Open Access

Clinicopathological characteristics, molecular features and novel diagnostic strategies for the detection of malignant transformation of endometriosis (Review)

  • Authors:
    • Hiroshi Kobayashi
  • View Affiliations

  • Published online on: April 24, 2023     https://doi.org/10.3892/etm.2023.11978
  • Article Number: 279
  • Copyright: © Kobayashi . This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Endometriosis is a benign gynecological disease that affects women of reproductive age. Although malignant transformation of endometriosis is rare, physicians must be aware of this due to the high incidence of clear cell carcinoma of the ovary (CCC) in Japan. The most prevalent histological subtype of ovarian cancer is CCC (~70%) followed by endometrioid carcinoma (30%). The present review discusses the clinicopathological and molecular features of endometriosis‑associated ovarian cancer (EAOC) as well as prospects for novel diagnostic strategies. Papers published between 2000 and 2022 in the PubMed and Google Scholar databases were included. Contents of the endometriotic cyst fluid may be involved in carcinogenesis, although the underlying mechanisms are largely unknown. Some studies have proposed a possible mechanism wherein excessive hemoglobin, heme and iron could cause an imbalance in intracellular redox homeostasis in endometriotic cells. Combined with DNA damage and mutations, the imbalances may induce the development of EAOC. Endometriotic cells evolve to adapt to the prolonged unfavorable oxidative microenvironmental stress. On the other hand, macrophages enhance the antioxidative defense mechanism and protect endometriotic cells against oxidative damage through intercellular crosstalk and signaling pathways. Therefore, changes in redox signaling, energy metabolism and the tumor immune microenvironment could be the key elements in the malignant transformation of certain endometriotic cell clones. Additionally, non‑invasive bioimaging (i.e., magnetic resonance relaxometry) and biomarkers (i.e., tissue factor pathway inhibitor 2) may be promising tools for early‑stage detection of the disease. In conclusion, the present review summarizes the latest advancements in research on the biological characteristics and early diagnosis of malignant transformation of endometriosis.

1. Introduction

Endometriosis is a common gynecological disease in women of reproductive age that is characterized by the development of functional endometrial tissue located outside the uterus (1). Endometriosis is a pathologically benign disease that is classified into ovarian endometriosis, superficial peritoneal disease, or deep infiltrating endometriosis. Endometriosis most commonly affects the ovaries and is associated with pelvic pain, infertility, or malignant transformation (1). Endometriosis is a possible precursor lesion of endometriosis-associated ovarian cancer (EAOC), including clear cell carcinoma of the ovary (CCC) and endometrioid ovarian carcinomas (2). CCC is the most common subtype of EAOC in Japan (70%) followed by endometrioid carcinoma (3).

The molecular mechanisms underlying the malignant transformation of endometriosis remain unclear; however, redox homeostasis imbalance might be involved. Endometriotic cysts contain high levels of iron, and endometriotic cells survive in reactive oxygen species (ROS)-rich environments (4). Hemoglobin, heme, and iron derivatives cause DNA damage and mutations, which promotes endometrial cell survival and proliferation and causes ectopic implantation (5-7). However, high levels of iron lead to cell death due to severe oxidative stress. It is necessary to elucidate the adaptive mechanisms that allow endometriotic cells to survive harsh environments and the key mediators that regulate redox homeostatic balance (7). Due to the greater number of patients with CCC in Japan compared with Western countries (25% vs. 8%), special attention has been paid to ensure the early diagnosis of malignant transformation of endometriosis (3).

The aim of this review is to summarize information on the clinicopathological characteristics, molecular mechanisms underlying redox homeostasis, and innovative diagnostic methods for malignant transformation of endometriosis. Finally, we discuss the current challenges and future directions.

2. Literature search

Search strategy and selection criteria

A computerized literature search was performed to identify relevant studies in English. The PubMed and Google Scholar electronic databases were searched for studies published between January 2000 and February 2022. The search terms included endometriosis, endometriosis-associated ovarian cancer, oxidative stress, antioxidant, energy metabolism, macrophages, imaging, and serodiagnosis. The references of each article were searched to identify potentially relevant studies. Publications of original studies and review papers were included. Given the heterogeneity in the research theme, data from studies were synthesized using a descriptive review design with narrative methods. Fig. 1 shows the first identification phase that includes records identified through a database search. Terms in the titles and abstracts were searched during the first screening. During the second screening, duplicates were removed, and titles, abstracts, and full-text articles were read to remove inappropriate papers. The final eligibility phase included full-text articles for analysis after excluding those wherein detailed data cannot be extracted.

3. Current understanding of malignant transformation of endometriosis

Current challenges in the malignant transformation of endometriosis in clinical practice

Most cases of endometriosis are benign and regress spontaneously after menopause. Sampson reported the first case of a malignant neoplasm arising from pre-existing endometriosis in 1925(8). Malignant transformation of endometriosis is rare, with an incidence of approximately <1.0% (2,9). Advances in genomics and molecular biology have made it possible to elucidate the potential mechanisms in the malignant transformation of endometriosis. Some researchers have proposed the clonal evolution model, wherein normal endometrial cells transform into neoplastic cells via endometriosis and atypical precursors based on oncogenic driver gene mutations (i.e., AT-rich interacting domain-containing protein 1A [ARID1A] gene) (10). Murakami et al (11) recently proposed a carcinogenic mechanism that ‘EAOC might not occur as a result of malignant transformation of endometrial cysts and might be caused by eutopic endometrial glandular epithelial cells that are refluxed to engraft in the ovary’. Genomic and epigenomic studies are required to better understand why and how endometriosis causes malignant transformation.

Epidemiological studies revealed that hysterectomy and unilateral oophorectomy reduces the risk of EAOC by 50-80% (12,13). Therefore, more than half of cases of EAOC originate from endometrial tissue fragments that have been regurgitated during menstruation, while other cases may have been caused by already existing endometriotic cysts (11). In fact, histopathological examination revealed that some cases of EAOC showed a continuous transition from benign endometriosis to atypical endometriosis and finally to invasive carcinoma (14-16). The prevalence of atypical endometriosis varies considerably, ranging from 1.7 to 40% (17-20). The difference in the results may be due to the lack of a unified international consensus regarding the definition of atypical endometriosis. In clinical practice, endometriosis is diagnosed based on clinical symptoms combined with diagnostic techniques, including transvaginal ultrasound (TVS) and blood biomarkers (e.g., carbohydrate antigen 125 [CA-125]) (21). TVS is the first-line diagnostic method for discriminating benign from malignant ovarian tumors. As shown in Fig. 2, EAOC is more common in women aged over 40 years, and the size of the cyst is larger than that of endometriosis (9). Benign ovarian endometriosis may pertain to any of the following: 1) pathologically benign endometriosis; 2) endometriosis with atypical lesions; and 3) endometriosis with clinically undetectable early-stage ovarian cancer. Although endometriosis with atypical lesions and with undetectable early-stage cancer may develop into ovarian cancer, patients with these conditions cannot be diagnosed as having cancer. Therefore, current screening options for early detection of EAOC are warranted.

Ovarian cancer initiation and progression via persistent oxidative stress mediated by iron overload

Oxidative stress is caused by an imbalance in the redox homeostasis between ROS overproduction and the antioxidant defense system (22). Iron-induced redox imbalance plays an important role in the pathophysiology of endometriosis, resulting in either cell death (anti-tumorigenic) or survival (pro-tumorigenic) (4,6,7). Repeated hemorrhage occurs in endometriotic cysts and in the peritoneal cavity during menstruation (2,7). Hemoglobin, heme, and iron are released following hemolysis from red blood cells (2,23). Cellular iron is imported through enhanced divalent metal transporter-1 (DMT1) and exported through ferroportin (FPN) (24). The differential expression of transporters (i.e., DMT1 upregulation and FPN downregulation) is associated with increased levels of intracellular iron (24). Iron content is reportedly elevated in the peritoneal cavity of women with endometriosis, in endometriotic cysts, in macrophages throughout the stroma of endometriotic cysts, and in the fimbriae of the fallopian tubes (24,25). Autoxidation (22) and the Fenton reaction (26) from the transformation of ferrous Fe2+ (oxyhemoglobin) to ferric Fe3+ (methemoglobin) produce large amounts of superoxide radicals (O2-) and hydroxyl radicals (OH). Accumulation of oxidative stress biomarkers (e.g., methemoglobin, 8-hydroxy-2'-deoxyguanosine, and lipid ROS) and inactivation of antioxidant enzymes (e.g., glutathione peroxidase 4) have been noted in endometriotic cysts (22,27). High levels of hemoglobin, heme, and iron derivatives in endometriotic cysts induce redox imbalance (22), leading to cell death through persistent DNA damage (7). Moreover, excessive accumulation of lipid ROS triggers ferroptotic cell death in an iron-dependent manner (28). In contrast, antioxidants protect cells from DNA damage by neutralizing excess ROS (29). A sublethal dose of iron overload participates in mutagenesis and aberrant pro-tumorigenic signaling to activate cell survival and promote carcinogenesis (29,30). Thus, low to moderate oxidative stress may contribute to the development of EAOC (26,27,29-31). This suggests that not only endometriosis but also conditions of iron overload, such as hemochromatosis, viral hepatitis B and C, and asbestos exposure, are risk factors for cancer development (32).

Metallobiology revealed diverse features involved in redox imbalance and the pathogenesis of malignant transformation of endometriosis (27). Compared to endometriosis, the contents of EAOC cysts have high antioxidant capacity due to lower levels of iron (14.2±36.6 mg/l vs. 244.4±204.9 mg/l) and higher levels of oxyhemoglobin (33) (Fig. 2). The contents of endometriotic and EAOC cysts are often dark and light brown, respectively, which may reflect iron concentrations and hemoglobin species. A study examining the expression of iron transport proteins showed that iron excretion is greater in CCC cells than in endometriotic cells (24), indicating that the intracellular levels of iron in CCC cells are maintained at low levels. Iwabuchi et al (27) provided a detailed characterization of hemoglobin species, such as methemoglobin and oxyhemoglobin, in endometriotic and EAOC cysts. The major components of hemoglobin species in endometriotic and EAOC cysts are methemoglobin and oxyhemoglobin, respectively (27) (Fig. 2). High levels of antioxidants allow EAOC cells to escape the lethal effects of excessive oxidative stress-induced cell death. Thus, iron levels should be maintained within a narrow range to prevent endometriotic cell death, which may in turn increase the risk of cancer progression when the cellular antioxidant capacity exceeds ROS production (2,6,27,29).

Dual role of antioxidants in the different stages of malignant transformation of endometriosis

Nagayasu et al (34) presented a comprehensive review on the endogenous antioxidant defense systems in endometriosis. The generated ROS can be scavenged by enzymatic (e.g., superoxide dismutase [SOD], catalase, reduced glutathione [GSH], glutathione peroxidase [GPX], and thioredoxin) and non-enzymatic (e.g., estradiol, melatonin, vitamin E, and vitamin C) antioxidants (35,36). Increased levels of oxidants (e.g., iron, ROS, nitric oxide [NO], lipid peroxidation, or advanced oxidation protein products) and decreased levels of antioxidants (e.g., SOD, catalase, GSH, GPX, glutathione reductase, total antioxidant capacity, vitamin C, or vitamin E) were observed in the serum, peritoneal fluid, follicular fluid, and tissue samples collected from patients with endometriosis compared to those in patients without endometriosis (37-39). Estradiol is a potent antioxidant that overcomes excessive oxidative stress by directly scavenging ROS or increasing the expression of SOD (34), and it is also involved in the development, maintenance, and progression of endometriosis. This may be the reason why endometriosis spontaneously regresses after menopause.

Overproduction of ROS and upregulation of endogenous antioxidant genes and signaling pathways are the main biological features of human cancers (29). Antioxidants have a dual role in tumor development and progression as both a tumor suppressor and promoter (29). Since nuclear factor erythroid 2-related factor 2 (NRF2) and CD44 variant isoform 9 (CD44v9) are well-studied regulators of antioxidants, we explain the role of these genes as an example. In genetically engineered mouse model, antioxidant (e.g., NRF2) depletion promotes carcinogenesis, demonstrating that antioxidants are involved in inhibiting cancer initiation and development (40). For example, vitamin supplementation may decrease the risk of developing human cancers (41). In contrast, treatment with antioxidant inhibitors reduced the growth of pre-existing cancer in xenograft animal models (42). Antioxidant inhibitors increase intracellular ROS and accelerate oxidative damage that induces cell death. For example, redox homeostasis in ovarian cancer is regulated by endogenous antioxidants, including NRF2 and CD44v9(29). Concurrent inhibition of CD44v9 and NRF2 triggers apoptotic cell death via the ROS-dependent p38 and p21 pathways (29). Animal experiments revealed that sublethal oxidative stress (low to moderate toxicity) is involved in the early phase of cancer initiation, while sufficient antioxidant capacity is necessary in the late phase of cancer progression. Similar results have been observed in changes in redox homeostasis in the process of malignant transformation of endometriosis (22,43). In the EAOC group, 8-OHdG levels in cystic fluids were significantly decreased, while levels of antioxidants (e.g., heme oxygenase-1 [HO-1], cytochrome P450 family, and glutathione transferase family) were increased compared with those in the endometriosis group (22,43). EAOC cells might be protected against excessive oxidative stress by an effective antioxidant defense (22). A fine-tuned pattern in redox homeostasis supports the idea that pro- and antioxidants may be involved in the initiation and progression of EAOC, respectively (7,23). EAOC is a disease that involves redox homeostasis and has predominant antioxidant capacity. Taken together, iron homeostatic pathways contribute to cancer initiation and progression via two major processes (Fig. 3). In the first step, hemoglobin, heme, and iron cause DNA damage and mutations by producing ROS, leading to cancer initiation. In the next step, antioxidants influence cancer progression.

Altered survival signals in endometriosis and EAOC

Energy production is required for cell adaptation and survival in stressful environmental conditions. Glucose is metabolized through glycolysis and oxidative phosphorylation (OXPHOS) to produce adenosine triphosphate (ATP), an important energy source. Endometriosis and EAOC, especially CCC, are characterized by common metabolic and molecular alterations (5,44,45). The key metabolic features include increased glucose uptake, anaerobic glycolysis, lactate production, and metabolic conversion from mitochondrial OXPHOS to aerobic glycolysis (44-47). Kirsten rat sarcoma (KRAS) and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) mutations found in endometriosis and EAOC are associated with glucose metabolism (10,48,49). However, much of our understanding about energy metabolism by mutated genes has come primarily from in vitro studies using other types of cancer cells rather than those in endometriosis or EAOC.

Transforming growth factor-beta (TGF-β) and hypoxia-inducible factor-1alpha (HIF-1α) have attracted attention as potential target genes related to energy metabolism in endometriosis (44,45) (Fig. 3). TGF-β is expressed in response to hypoxia, mitochondrial stress, oxidative stress, or tissue damage (44,45,50). TGF-β signaling enhances aerobic glycolysis by promoting glucose transporter 1 (GLUT1) expression, which converts glucose to lactate (44,45,50). HIF-1α induced by TGF-β1 modifies the expression of metabolism-related enzymes (44). Overexpression of HIF-1 inhibits pyruvate dehydrogenase (PDH) activation via upregulation of pyruvate dehydrogenase kinase (PDK) expression (44,45). PDH converts pyruvate to acetyl-CoA and stimulates the transfer of acetyl-CoA to the tricarboxylic acid cycle, enhancing the formation of ATP. Therefore, TGF-β and HIF-1α are major players in the metabolic switch from OXPHOS to glycolysis through the PDK-PDH pathway (44,45). Mitochondria not only produce ATP but also induce oxidative stress via the overproduction of ROS. Therefore, cells that survive in unfavorable environments (e.g., endometriotic and cancer cells) suppress ROS production in favor of glycolysis over mitochondrial OXPHOS even if they reduce energy production (44,45). Indeed, endometriotic lesions had higher expression of TGF-β and HIF-1α genes compared with eutopic endometrium (44). TGF-β plays multiple roles in tumorigenesis, possibly through the activation of the HIF-1α-PDK-PDH pathway (44,45). A recent study demonstrated that overexpression of PDK2 was linked to poor prognosis in patients with CCC due to decreased production of mitochondrial ROS (51). Emerging evidence indicates that TGF-β, HIF-1α, and glycolysis-related genes play critical roles in the pathogenesis and progression of endometriosis and CCC.

Hepatocyte nuclear factor-1 beta (HNF-1β) is a transcription factor that is overexpressed in almost all CCCs (52). HNF-1β is expressed only in CCC and not in other types of ovarian cancers (53). HNF-1β is also expressed in approximately 40% of endometriotic lesions (52). HNF-1β is a promising new marker for molecular diagnosis of both benign and malignant lesions (54). Single-nucleotide polymorphisms (rs11651755) in HNF-1β also correlated with the risk for ovarian cancer and endometriosis (55,56). Several papers suggested that HNF-1β affects a wide range of biological events, including tumorigenesis. For example, HNF-1β promotes glycogen accumulation in the cytoplasm of normal and gestational endometrial and CCC cells (28,57). HNF-1β is a key regulator of gene expression regulating metabolic remodeling, cell cycle arrest, and anti-apoptosis. First, HNF-1β induces glucose uptake into CCC cells via the overexpression of glucose transporter GLUT1 and facilitates glycolytic activity, lactate production, and glutathione synthesis, all of which are involved in the maintenance of redox homeostasis through enhanced anaerobic metabolism, decreased mitochondrial OXPHOS, and reduced ROS accumulation (46,53). Second, HNF-1β inhibits cell proliferation by blocking G1/S cell cycle progression through direct suppression of SMAD6-induced cyclin D1 expression (46,58). Furthermore, HNF-1β promotes G2/M cell cycle arrest in CCC cells through activation of the deubiquitinase ubiquitin-specific protease 28 (USP28)-Claspin-checkpoint kinase 1 (Chk1) signal transduction cascade (59). Finally, HNF-1β promotes cell proliferation and survival of endometriotic and CCC cells by upregulating the transcriptional regulator nuclear factor kappa B-induced antiapoptotic BCL2 gene expression (60,61). Thus, HNF-1β can facilitate DNA damage repair by inducing transient cell cycle arrest. HNF-1β protects endometriotic and CCC cells from oxidative damage, thereby contributing to the promotion of cell growth and survival.

Taken together, the potential target genes (i.e., KRAS, PIK3CA, TGF-β, HIF-1α and HNF-1β) confer a survival advantage against oxidative stress. Genome-scale reconstructions of metabolic, redox homeostasis, cell cycle, and survival signals are critical factors in disease progression (47,53). Accumulating evidence demonstrates that common metabolic reprogramming for cell survival, namely the Warburg effect, has been identified not only in cancer cells but also in endometriotic cells (20,44-46,53).

Functional roles of macrophages in the endometriotic/EAOC microenvironment

In response to inflammation, hypoxia, and oxidative stress, endometriosis creates a unique microenvironment composed of epithelium, stroma, mesenchyme, and infiltrative immune cells (62). Endometriotic cells adapt to and survive in hypoxic and oxidative stress conditions due to the high levels of hemoglobin, heme, iron, angiogenic factors (e.g., vascular endothelial growth factor), inflammatory cytokines and chemokines (e.g., TGF-β1, tumor necrosis factor alpha, interleukin-6 and -8, regulated on activation normal T cell expressed and secreted, and monocyte chemotactic protein-1 (62,63). These key mediators act as chemoattractants and enhance macrophage recruitment into endometriotic lesions. Iron-laden macrophages, especially M2 macrophages, are detected throughout the stroma of endometriotic cysts and CCC lesions (16,24,64). M2 macrophages scavenge iron and secrete antioxidants that relieve and eliminate excess oxidative stress in the surrounding environment (7). Heme oxygenase 1 (HO-1) is a key factor that contributes to antioxidant defenses (22). Co-culture experiments with macrophages and endometriotic cells have demonstrated that macrophage-derived HO-1 expression was induced by TGF-β1 produced by endometriotic cells (65). Addition of ROS to the co-culture system enhanced HO-1 production from macrophages (65). This crosstalk significantly enhances the antioxidant defense mechanism and protects endometriotic cells against oxidative injury, contributing to the progression of endometriosis (65). Moreover, a mouse model of endometriosis demonstrated that targeted depletion of macrophages blocks the growth of endometriotic lesions (66). Preclinical research suggests that endometriosis-macrophage crosstalk can provide a promising survival advantage to endometriotic cells (65,66).

Tumor-associated M2 macrophages contribute to tumor growth, invasion, metastasis, and treatment resistance in ovarian cancer (67); however, the effects of macrophages on the malignant transformation of endometriosis yielded inconsistent results. There were no statistically significant differences in the number of M2 macrophages in endometriotic cysts and CCC cells (24). In contrast, it was reported that the number of HO-1-positive M2 macrophages was significantly lower in the CCC group compared to the endometriotic cyst group (64). Additionally, CDC42-positive macrophages may prevent malignant transformation of endometriosis (68). CDC42, a small GTPase of the Rho-subfamily, regulates cell cycle progression. The number, polarization, and characteristics of infiltrating macrophages as well as their spatial distribution (e.g., closer proximity of M2 macrophages to target cells) play an important role in the carcinogenic process.

4. A promising tool for the early diagnosis of malignant transformation of endometriosis

Non-invasive diagnostic imaging

TVS is an effective imaging technique for the evaluation of ovarian masses, while magnetic resonance imaging may be used to identify TVS-indeterminate lesions. Increasing tumor size, rapidly growing mural nodules, papillary excrescences, or irregular and thick septations are features highly suspicious of malignancy (69). The conventional diagnostic modalities cannot distinguish benign from malignant lesions before the appearance of various morphological and anatomical changes in the cyst. Therefore, early-stage disease with little or no morphological changes may be missed during imaging surveillance. Histological examination is the gold standard for diagnosis, but it is an invasive procedure. Non-invasive diagnostic approaches to distinguish between endometriosis and EAOC are warranted. In 2008, Yamaguchi et al (4) showed for the first time that iron levels in endometriotic cysts are higher than in other benign ovarian cysts. Total iron, heme iron, and free iron levels in cystic fluids are useful markers in the differential diagnosis of endometriosis and EAOC (33). Total iron levels in patients with endometriosis were markedly higher than those with EAOC (median ± SD, 244.4±204.9 mg/l vs. 14.2±36.6 mg/l, P<0.001) (33). The total iron level that distinguishes endometriosis from EAOC provided 90.9% sensitivity and 100% specificity, with an optimal cut-off value of 64.8 mg/l (33). Recent advances in metallobiology have enabled the non-invasive quantification of iron levels. MR transverse relaxometry quantifies iron concentrations in endometriotic cysts using non-invasive techniques (70). MR relaxometry calculates the R2 value as a predicted value of iron concentration using a single-voxel multi-echo MR sequence (HISTO) by a 3T-MR system (70). R2 values highly correlated with iron concentrations, allowing for the rapid and non-invasive differentiation of endometriotic cysts from EAOC preoperatively, with a sensitivity of 86% and a specificity of 94% (70). MR relaxometry is a promising alternative for the early detection of malignant transformation of endometriosis (71-73). Additionally, real-time in vivo imaging methods used for the diagnosis of malignant transformation of endometriosis include electronic absorption spectroscopy and near infrared approach in addition to MR transverse relaxometry (74). However, diagnostic imaging techniques other than MR relaxometry are not available in clinical practice. Endometriosis causes changes in biochemical markers, including iron, during malignant transformation; hence, novel diagnostic modalities may allow physicians to detect biochemical changes early in the disease before anatomical changes occur. The non-invasive quantification of iron levels will aid in the early diagnosis and provide information that improves disease management strategies.

Potential serodiagnostic biomarkers

The final part highlights new serum biomarkers that may distinguish between benign and malignant ovarian tumors and improve the diagnostic accuracy for ovarian cancer. CA-125 is widely used in the diagnosis and monitoring of patients with ovarian cancer (75). However, the use of CA-125 is limited by its low sensitivity for CCC and a high false positivity rate in endometriosis (76). As CCC is the prevalent type of EAOC in Japan, novel non-invasive biomarkers that can accurately distinguish CCC from endometriosis are urgently needed. Human epididymis protein 4 (HE4) levels are not elevated in benign diseases, such as endometriosis, and are not affected by the menstrual cycle or hormone therapy. HE4 also has higher specificity than CA-125 (0.93 vs. 0.75, respectively) (77,78). Hence, HE4 could be useful in distinguishing suspected malignant ovarian tumors from endometriosis. However, HE4 is affected by several factors, including menopausal status, age, smoking, and renal dysfunction (79). Serum HE4 levels were elevated in 90% of patients with high-grade serous ovarian cancer and 69% of patients with CCC tumors, indicating that HE4 is reliable in diagnosing serous ovarian cancer, while it seems to be a less useful marker for CCC (80). Moreover, scoring systems and prediction algorithms may better assess the risk for ovarian cancer (81,82). The Risk of Ovarian Malignancy Algorithm and Copenhagen Index, including CA-125, HE4, and menopausal status or age, have high diagnostic performance for differentiating benign from malignant lesions (81,82). Additionally, recent advances in secretome analysis and bioinformatics revealed a serine protease inhibitor, namely tissue factor pathway inhibitor 2 (TFPI2), as a potential biomarker for the detection of ovarian cancer, especially CCC (76,83). TFPI2 is elevated in the serum and tumor tissues of patients with CCC, suggesting that serum TFPI2 may be a clinically useful biomarker for diagnosing CCC (76,83). A prospective validation study revealed that TFPI2 provided sufficient specificity for predicting CCC (79.5%) (84). Additionally, CA-125 had a high false-positive rate (71.4% [15/21]) in endometriosis, whereas none of the patients with elevated CA-125 levels had elevated TFPI2 levels (84). Miyagi et al (84) evaluated the diagnostic value of CA-125 and TFPI2 for distinguishing endometriosis and CCC, demonstrating that TFPI2 is superior to CA-125 as a marker of CCC (AUC 0.855 vs. 0.520). Based on these results, TFPI2 testing became available for ovarian cancers under the national health insurance in Japan since April 2021. We believe that TFPI2 can be used as a reliable diagnostic and treatment marker for malignant transformation of endometriosis into CCC.

5. Discussion and future challenges

We discuss the current challenges and future directions for malignant transformation of endometriosis. Fig. 3 shows the redox balance and metabolic switch regulating the endometriotic cell fate (death or survival) during malignant transformation. Redox homeostasis is regulated in response to environmental changes. Hemoglobin, heme, and iron are abundant in endometriotic cysts and cause oxidative stress (5-7,22). In the oxidative microenvironment, endometriotic cells stimulate the production of HO-1 from the surrounding macrophages via TGF-β production (64,65). HO-1 is a key regulator of redox homeostasis that eliminates excessive ROS in endometriotic cells and prevents cell death (64,65). This crosstalk can help macrophages promote endometriotic cell survival. On the other hand, the response of endometriotic cells to harsh environmental conditions requires energy generated through a metabolic shift from aerobic glycolysis to mitochondrial oxidation. In endometriotic cells under excessive oxidative stress (a redox meter showing the red area in Fig. 3), a metabolic shift to oxidative phosphorylation ultimately results in the death of endometriotic cells via extra ROS production from mitochondria. Proper energy homeostasis is regulated by specific control systems (43,44). The PDK-PDH axis plays a key role in mitochondrial dynamics during energy switching (44,45). When redox homeostasis is balanced in an oxidative microenvironment (a meter showing the yellow area in Fig. 3), HIF-1α stimulates PDK activation via TGF-β1 expression. As a result, ROS accumulation is reduced by fermentative glycolysis through a metabolic shift from mitochondrial OXPHOS to aerobic glycolysis. Accumulation of ROS at sublethal doses may increase tumorigenic potential via DNA mutagenesis (29,30).

Recent genomic studies revealed that somatic mutations accumulated in cancer-related genes (e.g., ARID1A, PIK3CA, and KRAS proto-oncogenes) not only in epithelial cells of endometriosis but also in the normal endometrium, and that mutagenesis in a certain gene (i.e., apolipoprotein B mRNA Editing Enzyme Catalytic Subunit) is involved in the genomic heterogeneity of endometriosis (85). Candidate gene mutations may affect cell survival via changes in inflammation, redox homeostasis, and metabolism. Genetic diversity occurs over time (85). Endometriosis clones that have adapted to harsh environments have also fueled adaptation to a suitable environment for carcinogenesis. Since the ARID1A gene is mutated in 46-70% of patients with CCC (10), epigenetic inactivation of this gene also contributes to the development of CCC. Methemoglobin and oxyhemoglobin represent the major hemoglobin species of endometriosis and EAOC cystic fluid, respectively (27). Endometriotic cells surviving in oxyhemoglobin-rich antioxidant environments (a meter showing the green area in Fig. 3) have a greater ability for carcinogenesis possibly through (epi)genetic modulations of CCC susceptibility genes, leading to the initiation and progression of malignant transformation. Furthermore, some antioxidants (e.g., HO-1) produced by macrophages may play a role in tumor promotion and progression. Therefore, the main hallmarks of malignant transformation are the changes in redox signaling, energy metabolism, and tumor immune microenvironment. Understanding the molecular mechanisms underlying these biological features will open new possibilities for the diagnosis, treatment, and management of the disease.

In clinical practice, conventional biomarkers and diagnostic imaging techniques are not sufficient for the early detection of malignant transformation of endometriosis. Novel imaging modalities (e.g., MR relaxometry) and biomarkers (e.g., TFPI2) are promising technologies. MR relaxometry offers the possibility of early diagnosis as changes in iron levels can be quantified quickly and non-invasively (70-73). Compared with conventional diagnostic imaging, MR relaxometry more accurately detects malignant transformation; however, there is limited data on its use. TFPI2 may become a useful biomarker for discriminating between endometriosis and CCC (76,83,84). In April 2021, TFPI2 testing was covered by the Japanese national health insurance system, enabling multicenter collaborative research to evaluate its diagnostic accuracy. Understanding the pathophysiology of malignant transformation may lead to the development of novel diagnostic imaging techniques and biomarkers. This review described the mechanisms of redox homeostasis, energy metabolism, and crosstalk with macrophages to better understand the malignant transformation of endometriosis and summarized the potential diagnostic and monitoring techniques for early diagnosis. Further studies are needed to determine how endometriotic cell clones that have acquired survival benefits undergo malignant transformation.

Acknowledgements

Figure 3 was created by Mrs. Toyomi Kobayashi (Ms.Clinic MayOne, Kashihara, Nara, Japan).

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

HK was in charge of conceptualization, data curation, formal analysis, funding acquisition, investigation, methodology, project administration, resources, supervision, validation, visualization, original draft writing, review and editing. Data authentication is not applicable. The author read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The author declares that they have no competing interests.

References

1 

Giudice LC: Clinical practice. Endometriosis. N Engl J Med. 362:2389–2398. 2010.PubMed/NCBI View Article : Google Scholar

2 

Wei JJ, William J and Bulun S: Endometriosis and ovarian cancer: A review of clinical, pathologic, and molecular aspects. Int J Gynecol Pathol. 30:553–568. 2011.PubMed/NCBI View Article : Google Scholar

3 

Machida H, Matsuo K, Yamagami W, Ebina Y, Kobayashi Y, Tabata T, Kanauchi M, Nagase S, Enomoto T and Mikami M: Trends and characteristics of epithelial ovarian cancer in Japan between 2002 and 2015: A JSGO-JSOG joint study. Gynecol Oncol. 153:589–596. 2019.PubMed/NCBI View Article : Google Scholar

4 

Yamaguchi K, Mandai M, Toyokuni S, Hamanishi J, Higuchi T, Takakura K and 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. 14:32–40. 2008.PubMed/NCBI View Article : Google Scholar

5 

Van Langendonckt A, Casanas-Roux F and Donnez J: Iron overload in the peritoneal cavity of women with pelvic endometriosis. Fertil Steril. 78:712–718. 2002.PubMed/NCBI View Article : Google Scholar

6 

Mandai M, Matsumura N, Baba T, Yamaguchi K, Hamanishi J and Konishi I: Ovarian clear cell carcinoma as a stress-responsive cancer: Influence of the microenvironment on the carcinogenesis and cancer phenotype. Cancer Lett. 310:129–133. 2011.PubMed/NCBI View Article : Google Scholar

7 

Kobayashi H: Potential scenarios leading to ovarian cancer arising from endometriosis. Redox Rep. 21:119–126. 2016.PubMed/NCBI View Article : Google Scholar

8 

Sampson JA: Endometrial carcinoma of the ovary, arising in endometrial tissue in that organ. Arch Surg. 10:1–72. 1925.

9 

Kobayashi H, Sumimoto K, Moniwa N, Imai M, Takakura K, Kuromaki T, Morioka E, Arisawa K and Terao T: Risk of developing ovarian cancer among women with ovarian endometrioma: A cohort study in Shizuoka, Japan. Int J Gynecol Cancer. 17:37–43. 2007.PubMed/NCBI View Article : Google Scholar

10 

Yachida N, Yoshihara K, Yamaguchi M, Suda K, Tamura R and Enomoto T: How does endometriosis lead to ovarian cancer? The molecular mechanism of endometriosis-associated ovarian cancer development. Cancers (Basel). 13(1439)2021.PubMed/NCBI View Article : Google Scholar

11 

Murakami K, Kotani Y, Nakai H and Matsumura N: Endometriosis-Associated ovarian cancer: The origin and targeted therapy. Cancers (Basel). 12(1676)2020.PubMed/NCBI View Article : Google Scholar

12 

Melin A, Sparén P, Persson I and Bergqvist A: Endometriosis and the risk of cancer with special emphasis on ovarian cancer. Hum Reprod. 21:1237–1242. 2006.PubMed/NCBI View Article : Google Scholar

13 

Dixon-Suen SC, Webb PM, Wilson LF, Tuesley K, Stewart LM and Jordan SJ: The association between hysterectomy and ovarian cancer risk: A population-based record-linkage study. J Natl Cancer Inst. 111:1097–1103. 2019.PubMed/NCBI View Article : Google Scholar

14 

LaGrenade A and Silverberg SG: Ovarian tumors associated with atypical endometriosis. Hum Pathol. 19:1080–1084. 1988.PubMed/NCBI View Article : Google Scholar

15 

Stern RC, Dash R, Bentley RC, Snyder MJ, Haney AF and Robboy SJ: Malignancy in endometriosis: Frequency and comparison of ovarian and extraovarian types. Int J Gynecol Pathol. 20:133–139. 2001.PubMed/NCBI View Article : Google Scholar

16 

Kondi-Pafiti A, Papakonstantinou E, Iavazzo C, Grigoriadis C, Salakos N and Gregoriou O: Clinicopathological characteristics of ovarian carcinomas associated with endometriosis. Arch Gynecol Obstet. 285:479–483. 2012.PubMed/NCBI View Article : Google Scholar

17 

Fukunaga M, Nomura K, Ishikawa E and Ushigome S: Ovarian atypical endometriosis: Its close association with malignant epithelial tumours. Histopathology. 30:249–255. 1997.PubMed/NCBI View Article : Google Scholar

18 

Van Gorp T, Amant F, Neven P, Vergote I and Moerman P: Endometriosis and the development of malignant tumours of the pelvis. A review of literature. Best Pract Res Clin Obstet Gynaecol. 18:349–371. 2004.PubMed/NCBI View Article : Google Scholar

19 

Vercellini P, Cribiù FM, Del Gobbo A, Carcangiu ML, Somigliana E and Bòsari S: The oncofetal protein IMP3: A novel biomarker and triage tool for premalignant atypical endometriotic lesions. Fertil Steril. 99:1974–1979. 2013.PubMed/NCBI View Article : Google Scholar

20 

Varga J, Reviczká A, Háková H, Švajdler P, Rabajdová M and Ostró A: Predictive factors of endometriosis progression into ovarian cancer. J Ovarian Res. 15(5)2022.PubMed/NCBI View Article : Google Scholar

21 

Dunselman GA, Vermeulen N, Becker C, Calhaz-Jorge C, D'Hooghe T, De Bie B, Heikinheimo O, Horne AW, Kiesel L, Nap A, et al: ESHRE guideline: Management of women with endometriosis. Hum Reprod. 29:400–412. 2014.PubMed/NCBI View Article : Google Scholar

22 

Iwabuchi T, Yoshimoto C, Shigetomi H and Kobayashi H: Oxidative stress and antioxidant defense in endometriosis and its malignant transformation. Oxid Med Cell Longev. 2015(848595)2015.PubMed/NCBI View Article : Google Scholar

23 

Kobayashi H, Yamada Y, Kawahara N, Ogawa K and Yoshimoto C: Integrating modern approaches to pathogenetic concepts of malignant transformation of endometriosis. Oncol Rep. 41:1729–1738. 2019.PubMed/NCBI View Article : Google Scholar

24 

Akashi K, Nagashima Y, Tabata T and Oda H: Immunochemical analysis of iron transporters and M2 macrophages in ovarian endometrioma and clear cell adenocarcinoma. Mol Clin Oncol. 15(159)2021.PubMed/NCBI View Article : Google Scholar

25 

Seidman JD: The presence of mucosal iron in the fallopian tube supports the ‘incessant menstruation hypothesis’ for ovarian carcinoma. Int J Gynecol Pathol. 32:454–458. 2013.PubMed/NCBI View Article : Google Scholar

26 

Yamada Y, Shigetomi H, Onogi A, Haruta S, Kawaguchi R, Yoshida S, Furukawa N, Nagai A, Tanase Y, Tsunemi T, et al: Redox-active iron-induced oxidative stress in the pathogenesis of clear cell carcinoma of the ovary. Int J Gynecol Cancer. 21:1200–1207. 2011.PubMed/NCBI View Article : Google Scholar

27 

Iwabuchi T, Yoshimoto C, Shigetomi H and Kobayashi H: Cyst fluid hemoglobin species in endometriosis and its malignant transformation: The role of metallobiology. Oncol Lett. 11:3384–3388. 2016.PubMed/NCBI View Article : Google Scholar

28 

Yamaguchi K, Kitamura S, Furutake Y, Murakami R, Yamanoi K, Taki M, Ukita M, Hamanishi J and Mandai M: Acquired evolution of mitochondrial metabolism regulated by HNF1B in ovarian clear cell carcinoma. Cancers (Basel). 13(2413)2021.PubMed/NCBI View Article : Google Scholar

29 

Kobayashi H, Imanaka S and Shigetomi H: Revisiting therapeutic strategies for ovarian cancer by focusing on redox homeostasis. Oncol Lett. 23(80)2022.PubMed/NCBI View Article : Google Scholar

30 

Vercellini P, Crosignani P, Somigliana E, Viganò P, Buggio L, Bolis G and Fedele L: The ‘incessant menstruation’ hypothesis: A mechanistic ovarian cancer model with implications for prevention. Hum Reprod. 26:2262–2273. 2011.PubMed/NCBI View Article : Google Scholar

31 

Defrère S, Van Langendonckt A, Vaesen S, Jouret M, González Ramos R, Gonzalez D and Donnez J: Iron overload enhances epithelial cell proliferation in endometriotic lesions induced in a murine model. Hum Reprod. 21:2810–2816. 2006.PubMed/NCBI View Article : Google Scholar

32 

Toyokuni S: Iron as a target of chemoprevention for longevity in humans. Free Radic Res. 45:906–917. 2011.PubMed/NCBI View Article : Google Scholar

33 

Yoshimoto C, Iwabuchi T, Shigetomi H and Kobayashi H: Cyst fluid iron-related compounds as useful markers to distinguish malignant transformation from benign endometriotic cysts. Cancer Biomark. 15:493–499. 2015.PubMed/NCBI View Article : Google Scholar

34 

Nagayasu M, Imanaka S, Kimura M, Maruyama S and Kobayashi H: Nonhormonal treatment for endometriosis focusing on redox imbalance. Gynecol Obstet Invest. 86:1–12. 2021.PubMed/NCBI View Article : Google Scholar

35 

Turrens JF: Mitochondrial formation of reactive oxygen species. J Physiol. 552(Pt 2):335–344. 2003.PubMed/NCBI View Article : Google Scholar

36 

He L, He T, Farrar S, Ji L, Liu T and Ma X: Antioxidants maintain cellular redox homeostasis by elimination of reactive oxygen species. Cell Physiol Biochem. 44:532–553. 2017.PubMed/NCBI View Article : Google Scholar

37 

Ota H, Igarashi S, Hatazawa J and Tanaka T: Endometriosis and free radicals. Gynecol Obstet Invest. 48 (Suppl 1):S29–S35. 1999.PubMed/NCBI View Article : Google Scholar

38 

Jana SK, Dutta M, Joshi M, Srivastava S, Chakravarty B and Chaudhury K: 1H NMR based targeted metabolite profiling for understanding the complex relationship connecting oxidative stress with endometriosis. Biomed Res Int. 2013(329058)2013.PubMed/NCBI View Article : Google Scholar

39 

Singh AK, Chattopadhyay R, Chakravarty B and Chaudhury K: Markers of oxidative stress in follicular fluid of women with endometriosis and tubal infertility undergoing IVF. Reprod Toxicol. 42:116–124. 2013.PubMed/NCBI View Article : Google Scholar

40 

Moon EJ and Giaccia A: Dual roles of NRF2 in tumor prevention and progression: Possible implications in cancer treatment. Free Radic Biol Med. 79:292–299. 2015.PubMed/NCBI View Article : Google Scholar

41 

Vahid F and Davoodi SH: Nutritional factors involved in the etiology of gastric cancer: A systematic review. Nutr Cancer. 73:376–390. 2021.PubMed/NCBI View Article : Google Scholar

42 

Müller MF, Florian S, Pommer S, Osterhoff M, Esworthy RS, Chu FF, Brigelius-Flohé R and Kipp AP: Deletion of glutathione peroxidase-2 inhibits azoxymethane-induced colon cancer development. PLoS One. 8(e72055)2013.PubMed/NCBI View Article : Google Scholar

43 

Fujimoto Y, Imanaka S, Yamada Y, Ogawa K, Ito F, Kawahara N, Yoshimoto C and Kobayashi H: Comparison of redox parameters in ovarian endometrioma and its malignant transformation. Oncol Lett. 16:5257–5264. 2018.PubMed/NCBI View Article : Google Scholar

44 

Young VJ, Brown JK, Maybin J, Saunders PT, Duncan WC and Horne AW: Transforming growth factor-β induced Warburg-like metabolic reprogramming may underpin the development of peritoneal endometriosis. J Clin Endocrinol Metab. 99:3450–3459. 2014.PubMed/NCBI View Article : Google Scholar

45 

Young VJ, Ahmad SF, Brown JK, Duncan WC and Horne AW: ID2 mediates the transforming growth factor-β1-induced Warburg-like effect seen in the peritoneum of women with endometriosis. Mol Hum Reprod. 22:648–654. 2016.PubMed/NCBI View Article : Google Scholar

46 

Okamoto T, Mandai M, Matsumura N, Yamaguchi K, Kondoh H, Amano Y, Baba T, Hamanishi J, Abiko K, Kosaka K, et al: Hepatocyte nuclear factor-1β (HNF-1β) promotes glucose uptake and glycolytic activity in ovarian clear cell carcinoma. Mol Carcinog. 54:35–49. 2015.PubMed/NCBI View Article : Google Scholar

47 

Kobayashi H, Shigetomi H and Imanaka S: Nonhormonal therapy for endometriosis based on energy metabolism regulation. Reprod Fertil. 2:C42–C57. 2021.PubMed/NCBI View Article : Google Scholar

48 

Ying H, Kimmelman AC, Lyssiotis CA, Hua S, Chu GC, Fletcher-Sananikone E, Locasale JW, Son J, Zhang H, Coloff JL, et al: Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism. Cell. 149:656–670. 2012.PubMed/NCBI View Article : Google Scholar

49 

Bhattacharya B, Low SH, Soh C, Kamal Mustapa N, Beloueche-Babari M, Koh KX, Loh J and Soong R: Increased drug resistance is associated with reduced glucose levels and an enhanced glycolysis phenotype. Br J Pharmacol. 171:3255–3267. 2014.PubMed/NCBI View Article : Google Scholar

50 

Zhou MY, Cheng ML, Huang T, Hu RH, Zou GL, Li H, Zhang BF, Zhu JJ, Liu YM, Liu Y and Zhao XK: Transforming growth factor beta-1 upregulates glucose transporter 1 and glycolysis through canonical and noncanonical pathways in hepatic stellate cells. World J Gastroenterol. 27:6908–6926. 2021.PubMed/NCBI View Article : Google Scholar

51 

Kitamura S, Yamaguchi K, Murakami R, Furutake Y, Higasa K, Abiko K, Hamanishi J, Baba T, Matsumura N and Mandai M: PDK2 leads to cisplatin resistance through suppression of mitochondrial function in ovarian clear cell carcinoma. Cancer Sci. 112:4627–4640. 2021.PubMed/NCBI View Article : Google Scholar

52 

Kato N, Sasou S and Motoyama T: Expression of hepatocyte nuclear factor-1beta (HNF-1beta) in clear cell tumors and endometriosis of the ovary. Mod Pathol. 19:83–89. 2006.PubMed/NCBI View Article : Google Scholar

53 

Amano Y, Mandai M, Yamaguchi K, Matsumura N, Kharma B, Baba T, Abiko K, Hamanishi J, Yoshioka Y and Konishi I: Metabolic alterations caused by HNF1β expression in ovarian clear cell carcinoma contribute to cell survival. Oncotarget. 6:26002–26017. 2015.PubMed/NCBI View Article : Google Scholar

54 

Chandra S, Srinivasan S and Batra J: Hepatocyte nuclear factor 1 beta: A perspective in cancer. Cancer Med. 10:1791–1804. 2021.PubMed/NCBI View Article : Google Scholar

55 

Shen H, Fridley BL, Song H, Lawrenson K, Cunningham JM, Ramus SJ, Cicek MS, Tyrer J, Stram D, Larson MC, et al: Epigenetic analysis leads to identification of HNF1B as a subtype-specific susceptibility gene for ovarian cancer. Nat Commun. 4(1628)2013.PubMed/NCBI View Article : Google Scholar

56 

Burghaus S, Fasching PA, Häberle L, Rübner M, Büchner K, Blum S, Engel A, Ekici AB, Hartmann A, Hein A, et al: Genetic risk factors for ovarian cancer and their role for endometriosis risk. Gynecol Oncol. 145:142–147. 2017.PubMed/NCBI View Article : Google Scholar

57 

Yamamoto S, Tsuda H, Aida S, Shimazaki H, Tamai S and Matsubara O: Immunohistochemical detection of hepatocyte nuclear factor 1beta in ovarian and endometrial clear-cell adenocarcinomas and nonneoplastic endometrium. Hum Pathol. 38:1074–1080. 2007.PubMed/NCBI View Article : Google Scholar

58 

Lu W, Sun J, Zhou H, Wang F, Zhao C, Li K, Fan C, Ding G and Wang J: HNF1B inhibits cell proliferation via repression of SMAD6 expression in prostate cancer. J Cell Mol Med. 24:14539–14548. 2020.PubMed/NCBI View Article : Google Scholar

59 

Ito F, Yoshimoto C, Yamada Y, Sudo T and Kobayashi H: The HNF-1β-USP28-Claspin pathway upregulates DNA damage-induced Chk1 activation in ovarian clear cell carcinoma. Oncotarget. 9:17512–17522. 2018.PubMed/NCBI View Article : Google Scholar

60 

Suzuki E, Kajita S, Takahashi H, Matsumoto T, Tsuruta T and Saegusa M: Transcriptional upregulation of HNF-1β by NF-κB in ovarian clear cell carcinoma modulates susceptibility to apoptosis through alteration in bcl-2 expression. Lab Invest. 95:962–972. 2015.PubMed/NCBI View Article : Google Scholar

61 

Preya UH, Woo JH, Choi YS and Choi JH: Hepatocyte nuclear factor-1 beta protects endometriotic cells against apoptotic cell death by up-regulating the expression of antiapoptotic genes. Biol Reprod. 101:686–694. 2019.PubMed/NCBI View Article : Google Scholar

62 

Wendel JRH, Wang X and Hawkins SM: The endometriotic tumor microenvironment in ovarian cancer. Cancers (Basel). 10(261)2018.PubMed/NCBI View Article : Google Scholar

63 

Capobianco A and Rovere-Querini P: Endometriosis, a disease of the macrophage. Front Immunol. 4(9)2013.PubMed/NCBI View Article : Google Scholar

64 

Yamada Y, Uchiyama T, Ito F, Kawahara N, Ogawa K, Obayashi C and Kobayashi H: Clinical significance of M2 macrophages expressing heme oxygenase-1 in malignant transformation of ovarian endometrioma. Pathol Res Pract. 215:639–643. 2019.PubMed/NCBI View Article : Google Scholar

65 

Ogawa K, Liu T, Kawahara N and Kobayashi H: Macrophages protect endometriotic cells against oxidative damage through a cross-talk mechanism. Reprod Sci. 29:2165–2178. 2022.PubMed/NCBI View Article : Google Scholar

66 

Bacci M, Capobianco A, Monno A, Cottone L, Di Puppo F, Camisa B, Mariani M, Brignole C, Ponzoni M, Ferrari S, et al: Macrophages are alternatively activated in patients with endometriosis and required for growth and vascularization of lesions in a mouse model of disease. Am J Pathol. 175:547–556. 2009.PubMed/NCBI View Article : Google Scholar

67 

Colvin EK: Tumor-associated macrophages contribute to tumor progression in ovarian cancer. Front Oncol. 4(137)2014.PubMed/NCBI View Article : Google Scholar

68 

Canet B, Pons C, Espinosa I and Prat J: CDC42-positive macrophages may prevent malignant transformation of ovarian endometriosis. Hum Pathol. 43:720–725. 2012.PubMed/NCBI View Article : Google Scholar

69 

Guerriero S, Alcazar JL, Coccia ME, Ajossa S, Scarselli G, Boi M, Gerada M and Melis GB: Complex pelvic mass as a target of evaluation of vessel distribution by color Doppler sonography for the diagnosis of adnexal malignancies: Results of a multicenter European study. J Ultrasound Med. 21:1105–1111. 2002.PubMed/NCBI View Article : Google Scholar

70 

Yoshimoto C, Takahama J, Iwabuchi T, Uchikoshi M, Shigetomi H and Kobayashi H: Transverse relaxation rate of cyst fluid can predict malignant transformation of ovarian endometriosis. Magn Reson Med Sci. 16:137–145. 2017.PubMed/NCBI View Article : Google Scholar

71 

Matsubara S, Kawahara N, Horie A, Murakami R, Horikawa N, Sumida D, Wada T, Maehana T, Yamawaki A, Ichikawa M, et al: Magnetic resonance relaxometry improves the accuracy of conventional MRI in the diagnosis of endometriosis-associated ovarian cancer: A case report. Mol Clin Oncol. 11:296–300. 2019.PubMed/NCBI View Article : Google Scholar

72 

Liu T, Sumida D, Wada T, Maehana T, Yamawaki A, Sugimoto S, Kawahara N, Yoshimoto C and Kobayashi H: A diagnostic challenge of seromucinous borderline tumor: A case report. Medicine (Baltimore). 98(e15707)2019.PubMed/NCBI View Article : Google Scholar

73 

Kawahara N, Miyake R, Yamanaka S and Kobayashi H: A novel predictive tool for discriminating endometriosis associated ovarian cancer from ovarian endometrioma: The R2 predictive index. Cancers (Basel). 13(3829)2021.PubMed/NCBI View Article : Google Scholar

74 

Kobayashi H, Yamada Y, Kawahara N, Ogawa K and Yoshimoto C: Modern approaches to noninvasive diagnosis of malignant transformation of endometriosis. Oncol Lett. 17:1196–1202. 2019.PubMed/NCBI View Article : Google Scholar

75 

Molina R, Ojeda B, Filella X, Borras G, Jo J, Mas E, Lopez JJ and Ballesta A: A prospective study of tumor markers CA 125 and CA 19.9 in patients with epithelial ovarian carcinomas. Tumour Biol. 13:278–286. 1992.PubMed/NCBI View Article : Google Scholar

76 

Arakawa N, Miyagi E, Nomura A, Morita E, Ino Y, Ohtake N, Miyagi Y, Hirahara F and Hirano H: Secretome-based identification of TFPI2, a novel serum biomarker for detection of ovarian clear cell adenocarcinoma. J Proteome Res. 12:4340–4350. 2013.PubMed/NCBI View Article : Google Scholar

77 

Urban N, Thorpe J, Karlan BY, McIntosh MW, Palomares MR, Daly MB, Paley P and Drescher CW: Interpretation of single and serial measures of HE4 and CA125 in asymptomatic women at high risk for ovarian cancer. Cancer Epidemiol Biomarkers Prev. 21:2087–2094. 2012.PubMed/NCBI View Article : Google Scholar

78 

Zapardiel I, Gorostidi M, Ravaggi A, Allende MT, Silveira M and Macuks R: Utility of human epididymis protein 4 serum marker for the detection of adnexal malignancy: A multicentric prospective study. Eur J Cancer Prev. 26:346–350. 2017.PubMed/NCBI View Article : Google Scholar

79 

Lycke M, Ulfenborg B, Malchau Lauesgaard J, Kristjansdottir B and Sundfeldt K: Consideration should be given to smoking, endometriosis, renal function (eGFR) and age when interpreting CA125 and HE4 in ovarian tumor diagnostics. Clin Chem Lab Med. 59:1954–1962. 2021.PubMed/NCBI View Article : Google Scholar

80 

Blackman A, Mitchell J, Rowswell-Turner R, Singh R, Kim KK, Eklund E, Skates S, Bast RC, Messerlian G, Miller MC and Moore RG: Analysis of serum HE4 levels in various histologic subtypes of epithelial ovarian cancer and other malignant tumors. Tumour Biol. 43:355–365. 2021.PubMed/NCBI View Article : Google Scholar

81 

Dikmen ZG, Colak A, Dogan P, Tuncer S and Akbiyik F: Diagnostic performances of CA125, HE4, and ROMA index in ovarian cancer. Eur J Gynaecol Oncol. 36:457–462. 2015.PubMed/NCBI

82 

Wang Z, Tao X and Ying C: CPH-I and HE4 Are more favorable than CA125 in differentiating borderline ovarian tumors from epithelial ovarian cancer at early stages. Dis Markers. 2019(6241743)2019.PubMed/NCBI View Article : Google Scholar

83 

Arakawa N, Kobayashi H, Yonemoto N, Masuishi Y, Ino Y, Shigetomi H, Furukawa N, Ohtake N, Miyagi Y, Hirahara F, et al: Clinical significance of tissue factor pathway inhibitor 2, a serum biomarker candidate for ovarian clear cell carcinoma. PLoS One. 11(e0165609)2016.PubMed/NCBI View Article : Google Scholar

84 

Miyagi E, Arakawa N, Sakamaki K, Yokota NR, Yamanaka T, Yamada Y, Yamaguchi S, Nagao S, Hirashima Y, Kasamatsu Y, et al: Validation of tissue factor pathway inhibitor 2 as a specific biomarker for preoperative prediction of clear cell carcinoma of the ovary. Int J Clin Oncol. 26:1336–1344. 2021.PubMed/NCBI View Article : Google Scholar

85 

Revathidevi S, Nakaoka H, Suda K, Fujito N, Munirajan AK, Yoshihara K, Enomoto T and Inoue I: APOBEC mediated mutagenesis drives genomic heterogeneity in endometriosis. J Hum Genet. 67:323–329. 2022.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

June-2023
Volume 25 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kobayashi H: Clinicopathological characteristics, molecular features and novel diagnostic strategies for the detection of malignant transformation of endometriosis (Review). Exp Ther Med 25: 279, 2023
APA
Kobayashi, H. (2023). Clinicopathological characteristics, molecular features and novel diagnostic strategies for the detection of malignant transformation of endometriosis (Review). Experimental and Therapeutic Medicine, 25, 279. https://doi.org/10.3892/etm.2023.11978
MLA
Kobayashi, H."Clinicopathological characteristics, molecular features and novel diagnostic strategies for the detection of malignant transformation of endometriosis (Review)". Experimental and Therapeutic Medicine 25.6 (2023): 279.
Chicago
Kobayashi, H."Clinicopathological characteristics, molecular features and novel diagnostic strategies for the detection of malignant transformation of endometriosis (Review)". Experimental and Therapeutic Medicine 25, no. 6 (2023): 279. https://doi.org/10.3892/etm.2023.11978