Next Article in Journal
Brianolide from Briareum stechei Attenuates Atopic Dermatitis-like Skin Lesions by Regulating the NFκB and MAPK Pathways
Previous Article in Journal
Positive Effects of (+)-Epicatechin on Traumatic Spinal Cord Injury Recovery
Previous Article in Special Issue
RIPK4 Downregulation Reduces ABCG2 Expression, Increasing BRAF-Mutated Melanoma Cell Susceptibility to Cisplatin- and Doxorubicin-Induced Apoptosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Relationship Between Thyroid Dysfunction and Ovarian Cancer

Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, PL 30387 Cracow, Poland
*
Authors to whom correspondence should be addressed.
Biomolecules 2025, 15(6), 870; https://doi.org/10.3390/biom15060870 (registering DOI)
Submission received: 25 April 2025 / Revised: 6 June 2025 / Accepted: 13 June 2025 / Published: 14 June 2025
(This article belongs to the Special Issue Molecular Advances in Drug Resistance and Novel Therapies for Cancer)

Abstract

:
This review looks at the causes of the association between thyroid dysfunction (hyperthyroidism and hypothyroidism) and ovarian cancer (OC) risk. Epidemiological data have revealed that thyroid dysfunction, particularly hyperthyroidism, is associated with increased risk, progression, and mortality in patients with OC. In addition, research studies and databases have demonstrated that both the expression and localization of thyroid hormone receptors alpha (TRα) and beta (TRβ) and membrane thyroid hormone receptor integrin alpha V beta 3 (αvβ3) affect OC progression and survival in OC patients. Furthermore, this review described the levels of the thyroid hormones (THs) thyroxine (T4) and 3,5,3′-triiodo-L-thyronine (T3) in the blood of OC patients and their role in OC progression. Moreover, we present studies that reported the relationship between hyperthyroidism and hypothyroidism and the levels of metabolic hormones in the blood and the possible effects on metabolic reprogramming in OC cells. We also report data indicating the relationship between the treatment of thyroid dysfunction and OC progression. Finally, the cited case studies described the essential case of struma ovarii, which is OC, including thyroid tissue. This review describes the link between thyroid dysfunction and OC risk and progression, which may be important in treating OC patients with thyroid dysfunction.

1. Introduction

The associations between thyroid disorders (hyperthyroidism and hypothyroidism) and hormone-dependent cancers have been highlighted in several epidemiological studies [1,2]. In particular, epidemiological data suggest a relationship between thyroid dysfunction and ovarian cancer (OC) [3,4,5]. In addition, abnormal levels of the thyroid hormones (THs) L-thyroxine (T4) and 3,5,3′-triiodo-L-thyronine (T3) may be responsible for the formation and progression of OC. Studies suggest that T3 and T4 promote OC development [6,7]. Furthermore, a significant positive or negative correlation has been identified between the survival of OC patients and the expression of nuclear thyroid hormone receptor alpha (TRα) and beta (TRβ) and the expression of the membrane thyroid hormone receptor integrin alpha V beta 3 (αvβ3). These studies suggest the potential role of thyroid hormone receptors in the adjuvant therapy of various cancers, including OC [8,9].
This paper reviews the epidemiological, clinical, and experimental evidence for the link between thyroid dysfunction (hyperthyroidism and hypothyroidism) and OC, particularly concerning the possible molecular mechanisms underlying these associations. The potential role of hypothyroidism and hyperthyroidism in promoting OC is of particular interest (Figure 1). This review will also consider the impact of THs and their receptors on the progression of OC, including the effects on the genomic and nongenomic TH pathways. In addition, we present information on the indirect associations of thyroid dysfunction with OC progression and metabolism through the regulation of metabolic hormones. These publications indicate the dependence between the treatment of thyroid dysfunction and OC development. Finally, we describe a specific type of OC, namely, struma ovarii (SO).

2. Materials and Methods

2.1. Search Strategy

The main focus of this review was the associations between thyroid dysfunction (hyperthyroidism and hypothyroidism) and OC risk, mortality, and progression. SCOPUS (https://www.scopus.com), PubMed (https://www.ncbi.nlm.nih.gov/pubmed), Google Scholar (https://scholar.google.com/), and Web of Science (https://www.webofscience.com) databases were searched on the basis of individual keywords and MeSH terms. Keywords were combined in various ways using Boolean operators ‘and’ or ‘or’ where appropriate and database-specific filters to maximize the identification of articles describing the association between thyroid dysfunction and OC. The search was performed, and all articles published up to March 2025 were retrieved. The articles included were clinical and epidemiological studies that reported the associations of the incidence of thyroid dysfunction with OC risk, mortality, and prognosis; studies that investigated the expression of nuclear and membrane TH receptors in OC; and studies that reported the level of TH in the blood of OC patients. There have also been studies that analyzed the effects of TH on OC progression and metabolism, studies that investigated the levels of metabolic hormones in OC, and analyses that explored the associations between thyroid dysfunction treatments and OC treatments. The search was limited to English literature, including meta-analyses, case reports, original studies, and reviews (Figure 2).

2.2. GEPIA Dataset Analysis

The GEPIA database [10] was used in this study to analyze the transcription levels and correlations of the THRA, THRB, ITGAV, and ITGB3 genes in OC (n = 426) and normal (n = 88) tissues on 06.03.2025. Student’s t test was used to determine the p value. The p value cut-off was 0.05.

2.3. Kaplan–Meier Plot Analysis

The Kaplan–Meier plotter online global survival database (http://kmplot.com/analysis/), accessed on 1 March 2025 was used to evaluate the correlation between target gene mRNA expression and progression-free survival (PFS) and overall survival (OS) in the serous OC groups. The results are presented as Kaplan–Meier survival curves. The hazard ratio (HR) and 95% confidence interval (CI) were calculated automatically via the website tool. The values for each group are expressed as the means ± SDs. A log-rank p value < 0.05 was considered statistically significant [11].

3. Ovarian Cancer

3.1. Ovarian Cancer Epidemiology and Stages

According to the findings of the Centers for Disease Control and Prevention, ovarian cancer (OC) has been identified as the leading cause of death from gynecological malignancies on a global scale [12]. The Global Cancer Observatory, which covers 185 countries, reported that in 2022, the global incidence of OC was 324,603 cases, with a mortality rate of 206,956 [13,14,15]. Projections indicate that the estimated incidence of OC will reach approximately 477 thousand cases by the year 2045, with a corresponding mortality rate of approximately 328 thousand [15].
OC staging guidelines are determined by the International Federation of Gynecology and Obstetrics (FIGO) [16]. The survival probability is contingent upon the stage of the disease. For patients with early-stage disease (FIGO stage I or II), the five-year survival probabilities are 87% and 62%, respectively, whereas for patients with advanced-stage disease (FIGO stage III or IV), the probabilities are reduced to 26% and 14%, respectively [17]. The stage of OC is a critical factor in determining the most appropriate treatment and is also the most significant element in establishing prognosis. The high OC mortality rate is associated with nonspecific symptoms in the early stage, which means that 75% of women are diagnosed at an advanced stage [18].

3.2. Ovarian Cancer Classification

OC is a heterogeneous tumor. Each histological subtype of OC has a different morphology, biological behavior, and prognosis. Ninety percent of OCs are epithelial OCs (EOCs), which originate from the epithelium [19]. The World Health Organization (WHO) divides EOCs into different groups on the basis of the type of cell: serous carcinoma (SC), mucinous carcinoma (MC), endometrioid carcinoma (EC), clear cell carcinoma (CCC), transitional cell, Brenner tumors, and mixed and undifferentiated. High-grade SC (HGSC) is the most common type of EOC [20]. Furthermore, recent molecular characterization of EOC correlated with clinicopathologic findings revealed that EOCs can be classified into two distinct groups. Type I (low-grade SC (LGSC), low-grade EC (LGEC), MC, and CCC) is relatively indolent and is diagnosed at an early stage, whereas type II (HGSC, undifferentiated carcinoma, and carcinosarcoma) is diagnosed at an advanced stage and has aggressive behavior, with a high rate of rapid dissemination [21]. Non-EOC comprises germ cell tumors (GeCTs), sex cord-stromal tumors (SCSTs), and rare tumors [22]. The most prevalent type of SCST is ovarian granulosa cell tumor (GCT), which accounts for approximately 5% of malignant OCs. GCTs can be categorized into two distinct subtypes, namely, adult-type GCTs (AGCTs) and juvenile GCTs (JGCTs), on the basis of their histological characteristics and clinical symptoms [23] (Figure 3).

4. The Associations of Hyperthyroidism and Hypothyroidism with Ovarian Cancer: Epidemiological, Clinical and Experimental Evidence

4.1. Thyroid Gland Dysfunction: Hyperthyroidism and Hypothyroidism

The thyroid is an endocrine organ responsible for the production and secretion of thyroid hormones (THs), including L-thyroxine (T4) and 3,5,3′-triiodo-L-thyronine (T3), which regulate metabolism and growth in organisms [24,25]. The synthesis and secretion of TH are regulated by the hypothalamus–pituitary–thyroid (HPT) axis [26,27] (Figure 4). The dysregulated production and secretion of TH by the thyroid gland are associated with the incidence of thyroid dysfunctions (hyperthyroidism or hypothyroidism). The prevalence is influenced by the iodine content of the diet [28,29].
Hyperthyroidism is characterized by increased TH synthesis and secretion from the thyroid gland, and the most common cause of hyperthyroidism is autoimmune inflammation of the thyroid gland, including Graves’ disease [30]. The prevalence of hyperthyroidism is approximately 0.8% in Europe and 1.3% in the USA [31,32]. The most prevalent etiological agent of hyperthyroidism is considered to be autoimmune inflammation of the thyroid gland [3]. Over the past decade, studies have indicated a correlation between elevated levels of inflammation and an increased risk of OC, decreased survival of patients with OC, and increased development of OC [33].
Hypothyroidism is characterized by a deficiency in TH. The condition is predominantly attributable to autoimmune thyroid disease, a subset of which is represented by Hashimoto thyroiditis [29]. The reported prevalence rates of hypothyroidism in Europe and the USA are 3% and 4.6%, respectively [30,31].
Thyroid dysfunctions are observed more frequently in women than in men [28]. There is a strong interaction between the HPT axis and the regulation of estradiol and progesterone secretion by the ovary [34]. Furthermore, hypothyroidism has been described as a potential causative agent in the formation of ovarian cysts [35]. In addition, Perveen et al. [36] described the association between hypothyroidism and multicystic ovarian enlargement in a 23-year-old woman in a case report.

4.2. The Prevalence of Thyroid Dysfunction and the Risk or Mortality of Ovarian Cancer

4.2.1. Hyperthyroidism

In 2000, a population-based case–control study of OC (767 OC patients and 1367 controls) revealed that the risk of OC almost doubled in women aged 20–69 years with hyperthyroidism compared with women without a history of thyroid disease. These studies also suggest a potential link between hyperthyroidism and inflammatory responses within the ovarian epithelium [3]. A few years later, a potential association between hyperthyroidism and OC risk was reported in experimental studies [37]. A large study of a large group of people revealed that women with Graves’ disease have an increased risk of OC compared with women in the control group. This was even after the first year of follow-up had been terminated. The increased risk remained the same after a follow-up period of more than five years [38]. A recent study using two-sample Mendelian randomization analysis also revealed a correlation between an increased risk of OC and a genetic predisposition to hyperthyroidism. This finding suggests a possible causal relationship between hyperthyroidism and the development of OC [4]. Furthermore, this study demonstrated a potential link between thyroid dysfunction and increased mortality risk in OC patients. This assertion is supported by the findings of Journy et al. [5], who reported that OC patients diagnosed with thyroid dysfunction and treated with additional radioactive iodine presented a greater mortality risk than women with a healthy thyroid in the control group. In addition, studies have reported on a few cases where women with hyperthyroidism were at an elevated risk of OC mortality [5]. The Ovarian Cancer Association Consortium (OCAC) reported that a history of hyperthyroidism was linked to worse five-year survival. A diagnosis of hyperthyroidism within five years before OC diagnosis was associated with an increased risk of death; however, no significant relationship between hyperthyroidism history and OC mortality was observed [39]. A study conducted by Ramadan [40] reported that hyperthyroidism was significantly associated with an elevated risk of OC mortality.
In contrast, a cohort study of hyperthyroidism in relation to gynecological cancers revealed no significant association with the risk of OC. The present study sought to evaluate the link between self-reported physician-diagnosed hyperthyroidism in the Nurses’ Health Study (NHS) and the risk of medical record-confirmed OC [41]. In the cohort study by Leung et al. [42], the Taiwan National Health Insurance Research Database was utilized to investigate the correlation between hyperthyroidism and gynecological cancers in women from 2000 to 2018. Studies have indicated that women diagnosed with hyperthyroidism exhibit a significantly reduced risk of developing gynecological cancers, including endometrial cancer (EC), uterine corpus cancer (UC), and OC, irrespective of the specific cancer type [42]. Table 1 summarizes the effects of hyperthyroidism on OC risk and mortality. In summary, few studies have shown no correlation or a negative correlation between hyperthyroidism and the risk and mortality of OC, but the vast majority of studies have shown a positive association. Therefore, on the basis of the data presented here, we concluded that the prevalence of hyperthyroidism is related to an increased risk and mortality of OC.

4.2.2. Hypothyroidism

Leung et al.’s cohort studies indicated that women with hypothyroidism presented a marginally elevated OC risk, a propensity that remained consistent irrespective of pharmaceutical interventions [42]. OCAC reported that an overall history of hypothyroidism was associated with worse five-year survival. A history of hypothyroidism was linked to a slightly increased risk of death [39].
Conversely, a study utilizing high-sensitivity chemiluminescence demonstrated that hypothyroidism does not impact the progression or prognosis of OC. The findings are somewhat contradictory, thereby hindering the ability to establish a definitive cause–effect relationship between thyroid dysfunction and OC [43]. A cohort study of hypothyroidism in relation to gynecological cancers also revealed no significant association with the risk of OC [41]. In addition, no such correlation was found between OC and hypothyroidism, the level of thyroid-stimulating hormone (TSH), or fT4 [4]. Furthermore, hypothyroidism has no statistically significant relationship with mortality in patients with OC [40]. The associations of hypothyroidism with OC risk and mortality are summarized in Table 1. There are fewer trials describing the relationships between hypothyroidism and OC risk and mortality, but most of them did not find a statistically significant association. On the basis of the small number of studies presented, it is not possible to establish a clear link between the incidence of hypothyroidism and OC, so further research and analysis are needed before a definitive conclusion can be reached.

4.3. Indirect Dependence of Thyroid Dysfunction and Ovarian Cancer

Furthermore, an indirect correlation between hypothyroidism and an elevated risk of OC has been indicated. The presence of hypothyroidism has been demonstrated to contribute to the development of polycystic ovary syndrome (PCOS), which affects ovulation and hormone balance [44]. This, in turn, has been linked to an increased risk of OC due to elevated androgen exposure [45]. According to a 2009 meta-analysis by Chittenden et al. [46], the incidence of OC is doubled in women with PCOS. Hansen et al. [47] reported a case of primary hypothyroidism with ovarian cyst formation and myxedematous infiltration of the ovary without luteinization.

5. The Expression Levels of Nuclear and Membrane Thyroid Hormone Receptors in Ovarian Cancer

5.1. The Expression of Nuclear Thyroid Hormone Receptors (TRs) in Ovarian Cancer: Classical Genomic Actions

THs bind with nuclear TR proteins, inducing transcription and activating or inhibiting various downstream effects on target genes [48]. Thyroid hormone receptors (TRs) belong to the nuclear receptor superfamily. Two TR proteins, TRα and TRβ, are encoded by two separate genes, THRA and THRB [49,50]. Distinct splicing forms of both TRα (TRα1, TRα2, and TRα3) and TRβ (TRβ1 and TRβ2) have been identified [51]. TRα1 is the sole protein that binds T3 and forms dimers with the truncated THRA gene products TRα2 and TRα3, which cannot bind this hormone [51]. However, both TRβ isoforms, TRβ1 and TRβ2, have been shown to bind T3 [52]. The binding of T3 to its receptors is critical for the biological effects of T3 across the body (Figure 4). TRα1 is present in many tissues, mainly in the heart, bones, and brain, whereas TRβ1 is more highly expressed in the liver, kidneys, and thyroid gland. In contrast, TRβ2 is found in the pituitary gland, hypothalamus, retina, and inner ear [53]. Since TRs have long been understood to function primarily by regulating gene transcription in the nucleus, their cytoplasmic localization could be interpreted as a regulator of their activity. Although TRs are generally considered to be predominantly located within the nucleus, TRα1 and TRβ1 have also been identified as capable of rapid shuttle movement between the nucleus and cytoplasm [54,55]. Furthermore, TR expression was observed in ovarian cells and OC tissue. As demonstrated by experimental studies, primary ovarian surface epithelial cells that potentially develop OC exhibit strong expression of TRs at the mRNA and protein levels [37,56].
According to the analysis via the GEPIA database, the expression level of THRA was significantly lower in OC than in noncancer ovarian tissue (Figure 5A; p < 0.05). However, the GEPIA database revealed no significant differences in the expression level of THRB between OC and noncancer ovarian tissues (Figure 5B). Importantly, these data were collected only for EOC, which accounts for the majority of OC cases, and did not consider the stage of the tumors.
Information on the expression levels of TRs (TRα and TRβ) in OC by type and stage is available from experimental literature data. Ditsch et al. [57] reported that among all OC subtypes, CC had the highest expression of TRα. Furthermore, nuclear TRα was associated with decreased survival in this subtype. There was a direct link between nuclear TRα1 or TRα2 expression and better survival in EOC patients, except in the CC subgroup. The group of OC patients with different CC subtypes seemed to have different characteristics in terms of TRα expression. In contrast, a shift in TRα1 or TRα2 expression to the cytoplasm seemed to be associated with reduced overall survival in EOC patients [57]. In the context of OC, both TRβ and its isoform TRβ1 are extensively expressed, with these proteins extending beyond the tumor cell nucleus to the cytoplasm. However, the subcellular distribution of these proteins varies among distinct histologic subtypes, grades, and FIGO stages. Notably, TRβ positivity has been strongly associated with decreased overall survival in OC patients. This correlation is attributed primarily to patients exhibiting complete or incomplete cytoplasmic shifts in TRβ. However, multivariate testing revealed that the prognostic significance of TRβ or its localization was lost [58]. Research has highlighted that the importance of TRs in terms of the progression and survival of OC patients is not contingent upon their level of expression but rather upon their localization.
However, Kaplan–Meier survival analysis revealed better overall survival (OS) and progression-free survival (PFS) in all OC patients of all stages and grades with high THRA expression and worse OS and PFS in all OC patients with high THRB expression. The risk of death is decreased by approximately 13% in OC patients with high THRA expression and increased by approximately 55% in OC patients with high THRB expression. The median survival of OC patients with low THRA expression was estimated to be 44 months, and that of OC patients with high THRA expression was 46.6 months. The median survival of OC patients with low THRB expression was estimated to be 57 months, and that of OC patients with high THRB expression was 38.4 months (Figure 6A,C; p = 0.042 and p < 0.001, respectively). Furthermore, PFS was decreased by approximately 52% in OC patients with high THRB expression (median survival: 19.55 months for OC patients with low THRB expression and 16 months for OC patients with high THRB expression) (Figure 6C,D; p = 0.058 and p < 0.001, respectively).
The available studies do not allow definitive conclusions to be drawn. However, in summary, the database revealed a correlation between poor overall survival in OC patients with low THRB expression and high THRA expression. In addition, research has shown that the level of TRα and TRβ expression and the correlation of TR expression with survival in OC patients are dependent on tumor type and stage. These studies highlighted that the subcellular localization of TRs, particularly the transition from nuclear to cytoplasmic localization, is a more significant predictor of survival in patients with OC than the level of TR expression. However, further studies are needed to support this.

5.2. Integrin αvβ3 Expression in Ovarian Cancer: Nongenomic Mechanism of Action

As demonstrated in the study by Bergh et al. [59], nongenomic actions of TH (T3 and T4) have been shown to bind to integrin αvβ3, which is a structural protein located within the plasma membrane, thereby activating protein kinase/extracellular signal-regulated kinase pathways [59,60] (Figure 4). The integrin αvβ3, also known as the vitronectin receptor, consists of a 125 kDa αv subunit (encoded by the ITGAV gene) and a 105 kDa β3 subunit (encoded by the ITGB3 gene) [61]. The integrin αvβ3 pathway is critical for regulating processes such as cell proliferation, apoptosis, and metastasis in cancer cells [62].
GEPIA database analysis of 426 samples of EOC and 88 noncancer ovarian tissues, without considering the stage of the tumor, revealed that the expression levels of ITGAV and ITGB3 were not significantly different between OC and noncancer ovarian tissues (Figure 7A).
However, experimental data have provided evidence both for the current expression of integrin αvβ3 and for its role in the progression of OC. In their 2020 study, Seraya-Bareket et al. [63] reported the presence of the nuclear integrin αvβ3 in HGSC cells and tissues but not in normal ovaries or fallopian tubes (FTs). The nuclear integrin αvβ3 was found to be functionally active. This study demonstrated that OVCAR-3 cells with high expression of nuclear integrin αvβ3 exhibited increased proliferation and oncogenic signaling, inhibiting migration and intact colony formation ability. Proteomic analyses revealed a network of nuclear integrin αvβ3-bound proteins, many of which have key cancer-related activities. As Shinderman-Maman et al. [6] demonstrated, T3 has been shown to bind to the integrin αvβ3, which is overexpressed in OC cells. The OVCAR-3 cell line, which represents HGSC (type II EOC), presented the highest level of integrin αvβ3. In contrast, LGSC (type I EOCs (SKOV-3 and A2780 cell lines)) presented lower expression of integrin αvβ3. These findings suggest that higher levels of integrin expression are observed in late-stage disease [6]. Furthermore, the expression of integrin αvβ3 is less common in EOCs with low malignant potential, which is characterized by a histological absence of invasion and a very good prognosis. This is in contrast to invasive OC [64]. As Wang et al. [65] demonstrated, the expression rate of integrin αvβ3 in malignant EOC was significantly greater than that in the borderline benign group and the normal ovary group. In addition, T4-induced nuclear accumulation of the nuclear αvβ3 integrin transcription complex enhances OC proliferation [66]. The EOC integrin αvβ3-transfected cell model used by Dolinschek et al. [67] demonstrated the ability of integrin αvβ3 to promote the activation of key players involved in anoikis resistance, including epidermal growth factor receptor (EGF-R), mitogen-activated protein kinase (MAPK), focal adhesion kinase (FAK), src, and PKB/Akt kinases, as well as the induction of antiapoptotic factors. Furthermore, research findings have demonstrated that the interaction between integrin αvβ3 plays a pivotal role in stimulating the proliferation and motility of OC cells (OV-MZ-6) [68]. Lössner et al.’s [69] findings suggest that the upregulation of EGF-R expression and activity in human OCs is mediated by integrin αvβ3. In addition, Gao et al. [70] reported a close correlation between integrin αvβ3 and Lewis and antigen expression, indicating that there are independent drug resistance-related risk factors in patients with OC. In contrast, one study by Kaur et al. [71] revealed that the inhibition of integrin αvβ3 in OC cells increased proliferation and invasion but decreased adhesion. OC patients with high tumor expression of integrin αvβ3 had significantly better overall survival, suggesting that integrin αvβ3 acts as a tumor suppressor in OC cells [71].
The Kaplan–Meier survival analysis revealed worse overall survival (OS) and progression-free survival (PFS) in all OC patients of all stages and grades with high mean expression of ITGAV and ITGB3. The risk of death is increased by approximately 23% in OC patients with high mean expression of IGAV and ITGB3. The median survival of OC patients with low mean expression of ITGAV and ITGB3 was estimated to be 47.94 months, and that of OC patients with high mean expression of ITGAV and ITGB3 was 40.54 months. Progression-free survival was reduced by approximately 20% (median survival: 20.77 months for OC patients with low mean expression of ITGAV and ITGB3 and 18 months for OC patients with high mean expression of ITGAV and ITGB3) (Figure 8A,B; p = 0.0029 and p = 0.01, respectively).
In conclusion, studies indicate that integrin αvβ3 is overexpressed in OC, highlighting differences in its expression levels and localization depending on the OC type and disease stage. Moreover, databases have indicated that high expression levels of integrin αvβ3 in OC patients are correlated with poor overall survival and progression-free survival. Furthermore, on the basis of the numerous studies presented, we conclude that integrin αvβ3 plays a role in OC proliferation and metastasis.

6. Relationship Between Thyroid Hormone Levels and the Progression of Ovarian Cancer

6.1. Thyroid Hormone Levels in Ovarian Cancer Patients

THs play an important role in several key processes, including growth, development, tissue homeostasis, and cancer [48]. Given the established relationship between thyroid disorders and alterations in TH expression on one hand, and ovulation and estrogen levels on the other hand, further exploration into the possible association with OC is needed.
A study by Rasool et al. [72] revealed elevated levels of T4, but no statistically significant differences were observed in the levels of T3 in the serum samples compared with those in the control group. However, the TH levels in patients with OC deviated from the standard reference ranges compared with those observed in healthy individuals [72]. Studies conducted on samples from 100 healthy women and 144 women with diagnosed gynecological cancer (128 cases of OC and 16 cases of endometrioid cancer (EC)) indicated a deranged thyroid profile with gynecological cancer. Elevated serum TSH levels are associated with the occurrence of OC and EC [73].
Correlation analysis revealed abnormal levels of TSH, fT3, fT4, and thyroid-stimulating hormone receptor (TRAb) antibodies in the serum of OC patients. The positive rates of TSH, fT3, fT4, and TRAb varied according to the type of OC (serous cystadenocarcinoma showed higher rates than did mucinous cystadenocarcinoma and other types), stage (lower rates in stages III and IV than in stages I and II), and differentiation (higher rates in poorly differentiated OC than in moderately differentiated and well-differentiated OC compared with the control group). In addition, significant differences in the serum levels of TSH, fT3, fT4 and TRAb were detected compared with those in the control group, both before and after surgery and after four cycles of OC [74].
In conclusion, determining a direction of change on the basis of these data is difficult, although several meta-analyses have shown abnormalities in the TH profile of patients with OC.

6.2. The Role of Thyroid Hormones in Ovarian Cancer Progression

T3 exerts direct inflammatory effects on ovarian surface epithelial (OSE) cell function in vitro, with the potential to affect the tumor microenvironment. As shown in in vitro studies, T3 affects OSE cells by inducing inflammation in OSE cells and increasing the expression of inflammation-associated genes such as cyclooxygenase-2 (COX-2) and matrix metalloproteinase-9 (MMP-9). In addition, T3 stimulates the expression of estrogen receptor alpha (ERα) mRNA, which is strongly associated with OC progression [37] (Figure 9).
Recent findings have indicated a potential correlation between exposure to THs and the development and progression of OC. An in vitro study demonstrated that physiological levels of T3 (1 nM) and T4 (100 nM) can induce MAPK-dependent proliferation and increase the survival of OC cells (OVCAR-3) after three- and five-day incubation. Furthermore, in A2780 cells, longer incubation with either hormone (five days) was needed to observe the effect on A2780 cell viability. Conversely, exposure to these hormones at concentrations below normal serum levels may reduce the growth of OC cells by approximately 10% (OVCAR-3). Additionally, the inhibitory effects of THs on the expression of six genes associated with tumor suppression (GDF-15, IGFBP-6), apoptosis (Nix, PUMA), and cell cycle regulation (p21, p16) were documented in OC cell lines [6]. Two years later, Shinderman-Maman et al. [75] reported that THs can downregulate the expression of apoptosis-related genes (Apaf-1, NOXA, Caspase-3, BAX, FAS, Bnip3), a tumor suppressor gene (ZMAT-3) and two DNA repair genes (ERCC5 and PolR2A). This finding highlights the role of THs in influencing the molecular pathways related to OC progression [75]. In addition, Shinderman-Maman et al. [76] reported that derivatives of TH, including tetrac, triac, and T1AM, have the capacity to reduce proliferation and induce cell death and DNA damage in the OC cell lines OVCAR-3 and A2780. This finding suggests a potential therapeutic approach for OC [76]. Furthermore, studies by Hsieh et al. [66] reported the proliferative effects of THs (100 nM T4 and 10 nM T3) on OC cells (OVCAR-3 and SKOV-3). These processes are initiated via integrin αvβ3, which has the potential for subsequent crosstalk with ERα. The activation of ERK1/2 by THs was found to be integrin αvβ3-dependent. Furthermore, it was determined that activated ERK1/2 was responsible for the phosphorylation of ERα. The collaboration between these two signaling pathways is pivotal in promoting OC cell proliferation. The findings of this study provide a novel direction for the management of ERα-positive OC, with a focus on the role of THs in the absence of host estrogen [66] (Figure 9).
Yu et al. [77] reported that T3 increases cell viability and proliferation in ovarian adult granulosa cell tumors (KGN cell line). This effect is mediated by the TR and involves the phosphorylation of signal transducer and activator of transcription (STAT3) and the expression of octamer-binding transcription factor 4 (OCT4) proteins. Blocking TR or inhibiting STAT3 reduces T3-induced cell growth, suggesting that T3 promotes cell development via the STAT3-OCT4 pathway, which is associated with poor overall survival in OC patients [77]. Our previous studies demonstrated that T3, in a dose-dependent manner ranging from 0.1 nM to 100 nM, increased the viability of nonluteinized ovarian granulosa cells (HGrC1) and rare OC cells (COV434 and KGN cells). This effect was observed in comparison with that in untreated control cells [9]. A report by Verga Falzacappa et al. [78] indicated that T3, but not T4, is capable of protecting COV434 cells from apoptosis. This protective effect is achieved through the regulation of the cell cycle and cell growth. Moreover, the protective effect of T3, as evaluated by FACS analysis in the presence of a PI3K inhibitor, is significant. This finding was further substantiated by Western blot analysis of pAkt, which confirmed the crucial role of the PI3K pathway in T3 survival [78] (Figure 9).
Furthermore, T3 can enhance angiogenesis and tumor metastasis [79]. In OC, T3 through the integrin αvβ3 pathway is involved in epithelial-to-mesenchymal transition (EMT) activity. This finding highlights a novel role for THs in OC metastasis [80] (Figure 9). In addition, T4 inhibits resveratrol-induced apoptosis via programmed death-ligand 1 (PD-L1) in in vitro model OC cells. The inhibition of resveratrol-induced nuclear accumulation of COX-2 and increased expression and cytoplasmic accumulation of PD-L1 were observed after treatment with T4 [81].
In summary, the available information from in vitro studies strongly suggests that THs may influence increased inflammation in OSE as well as OC progression and metastasis. To date, the molecular mechanism by which THs promote OC proliferation and inhibit OC apoptosis by regulating the expression of suppressive tumor genes, cell cycle regulatory genes, apoptosis genes, and DNA repair genes has been described.
Figure 9. Schematic representation of the possible actions of thyroid hormones (THs) in the ovarian surface epithelium (OSE) and ovarian cancer (OC). T3 exerts direct inflammatory effects on OSE. THs increase OC progression (both nonepithelial OC and epithelial OC (EOC)) by regulating the proliferation and expression of genes associated with tumor suppression, apoptosis, DNA repair, and cell cycle regulation. T3 promotes OC metastasis by enhancing epithelial-to-mesenchymal transition (EMT). ↑—significant increase, ↓— significant decrease; COX-2—cyclooxygenase-2; MMP-9—matrix metalloproteinase-9; STAT3—signal transducer and activator of transcription 3; OCT4—octamer-binding transcription factor 4; ERCC5—gene encoding the DNA repair protein complementing XP-G cells; PolR2A—gene encoding the DNA-directed RNA polymerase II subunit RPB1; Erα—estrogen receptor alpha; GDF-15—growth/differentiation factor 15; IGFBP-6—insulin-like growth factor-binding protein 6; ZMAT-3—zinc finger matrin-type 3; p21—cyclin-dependent kinase inhibitor 1; p16—cyclin-dependent kinase inhibitor 2A; Nix—membrane-anchored outer mitochondrial protein; PUMA—p53 upregulated modulator of apoptosis; Apaf-1—apoptotic protease activating factor 1; NOXA—phorbol-12-myristate-13-acetate-induced protein 1; BAX—bcl-2-like protein 4; FAS—cell surface death receptor; Bnip3—BCL2 interacting protein 3; COV434—juvenile granulosa tumor cell line; KGN—adult granulosa tumor cell line; OVCAR-3 and SKOV-3—epithelial OC (EOC) cell lines [6,9,37,66,75,77,78,80].
Figure 9. Schematic representation of the possible actions of thyroid hormones (THs) in the ovarian surface epithelium (OSE) and ovarian cancer (OC). T3 exerts direct inflammatory effects on OSE. THs increase OC progression (both nonepithelial OC and epithelial OC (EOC)) by regulating the proliferation and expression of genes associated with tumor suppression, apoptosis, DNA repair, and cell cycle regulation. T3 promotes OC metastasis by enhancing epithelial-to-mesenchymal transition (EMT). ↑—significant increase, ↓— significant decrease; COX-2—cyclooxygenase-2; MMP-9—matrix metalloproteinase-9; STAT3—signal transducer and activator of transcription 3; OCT4—octamer-binding transcription factor 4; ERCC5—gene encoding the DNA repair protein complementing XP-G cells; PolR2A—gene encoding the DNA-directed RNA polymerase II subunit RPB1; Erα—estrogen receptor alpha; GDF-15—growth/differentiation factor 15; IGFBP-6—insulin-like growth factor-binding protein 6; ZMAT-3—zinc finger matrin-type 3; p21—cyclin-dependent kinase inhibitor 1; p16—cyclin-dependent kinase inhibitor 2A; Nix—membrane-anchored outer mitochondrial protein; PUMA—p53 upregulated modulator of apoptosis; Apaf-1—apoptotic protease activating factor 1; NOXA—phorbol-12-myristate-13-acetate-induced protein 1; BAX—bcl-2-like protein 4; FAS—cell surface death receptor; Bnip3—BCL2 interacting protein 3; COV434—juvenile granulosa tumor cell line; KGN—adult granulosa tumor cell line; OVCAR-3 and SKOV-3—epithelial OC (EOC) cell lines [6,9,37,66,75,77,78,80].
Biomolecules 15 00870 g009

6.3. The Role of Iodothyronine Deiodinases in Ovarian Cancer Progression

TH deiodinases play pivotal roles in various physiological processes, including development, growth, and metabolic regulation. These enzymes function as regulatory agents, maintaining the balance of THs within the body. To date, three distinct deiodinases have been identified: iodothyronine deiodinase 1 (DIO1), DIO2, and DIO3 [82]. DIO1 and DIO2 convert T4 or reverse T3 (rT3) to T3 [83] (Figure 4).
As Moskovich et al. [84] reported, an increase in T3 bioavailability through stable knockdown of DIO3 (DIO3-KD) resulted in the induction of apoptosis and the attenuation of proliferation, migration, colony formation, oncogenic signaling, and tumor growth in a mouse model of HGSC. Lower levels of DIO1 were observed in HGSC tissues than in normal tissues. TCGA analyses confirmed that low DIO1 mRNA expression was correlated with poorer survival and resistance to therapy in HGSC patients. Silencing or inhibiting this enzyme resulted in increased HGSC proliferation. These studies showed that DIO1 has a potential role in suppressing OC [85]. However, DIO2 has been shown to act as a tumor promoter in OC and is proposed as an oncogenic marker [86].

7. Thyroid Dysfunctions and Ovarian Cancer Metabolism

There is evidence that THs have a strong effect on glucose metabolism, which was reported more than 100 years ago [87]. The uninterrupted proliferation that is characteristic of cancer, including OC, is connected with a phenomenon termed the “Warburg effect”. This effect refers predominantly to the production of energy through aerobic glycolysis rather than mitochondrial respiration [88]. The associations of metabolic reprogramming and the Warburg effect with THs in a variety of cancers, including breast cancer and renal cell carcinoma, have been described in in vitro studies [89,90].
Only four studies have reported the role of THs in OC metabolism. Studies suggest that T3 plays a role in the PI3K/AKT/mTOR pathway, which is frequently altered in OC and is involved in cellular metabolism and tumor progression [37,91]. In vivo studies revealed that stable DIO3 knockdown, which led to increased T3 bioavailability in HGSC cells, promoted metabolic reprogramming by inducing the Warburg effect in a HGSC mouse model [84]. In our recent study, no alteration in the metabolic activity of adult granulosa cell tumor (KGN) cells was detected after treatment with T3 (10 nM). However, a decrease in metabolic activity was observed following treatment with GW0742 (a potential novel TRα and TRβ antagonist) at a dose of 10 μM. The findings indicated a decrease in total ATP levels, accompanied by a decrease in glycoATP and mitoATP production rates, as well as an alteration in the oxygen consumption rate (OCR) and the extracellular acidification rate (ECAR). These observations suggest a substantial reduction in the mitochondrial and glycolytic pathways in KGN cells [9].
In summary, despite the potential role of THs in the regulation of OC metabolism, there is an emerging knowledge gap. This is due to the lack of research on this topic.

7.1. Associations Between Thyroid Dysfunction and the Blood Levels of Metabolic Hormones, Including Adipokines

Single cells or spheroids of OC cells are capable of surviving in nonadherent conditions, as evidenced by their resistance to anoikis (a specific form of apoptosis triggered by detachment from the extracellular matrix) [92,93]. This process is closely linked to the regulation of metabolic reprogramming, which can be regulated by individual metabolic hormones, including adipokines, which are released by adipose tissue [94,95]. The detection of adipokines (e.g., visfatin, adiponectin, and leptin) in the peritoneal fluid of OC patients suggests that they may act on OC cells, which have receptors for certain metabolic hormones on their surface [96,97,98]. Therefore, as key mediators of metabolic regulation, they can alter OC metabolism via paracrine signaling [99]. The impact of THs on metabolic hormone production has received little attention from the scientific community.

7.1.1. Insulin

The Mendelian randomization data revealed that an increased risk of OC is associated with a decreased rate of insulin secretion. These findings suggest that glucose metabolism, which is regulated by insulin secretion, plays an important role in the pathogenesis of OC, which involves the expression of a functional insulin receptor [100,101]. Furthermore, Joung et al. [102] reported that hyperinsulinemia is linked with an elevated risk of OC in women after menopause.
THs have been shown to induce hyperinsulinemia and stimulate insulin secretion [103,104]. Yang et al. [105] reported that subclinical hypothyroidism is connected with increased hyperinsulinemia. In addition, the half-life of insulin is reduced by an increased rate of degradation in diabetic patients with hyperthyroidism [106]. Furthermore, insulin resistance has been described as a result of thyroid dysfunction, both hyperthyroidism and hypothyroidism. In hyperthyroidism, an imbalance of THs can lead to glucose intolerance, mainly due to hepatic insulin resistance [107]. Another study confirmed that hypothyroidism leads to decreased synthesis and release of insulin [108] and insulin resistance [109]. Moreover, insulin also regulates TH levels by increasing fT4 while suppressing T3 levels by inhibiting the conversion of T4 to T3 [110] (Table 2).

7.1.2. Adiponectin and Leptin

The roles of adiponectin and leptin in OC have been linked. The expression of adiponectin (AdipoR1) and leptin receptor was detected in OC cells [97,98]. In a seminal study, Diaz et al. [111] demonstrated that a high leptin/adiponectin ratio was correlated with a poor prognosis for patients diagnosed with OC.
The study revealed higher concentrations of serum adiponectin in patients with hyperthyroidism than in patients with hypothyroidism (5.73 vs. 3.0 ng/mL) and revealed a positive correlation between adiponectin levels and fT4 and fT3 [112]. The levels of leptin in the serum did not change with thyroid functional status [112] (Table 2).

7.1.3. Apelin

The presence of apelin and its secretion at low concentrations (<0.3 ng/mL) has been identified in HGSC [113,114]. Moreover, apelin and its receptor (APJ) are expressed in various OC cell lines [115]. Research has revealed that apelin promotes the progression and metastasis of OC by regulating energy metabolism and lymphatic vessel formation in serous OC [116]. Furthermore, human OC cells with high APJ expression presented significant increases in migration and invasion in vitro [117].
The results revealed that the serum levels of apelin were increased in hypothyroid rats. In addition, apelin in the presence or absence of THs decreases serum levels of TSH, but lowering TSH is more effective in the coadministration of T4 [118,119]. Analysis of apelin levels in different TH states revealed that apelin levels were higher in patients with hypothyroidism (4.8 ± 2.5 ng/mL) and hyperthyroidism (3.7 ± 1.9 ng/mL) than in the normal population (3.4 ± 1.4 ng/mL), but the difference was not statistically significant [120]. Zorlu et al. [121] reported that apelin levels did not differ between patients with subclinical hypothyroidism and healthy subjects (Table 2).

7.1.4. Chemerin

Chemerin was found to be abundant in the ascitic fluid of OC patients [122]. Furthermore, chemerin expression is higher in OC tissue than in noncancer ovarian tissue [123], and the chemerin receptor chemokine-like receptor 1 (CMKLR1) is expressed in OC cells [124]. The biological impact of chemerin on signaling pathways in OC has been described [125,126].
Serum chemerin levels were found to be significantly elevated by approximately 13 µg/L in patients diagnosed with hyperthyroidism compared with those in the control group (88 vs. 75 µg/L, respectively). Furthermore, a positive correlation was detected between serum chemerin concentrations and T3, T4, and fT3, whereas a negative correlation was detected between chemerin levels and TSH and FT4 [127] (Table 2).

7.1.5. Resistin

Studies by Pang and Chang [128] indicated that resistin can promote the proliferation and migration of OC cells.
A meta-analysis of the extant literature demonstrated that resistin levels are significantly higher in patients with thyroid dysfunction. This finding suggests a potential association between higher TH levels and increased resistin. Moreover, a significant decrease in resistin levels after treatment in patients with thyroid dysfunction was observed in comparison to levels before treatment [129]. As demonstrated in the literature, serum resistin concentrations were found to be higher in hyperthyroidism patients than in hypothyroidism patients (6.378 vs. 5.81 ng/mL, respectively) [112]. Additionally, the successful treatment of hyperthyroidism is associated with decreased resistin levels [130]. As demonstrated in the study by Eke Koyuncu et al. [131], an increase in resistin levels was found to be directly related to thyroid dysfunction. The levels of resistin in the hypothyroid group were significantly greater than those in the control group (12.66 and 8.45 ng/mL, respectively). Similarly, resistin levels were significantly higher in the subclinical hyperthyroid group than in the control group (14.88 and 8.45 ng/mL, respectively). Furthermore, changes in TH levels affect the synthesis and/or secretion of resistin in adipose tissue and/or macrophages [131]. Additionally, a positive correlation between resistin levels and fT3 levels in patients with thyroid dysfunction was identified [129,130]. Moreover, a positive correlation between resistin levels and fT4 levels, as well as a negative correlation with TSH, was observed [130].
Studies conducted by Owecki et al. [132] demonstrated that short-term profound hypothyroidism resulted in a decrease in resistin levels. However, no significant correlations were detected between the levels of circulating resistin and THs or TSH [132]. In addition, Peng et al. [133] reported that downregulated resistin levels in Grave’s disease patients are the consequence of decreased neutrophil counts (Table 2).

7.1.6. Visfatin (NAMPT)

Li et al. [96] reported that visfatin is present in the ascitic fluid of OC patients. The increased expression of visfatin is associated with OC progression through the upregulation of pro-proliferative cytokine secretion and the stimulation of OC growth, development, angiogenesis, and metastasis [96,134,135].
Ozkaya et al. [136] suggested that serum visfatin levels in a cohort of patients with hyperthyroidism and hypothyroidism may be modulated by THs, such as T3 and T4. Low visfatin levels were detected in patients with hyperthyroidism (9.44 ng/mL) compared with those in patients with hypothyroidism (49.93 ng/mL) and the control group (38.6 ng/mL). In patients diagnosed with hypothyroidism, a significant decrease in plasma visfatin levels was observed following treatment (58.58 ng/mL vs. 40.00 ng/mL). Furthermore, a negative correlation was identified between visfatin levels and both fT3 and fT4 levels, whereas a positive correlation was detected between visfatin and TSH levels. Moreover, visfatin, as a potential proinflammatory mediator, may contribute to proinflammatory conditions in the thyroid, leading to thyroid dysfunction [136]. Serum visfatin concentrations were found to be significantly lower in patients with hyperthyroidism than in controls. Furthermore, a negative correlation was identified between fT4 and T4 concentrations, on the one hand, and serum visfatin concentrations, on the other hand. Conversely, TSH was found to correlate positively with serum visfatin levels [127] (Table 2).

7.1.7. Vaspin

As González et al. [137] demonstrated, thyroid dysfunction has the potential to affect vaspin expression. Researchers have reported that the mRNA levels of vaspin are considerably lower in hyperthyroid rats and significantly greater in hypothyroid rats than in euthyroid rats [137]. Handisurya et al. [138] reported a significant decrease in TSH levels that was positively correlated with changes in serum vaspin levels. In contrast, a study by Cinar et al. [139] demonstrated that TH status does not influence serum vaspin levels in humans. Vaspin levels were similar in euthyroid and hypothyroid patients. However, vaspin levels do not correlate with TSH levels [139] (Table 2).

7.1.8. Omentin (ITLN1, Intelectin-1)

In patients diagnosed with neoplasia, omentin levels are lower than those observed in patients with benign gynecological lesions and healthy women. Furthermore, the downregulation of mesothelial cell-derived ITLN1 in the omental tumor microenvironment has been demonstrated to facilitate OC progression [140].
As Alshaikh et al. [127] reported, serum omentin concentrations were significantly lower in patients with hyperthyroidism than in the control group (~30 vs. ~70 µg/L). Cerit et al. [141] reported that, in hypothyroid patients, omentin-1 levels were lower than those in the control group but increased after six months of TH replacement therapy. However, no significant correlation was observed between serum omentin concentrations and any of the thyroid profile variables except fT3 [127] (Table 2).
Table 2. The blood levels of metabolic hormones, including adipokines (insulin, adiponectin, leptin, apelin, chemerin, resistin, visfatin, vaspin, and omentin), in patients with thyroid dysfunction (hyperthyroidism and hypothyroidism) and correlations between thyroid and metabolic hormones.
Table 2. The blood levels of metabolic hormones, including adipokines (insulin, adiponectin, leptin, apelin, chemerin, resistin, visfatin, vaspin, and omentin), in patients with thyroid dysfunction (hyperthyroidism and hypothyroidism) and correlations between thyroid and metabolic hormones.
Metabolic HormonesBlood Levels of Metabolic Hormones in Patients with Thyroid Dysfunction Compared with Control SubjectsCorrelation Between Metabolic Hormone Level and TH Levels
HyperthyroidismHypothyroidismPositiveNegative
insulinreduced half-life by an increased rate of degradation in diabetic patients [106]associated with increased hyperinsulinemia
[105]
fT4
[110]
T3
[110]
insulin resistance—result of both thyroid dysfunctions [107]THs induce hyperinsulinemia and stimulate insulin secretion
[103,104]
adiponectin↑ (5.73 ng/mL)
[112]
↓ (3 ng/mL)
[112]
fT3, fT4 level
[112]
Nd.
leptinno change detected [112]Nd.
apelin↓ (3.7 ± 1.9 ng/mL)
[120]
↑ 4.8 ± 2.5 ng/mL)
[120]
Nd.↓ TSH levels
(with a stronger effect when coadministered with T4)
[118,119]
normal population (3.4 ± 1.4 ng/mL)
[120]
apelin levels remain unchanged in subclinical hypothyroidism [121]
chemerin↑ 88 µg/L
[127]
Nd.TT3, TT4 and fT3 levels
[127]
TSH and fT4 levels
[127]
resistin↑ 6.378 ng/mL
[112]
↑ 14.88 ng/mL
[131]
control (8.45 ng/mL)
[131]
↑ 5.81 ng/mL [112]
↑ 12.66 ng/mL
[131]
control (8.45 ng/mL)
[131]
fT3
[129,130]
fT4
[130]
successful treatment of hyperthyroidism associated with decreased resistin levels
[112,130]
TSH
[130]
short-term profound hypothyroidism decreases resistin level
[132]
↓ in Graves’ disease, as a result of a decrease in the number of neutrophils
[133]
no significant correlations
between TH or TSH levels
[131]
visfatin↓ (9.44 ng/mL)
[136]
↑ 49.93 (ng/mL)
[136]
TSH
[127,136]
fT3
[136]
fT4 and TT4
[127,136]
control—38.6 ng/mL [136]visfatin levels is decreased by T3 and T4 [112]
vaspinNd.overt 1.20 ± 1.17 ng/mL
[139]
subclinical 1.48 ± 0.93 ng/mL
[139]
TSH
[138]
Nd.
control 0.95 ± 0.75 ng/mL
[139]
TSH level is not correlated with vaspin level
[139]
omentin↓ ~30 µg/L
[127]
↓ (before thyroid hormone therapy)
[141]
Nd.fT3
[127]
—increased compared to euthyroid control, —decreased compared to euthyroid control, THs—thyroid hormones, TSH—thyroid-stimulating hormone, T3—triijodothyronine, fT3—free triiodothyronine, T4—thyroxine, fT4—free thyroxine, TT4—total thyroxine, Nd.—no data.
In conclusion, the results of this study suggest that the levels of metabolic hormones, including adipokines, change with the occurrence of thyroid dysfunction (hyperthyroidism and hypothyroidism). Furthermore, the correlations shown between blood levels of THs and metabolic hormones confirm that metabolic hormone levels are regulated by THs. This finding provides a basis for the hypothesis that THs may exert an indirect influence on the progression of OC, possibly through the regulation of metabolic hormones whose effects on OC progression have been described. However, further molecular studies demonstrating this effect are needed to confirm this hypothesis.

8. Correlation Between the Treatment of Thyroid Dysfunction and Ovarian Cancer

8.1. Effects of Thyroid Dysfunction Treatments on Ovarian Cancer Risk and Progression

The standard treatment for patients with thyroid dysfunction is antithyroid medication, radioactive iodine ablation, and synthetic iodine or surgery [30,142]. Studies by Minlikeeva et al. [39] reported that medications used for hyperthyroidism or hypothyroidism were not associated with mortality among OC patients. A study conducted by Leung et al. [42] revealed that there was no statistically significant risk of developing gynecological cancers, including OC, in patients with hyperthyroidism or hypothyroidism after medication adjustment. Nevertheless, the impact of interferon administration on patients diagnosed with hepatitis C [143], particularly women afflicted with thyroid disease, may be associated with the development of OC [72]. In addition, Muderris et al. [144] reported that patients with hypothyroidism had significantly larger ovarian volumes than controls did, which were significantly reduced after TH therapy.

8.2. Effects of Treatments for Ovarian Cancer on Thyroid Dysfunction

In a 50-year-old woman diagnosed with stage III HGSC, after surgery and complete therapy, the TSH level was undetectable, and the fT4 level was elevated, confirming the diagnosis of Graves’ disease [145]. A 55-year-old female with OC that metastasized to the thyroid resulting in clinical hypothyroidism was reported by Skarf et al. [146]. Kimura et al. [147] reported a 31-year-old female patient with massive ascites and elevated serum CA 125 levels and no typical features of hypothyroidism.

9. Ovarian Cancer, Including Thyroid Tissue (Struma Ovarii)

Thyroid tissue may be present in OC, known as struma ovarii (SO). GeCTs account for approximately 20% of all OC cases, with up to 20% of OC cases containing thyroid tissue. However, only 5% of these cancers contain more than 50% thyroid tissue and are termed SO [148]. Notably, the vast majority of SO cases (95%) are benign, with only a small percentage (5–20%) associated with hyperthyroidism [149]. In rare instances, malignant thyroid tumors can develop within the SO, which can lead to hyperthyroidism due to the capacity of the thyroid tissue within the tumor to produce THs. Hypothyroidism, on the other hand, occurs in 51.5% of SO patients and is more prevalent in benign SO patients than in malignant patients [150]. Thyroid carcinoma on struma ovarii (TCSO) is a rare OC derived from monodermic teratomas. It represents approximately 0.01% of all OCs and 5–10% of all SOs [151]. A case report by Koehler et al. [152] described a case of a 61-year-old female patient with Hashimoto’s thyroiditis on levothyroxine therapy who progressed to clinical and biochemical hyperthyroidism despite antithyroid medication, with a new diagnosis of a mature teratoma composed predominantly of thyroid tissue with high levels of sodium iodide symporter protein expression. Following the surgical resection of SO and the subsequent resolution of autonomous TH production, the patient presented symptoms consistent with hypothyroidism, as evidenced by a significant decrease in TSH levels of both fT4 and fT3. This finding suggests secondary hypothyroidism, a diagnosis that is substantiated by the known coexistence of chronic lymphocytic thyroiditis in the orthotopic thyroid gland [152]. Furthermore, hypothyroidism in conjunction with OC has been documented in patients after SO tumor resection [153].

10. Conclusions

In conclusion, in this review, we sought to elucidate the relationships between the presence of hyperthyroidism or hypothyroidism and OC. This is very important, especially in the context of the discovery of the expression of the nuclear TRs (TRα and TRβ), as well as the membrane TR (αvβ3 integrin), in human tissues and OC cell lines. The presented data from the database revealed that changes in TR expression levels regulate survival in patients with OC. The results of these studies demonstrated that TH, through binding to these receptors, may facilitate the progression, metabolism, and metastasis of OC, confirming the correlation between thyroid dysfunction and the occurrence of OC.
Given this connection, clinicians treating OC patients with known thyroid dysfunction should consider how treating this condition might affect the effectiveness of OC treatment. Furthermore, compelling evidence may encourage gynecologic oncologists to consider thyroid status when evaluating OC patients, potentially allowing more personalized clinical management of these patients. Interventional studies could facilitate the development of novel, effective treatments, which are currently lacking in OC therapy.
However, further molecular studies are needed to elucidate the mechanism by which the HPT axis promotes the development of OC to improve the efficacy of treatment and the quality of life of OC patients with hyper- or hypothyroidism.

Author Contributions

All the authors contributed to the conception and design of the study. J.G.-M. and A.P. had the initial idea for the article. A.S., I.K., G.W., K.R. and K.K. conducted the literature search. J.G.-M. drafted the first draft of the manuscript. All the authors have commented on earlier versions of the manuscript. I.K. and G.W. prepared the figures, and A.S. and K.R. prepared the tables included in the manuscript. A.P. supervised the study. K.K. compiled the bibliography for the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding. JGM is supported by the Foundation for Polish Science (FNP) Scholarship.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were generated for this review.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Søgaard, M.; Farkas, D.K.; Ehrenstein, V.; Jørgensen, J.O.; Dekkers, O.M.; Sørensen, H.T. Hypothyroidism and hyperthyroidism and breast cancer risk: A nationwide cohort study. Eur. J. Endocrinol. 2016, 174, 409–414. [Google Scholar] [CrossRef] [PubMed]
  2. Bach, L.; Kostev, K.; Schiffmann, L.; Kalder, M. Association between thyroid gland diseases and breast cancer: A case–control study. Breast Cancer Res. Treat. 2020, 182, 207–213. [Google Scholar] [CrossRef]
  3. Ness, R.B.; Grisso, J.A.; Cottreau, C.; Klapper, J.; Vergona, R.; Wheeler, J.E.; Morgan, M.; Schlesselman, J.J. Factors related to inflammation of the ovarian epithelium and risk of ovarian cancer. Epidemiology 2000, 11, 111–117. [Google Scholar] [CrossRef]
  4. Wang, T.; Wang, X.; Wu, J.; Li, X. Cusal relationship between thyroid dysfunction and ovarian cancer: A two-sample Mendelian randomization study. BMC Cancer 2024, 24, 629. [Google Scholar]
  5. Journy, N.M.Y.; Bernier, M.O.; Doody, M.M.; Alexander, B.H.; Linet, M.S.; Kitahara, C.M. Hyperthyroidism, hypothyroidism, and cause-specific mortality in a large cohort of women. Thyroid 2017, 27, 1001–1010. [Google Scholar] [CrossRef]
  6. Shinderman-Maman, E.; Cohen, K.; Weingarten, C.; Nabriski, D.; Twito, O.; Baraf, L.; Hercbergs, A.; Davis, P.J.; Werner, H.; Ellis, M.; et al. The thyroid hormone-αvβ3 integrin axis in ovarian cancer: Regulation of gene transcription and MAPK-dependent proliferation. Oncogene 2016, 35, 1977–1987. [Google Scholar] [CrossRef]
  7. Zhang, C.; Guo, L.; Zhu, B.; Feng, Y.; Yu, S.; An, N.; Wang, X. Effects of 3,5,3’-triiodothyronine (T3) and follicle-stimulating hormone on apoptosis and proliferation of rat ovarian granulosa cells. Chin. J. Physiol. 2013, 56, 298–305. [Google Scholar]
  8. Zhu, L.; Tian, G.; Yang, Q.; De, G.; Zhang, Z.; Wang, Y.; Nie, H.; Zhang, Y.; Yang, X.; Li, J. Thyroid hormone receptor β1 suppresses proliferation and migration by inhibiting PI3K/Akt signalling in human colorectal cancer cells. Oncol. Rep. 2016, 36, 1419–1426. [Google Scholar] [CrossRef] [PubMed]
  9. Gogola-Mruk, J.; Kumor, I.; Wojtaszek, G.; Kulig, K.; Ptak, A. GW0742 as a potential TRα and TRβ antagonist reduces the viability and metabolic activity of an adult granulosa tumour cell line and simultaneously upregulates TRβ expression. Cancers 2024, 16, 4069. [Google Scholar] [CrossRef]
  10. Tang, Z.; Li, C.; Kang, B.; Gao, G.; Li, C.; Zhang, Z. GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017, 45, W98–W102. [Google Scholar] [CrossRef]
  11. Győrffy, B. Discovery and ranking of the most robust prognostic biomarkers in serous ovarian cancer. GeroScience 2023, 45, 1889–1898. [Google Scholar] [CrossRef] [PubMed]
  12. Huang, J.; Chan, W.C.; Ngai, C.H.; Lok, V.; Zhang, L.; Lucero-Prisno, D.E.; Xu, W.; Zheng, Z.J.; Elcarte, E.; Withers, M.; et al. Worldwide burden, risk factors, and temporal trends of ovarian cancer: A global study. Cancers 2022, 14, 2230. [Google Scholar] [CrossRef] [PubMed]
  13. 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 Cancer J. Clin. 2020, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  14. Ferlay, J.; Colombet, M.; Soerjomatara, I.; Parkin, D.M.; Piñeros, M.; Znaor, A.; Bray, F. Cancer statistics for the year 2020: An overview. Int. J. Cancer 2021, 149, 778–789. [Google Scholar] [CrossRef]
  15. Ferlay, J.; Ervik, M.; Lam, F.; Laversanne, M.; Colombet, M.; Mery, L.; Piñeros, M.; Znaor, A.; Soerjomataram, I.; Bray, F. Global Cancer Observatory: Cancer Today; International Agency for Research on Cancer: Lyon, France, 2024; Available online: https://gco.iarc.who.int/today (accessed on 3 March 2025).
  16. Prat, J.; FIGO Committee on Gynecologic Oncology. Staging classification for cancer of the ovary, fallopian tube, and peritoneum. Int. J. Gynaecol. Obstet. 2014, 124, 1–5. [Google Scholar] [CrossRef]
  17. Gaitskell, K.; Hermon, C.; Barnes, I.; Pirie, K.; Floud, S.; Green, J.; Beral, V.; Reeves, G. K; Million Women Study Collaborators. Ovarian cancer survival by stage, histotype, and prediagnostic lifestyle factors, in the prospective UK Million Women Study. Cancer Epidemiol. 2022, 76, 102074. [Google Scholar] [CrossRef]
  18. Mori, K.M.; Neubauer, N.L. Minimally invasive surgery in gynecologic oncology. Obs. Gynecol 2013, 312982. [Google Scholar] [CrossRef]
  19. Flaum, N.; Crosbie, E.J.; Edmondson, R.J.; Smith, M.J.; Evans, D.G. Epithelial ovarian cancer risk: A review of the current genetic landscape. Clin. Genet. 2020, 97, 54–63. [Google Scholar] [CrossRef]
  20. Zamwar, U.M.; Anjankar, A.P. Aetiology, epidemiology, histopathology, classification, detailed evaluation, and treatment of ovarian cancer. Cureus 2022, 14, e30561. [Google Scholar] [CrossRef]
  21. Kurman, R.J.; Shih, I.M. The dualistic model of ovarian carcinogenesis: Revisited, revised, and expanded. Am. J. Pathol. 2016, 186, 733–747. [Google Scholar] [CrossRef]
  22. Van Nieuwenhuysen, E.; Lambrechts, S.; Lambrechts, D.; Leunen, K.; Amant, F.; Vergote, I. Genetic changes in nonepithelial ovarian cancer. Expert Rev. Anticancer. Ther. 2013, 13, 871–882. [Google Scholar] [CrossRef] [PubMed]
  23. Pectasides, D.; Pectasides, E.; Psyrri, A. Granulosa cell tumor of the ovary. Cancer Treat. Rev. 2008, 34, 1–12. [Google Scholar] [CrossRef]
  24. Khan, Y.S.; Farhana, A. Histology, Thyroid Gland; StatPearls: Treasure Island, FL, USA, 2021. [Google Scholar]
  25. Kelly, G.S. Peripheral metabolism of thyroid hormones: A review. Altern. Med. Rev. 2000, 5, 306–333. [Google Scholar]
  26. Vagenakis, A.G. Pituitary-thyroid interaction: Effects of thyroid hormone, non thyroidal illness and various agents on TSH secretion. Acta Med. Austriaca 1988, 15, 52–56. [Google Scholar] [PubMed]
  27. Nikrodhanond, A.A.; Ortiga-Carvalho, T.M.; Shibusawa, N.; Hashimoto, K.; Liao, X.H.; Refetoff, S.; Yamada, M.; Mori, M.; Wondisford, F.E. Dominant role of thyrotropin-releasing hormone in the hypothalamic-pituitary-thyroid axis. J. Biol. Chem. 2006, 281, 5000–5007. [Google Scholar] [CrossRef]
  28. Mathew, P.; Kaur, J.; Rawla, P. Hyperthyroidism; StatPearls: Treasure Island, FL, USA, 2023. [Google Scholar]
  29. Chaker, L.; Bianco, A.C.; Jonklaas, J.; Peeters, R.P. Hypothyroidism. Lancet 2017, 390, 1550–1562. [Google Scholar] [CrossRef] [PubMed]
  30. De Leo, S.; Lee, S.Y.; Braverman, L.E. Hyperthyroidism. Lancet 2016, 388, 906–918. [Google Scholar] [CrossRef]
  31. Garmendia Madariaga, A.; Santos Palacios, S.; Guillén-Grima, F.; Galofré, J.C. The incidence and prevalence of thyroid dysfunction in Europe: A meta-analysis. J. Clin. Endocrinol. Metab. 2014, 99, 923–931. [Google Scholar] [CrossRef] [PubMed]
  32. Hollowell, J.G.; Staehling, N.W.; Flanders, W.D.; Hannon, W.H.; Gunter, E.W.; Spencer, C.A.; Braverman, L.E. Serum TSH, T4, and thyroid antibodies in the United States population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). J. Clin. Endocrinol. Metab. 2002, 87, 489–499. [Google Scholar] [CrossRef]
  33. Savant, S.S.; Sriramkumar, S.; O’Hagan, H.M. The role of inflammation and inflammatory mediators in epithelial ovarian cancer. Cancers 2018, 10, 251. [Google Scholar] [CrossRef]
  34. Silva, J.F.; Ocarino, N.M.; Serakides, R. Thyroid hormones and female reproduction. Biol. Reprod. 2018, 99, 907–921. [Google Scholar] [CrossRef] [PubMed]
  35. Tresa, A.; Rema, P.; Suchetha, S.; Dinesh, D.; Sivaranjith, J.; Nath, A.G. Hypothyroidism presenting as ovarian cysts: A case series. Indian J. Surg. Oncol. 2021, 12, 343–347. [Google Scholar] [CrossRef]
  36. Perveen, R.; Asaduzzaman, M.D. Massive ovarian enlargement with multiple cysts mimicking ovarian neoplasm and primary hypothyroidism. Bangladesh J. Obs. Gynaecol. 2020, 33, 78–81. [Google Scholar] [CrossRef]
  37. Rae, M.T.; Gubbay, O.; Kostogiannou, A.; Price, D.; Critchley, H.O.; Hillier, S.G. Thyroid hormone signalling in human ovarian surface epithelial cells. J. Clin. Endocrinol. Metab. 2007, 92, 322–327. [Google Scholar] [CrossRef] [PubMed]
  38. Lee, J.Y.; Lee, M.K.; Lee, J.H.; Kim, K.; Bae, K.; Sohn, S.Y. Cancer risks of patients with Graves’ disease who received antithyroid drugs as initial treatment: A nationwide population-based analysis. Thyroid 2024, 34, 1271–1279. [Google Scholar] [CrossRef]
  39. Minlikeeva, A.N.; Freudenheim, J.L.; Cannioto, R.A.; Eng, K.H.; Szender, J.B.; Mayor, P.; Etter, J.L.; Cramer, D.W.; Diergaarde, B.; Doherty, A.J. History of thyroid disease and survival of ovarian cancer patients: Results from the Ovarian Cancer Association Consortium. Br. J. Cancer 2017, 117, 1063–1069. [Google Scholar] [CrossRef]
  40. Ramadan, H. Navigating the thyroid-gynecologic interplay: A systematic review and meta-analysis. Obstet. Gynecol. Sci. 2024, 67, 525–533. [Google Scholar] [CrossRef]
  41. Kang, J.H.; Kueck, A.S.; Stevens, R.; Curhan, G.; De Vivo, I.; Rosner, B.; Alexander, E.; Tworoger, S.S. A large cohort study of hypothyroidism and hyperthyroidism in relation to gynecologic cancers. Obstet. Gynecol. Int. 2013, 2013, 743721. [Google Scholar] [CrossRef] [PubMed]
  42. Leung, J.H.; Wang, S.Y.; Leung, H.W.C.; Yu, T.S.; Chan, A.L.F. Hypothyroidism and hyperthyroidism related to gynecologic cancers: A nationwide population-based cohort study. Sci. Rep. 2024, 14, 1892. [Google Scholar] [CrossRef]
  43. Elgebaly, M.M.; Abdel-Hamed, A.R.; Mesbah, N.M.; Abo-Elmatty, D.M.; Abouzid, A.; Abdelrazek, M.A. Hypothyroidism affects progression and outcomes of breast cancer but not ovarian cancer. J. Immunoass. Immunochem. 2022, 43, 288–298. [Google Scholar] [CrossRef]
  44. Mukherjee, P.; Sanyal, S.; Chadha, S.; Mukherjee, S. Impact of PCOS on ovarian cancer and thyroid disorders: A review. Endocr. Metab. Immune Disord. Drug Targets 2024, 24, 562–572. [Google Scholar] [CrossRef] [PubMed]
  45. Harris, H.R.; Terry, K.L. PCOS and risk of endometrial, ovarian, and breast cancer: A systematic review. Fertil. Res. Pract. 2016, 2, 14. [Google Scholar] [CrossRef] [PubMed]
  46. Chittenden, B.G.; Fullerton, G.; Maheshwari, A.; Bhattacharya, S. PCOS and the risk of gynecological cancer: A systematic review. Reprod. Biomed. Online 2009, 19, 398–405. [Google Scholar] [CrossRef]
  47. Hansen, K.A.; Tho, S.P.; Hanly, M.; Moretuzzo, R.W.; McDonough, P.G. Massive ovarian enlargement in primary hypothyroidism. Fertil. Steril. 1997, 67, 169–171. [Google Scholar] [CrossRef]
  48. Pascual, A.; Aranda, A. Thyroid hormone receptors, cell growth and differentiation. Biochim. Biophys. Acta 2013, 1830, 3908–3916. [Google Scholar] [CrossRef]
  49. Cheng, S.Y.; Leonard, J.L.; Davis, P.J. Molecular aspects of thyroid hormone actions. Endocr. Rev. 2010, 31, 139–170. [Google Scholar] [CrossRef]
  50. Ortiga-Carvalho, T.M.; Sidhaye, A.R.; Wondisford, F.E. Thyroid hormone receptors and resistance to thyroid hormone disorders. Nat. Rev. Endocrinol. 2014, 10, 582–591. [Google Scholar] [CrossRef] [PubMed]
  51. Brent, G.A. Mechanisms of thyroid hormone action. J. Clin. Investig. 2012, 122, 3035–3043. [Google Scholar] [CrossRef]
  52. Williams, G.R. Cloning and characterization of two novel thyroid hormone receptor beta isoforms. Mol. Cell. Biol. 2000, 20, 8329–8342. [Google Scholar] [CrossRef]
  53. Wondisford, F.E. Thyroid hormone action: Insight from transgenic mouse models. J. Investig. Med. 2003, 51, 215–220. [Google Scholar] [CrossRef]
  54. Roggero, V.R.; Zhang, J.; Parente, L.E.; Doshi, Y.; Dziedzic, R.C.; McGregor, E.L.; Varjabedian, A.D.; Schad, S.E.; Bondzi, C.; Allison, L.A. Nuclear import of the thyroid hormone receptor α1 is mediated by importin 7, importin β1, and adaptor importin α1. Mol. Cell. Endocrinol. 2016, 419, 185–197. [Google Scholar] [CrossRef] [PubMed]
  55. Subramanian, K.S.; Dziedzic, R.C.; Nelson, H.N.; Stern, M.E.; Roggero, V.R.; Bondzi, C.; Allison, L.A. Multiple exportins influence thyroid hormone receptor localization. Mol. Cell. Endocrinol. 2015, 411, 86–96. [Google Scholar] [CrossRef] [PubMed]
  56. Aghajanova, L.; Lindeberg, M.; Carlsson, I.B.; Stavreus-Evers, A.; Zhang, P.; Scott, J.E.; Hovatta, O.; Skjöldebrand-Sparre, L. Receptors for thyroid-stimulating hormone and thyroid hormones in human ovarian tissue. Reprod. Biomed. Online 2009, 18, 337–347. [Google Scholar] [CrossRef]
  57. Ditsch, N.; Heublein, S.; Jeschke, U.; Sattler, C.; Kuhn, C.; Hester, A.; Czogalla, B.; Trillsch, F.; Mahner, S.; Engel, J.; et al. Cytoplasmic versus nuclear THR alpha expression determines survival of ovarian cancer patients. J. Cancer Res. Clin. Oncol. 2020, 146, 1923–1932. [Google Scholar] [CrossRef]
  58. Heublein, S.; Jeschke, U.; Sattler, C.; Kuhn, C.; Hester, A.; Czogalla, B.; Trillsch, F.; Mahner, S.; Mayr, D.; Schmoeckel, E.; et al. Subcellular distribution of thyroid hormone receptor beta in ovarian cancer. Int. J. Mol. Sci. 2022, 23, 2698. [Google Scholar] [CrossRef]
  59. Bergh, J.J.; Lin, H.Y.; Lansing, L.; Mohamed, S.N.; Davis, F.B.; Mousa, S.; Davis, P.J. Integrin alphaVbeta3 contains a cell surface receptor site for thyroid hormone that is linked to activation of mitogen-activated protein kinase and induction of angiogenesis. Endocrinology 2005, 146, 2864–2871. [Google Scholar] [CrossRef] [PubMed]
  60. Cayrol, F.; Díaz Flaqué, M.C.; Fernando, T.; Yang, S.N.; Sterle, H.A.; Bolontrade, M.; Amorós, M.; Isse, B.; Farías, R.N.; Ahn, H.; et al. Integrin αvβ3 acting as membrane receptor for thyroid hormones mediates angiogenesis in malignant T cells. Blood 2015, 125, 841–851. [Google Scholar] [CrossRef]
  61. Liu, Z.; Wang, F.; Chen, X. Integrin alpha(v)beta(3)-targeted cancer therapy. Drug Dev. Res. 2008, 69, 329–339. [Google Scholar] [CrossRef]
  62. Yang, Y.S.H.; Ko, P.J.; Pan, Y.S.; Lin, H.Y.; Whang-Peng, J.; Davis, P.J.; Wang, K. Role of thyroid hormone-integrin αvβ3-signal and therapeutic strategies in colorectal cancers. J. Biomed. Sci. 2021, 28, 24. [Google Scholar] [CrossRef]
  63. Seraya-Bareket, C.; Weisz, A.; Shinderman-Maman, E.; Teper-Roth, S.; Stamler, D.; Arbib, N.; Kadan, Y.; Fishman, A.; Kidron, D.; Edelstein, E.; et al. The identification of nuclear αvβ3 integrin in ovarian cancer: Nonparadigmal localization with cancer promoting actions. Oncogenesis 2020, 9, 69. [Google Scholar] [CrossRef]
  64. Liapis, H.; Adler, L.M.; Wick, M.R.; Rader, J.S. Expression of alpha(v)beta3 integrin is less frequent in ovarian epithelial tumors of low malignant potential in contrast to ovarian carcinomas. Hum. Pathol. 1997, 28, 443–449. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, Y.; Liu, J.; Lin, B.; Wang, C.; Li, Q.; Liu, S.; Yan, L.; Zhang, S.; Iwamori, M. Study on the expression and clinical significances of lewis y antigen and integrin αv, β3 in epithelial ovarian tumors. Int. J. Mol. Sci. 2011, 12, 3409–3421. [Google Scholar] [CrossRef] [PubMed]
  66. Hsieh, M.T.; Wang, L.M.; Changou, C.A.; Chin, Y.T.; Yang, Y.S.H.; Lai, H.Y.; Lee, S.Y.; Yang, Y.N.; Whang-Peng, J.; Liu, L.F.; et al. Crosstalk between integrin αvβ3 and ERα contributes to thyroid hormone-induced proliferation of ovarian cancer cells. Oncotarget 2017, 8, 24237–24249. [Google Scholar] [CrossRef]
  67. Dolinschek, R.; Hingerl, J.; Benge, A.; Zafiu, C.; Schüren, E.; Ehmoser, E.K.; Lössner, D.; Reuning, U. Constitutive activation of integrin αvβ3 contributes to anoikis resistance of ovarian cancer cells. Mol. Oncol. 2021, 15, 503–522. [Google Scholar] [CrossRef] [PubMed]
  68. Hapke, S.; Kessler, H.; Luber, B.; Benge, A.; Hutzler, P.; Höfler, H.; Schmitt, M.; Reuning, U. Ovarian cancer cell proliferation and motility is induced by engagement of integrin alpha(v)beta3/Vitronectin interaction. Biol. Chem. 2003, 384, 1073–1083. [Google Scholar] [CrossRef]
  69. Lössner, D.; Abou-Ajram, C.; Benge, A.; Reuning, U. Integrin alphavbeta3 mediates upregulation of epidermal growth-factor receptor expression and activity in human ovarian cancer cells. Int. J. Biochem. Cell Biol. 2008, 40, 2746–2761. [Google Scholar] [CrossRef]
  70. Gao, J.; Hu, Z.; Liu, D.; Liu, J.; Liu, C.; Hou, R.; Gao, S.; Zhang, D.; Zhang, S.; Lin, B. Expression of Lewis y antigen and integrin αv, β3 in ovarian cancer and their relationship with chemotherapeutic drug resistance. J. Exp. Clin. Cancer Res. 2013, 32, 36. [Google Scholar] [CrossRef]
  71. Kaur, S.; Kenny, H.A.; Jagadeeswaran, S.; Zillhardt, M.R.; Montag, A.G.; Kistner, E.; Yamada, S.D.; Mitra, A.K.; Lengyel, E. {beta}3-integrin expression on tumor cells inhibits tumor progression, reduces metastasis, and is associated with a favourable prognosis in patients with ovarian cancer. Am. J. Pathol. 2009, 175, 2184–2196. [Google Scholar] [CrossRef]
  72. Rasool, M.; Naseer, M.I.; Zaigham, K.; Malik, A.; Riaz, N.; Alam, R.; Manan, A.; Sheikh, I.A.; Asif, M. Comparative study of alterations in tri-iodothyronine (T3) and thyroxine (T4) hormone levels in breast and ovarian cancer. Pak. J. Med. Sci. 2014, 30, 1356–1360. [Google Scholar]
  73. Pandey, S.K.; Pandey, H.S.; Kalra, V. Study of prevalence of thyroid disorders in pregnant females of a tertiary care hospital of Garhwal region of Uttarakhand. Ann. Int. Med. Dent. Res. 2018, 4, 25. [Google Scholar] [CrossRef]
  74. Yang, M.; Guo, X. Detection of TSH, FT3, FT4, and TRAb in ovarian cancer with highly sensitive electrochemical immunoassay. Eur. J. Inflamm. 2018, 16, 205873921879670. [Google Scholar] [CrossRef]
  75. Shinderman-Maman, E.; Weingarten, C.; Moskovich, D.; Werner, H.; Hercbergs, A.; Davis, P.J.; Ellis, M.; Ashur-Fabian, O. Molecular insights into the transcriptional regulatory role of thyroid hormones in ovarian cancer. Mol. Carcinog. 2018, 57, 97–105. [Google Scholar] [CrossRef] [PubMed]
  76. Shinderman-Maman, E.; Cohen, K.; Moskovich, D.; Hercbergs, A.; Werner, H.; Davis, P.J.; Ellis, M.; Ashur-Fabian, O. Thyroid hormones derivatives reduce proliferation and induce cell death and DNA damage in ovarian cancer. Sci. Rep. 2017, 7, 16475. [Google Scholar] [CrossRef]
  77. Yu, Y.; Yao, Y.; Liu, Y.; Sun, Y.; Feng, H.; Kong, N.; Chen, R.; Wu, M.; Guo, S.; Tian, S.; et al. Effects of thyroid hormones on cellular development in human ovarian granulosa tumor cells (KGN). Reprod. Sci. 2024, 32, 1545–1556. [Google Scholar] [CrossRef]
  78. Verga Falzacappa, C.; Mangialardo, C.; Patriarca, V.; Bucci, B.; Amendola, D.; Raffa, S.; Torrisi, M.R.; Silvestrini, G.; Ballanti, P.; Moriggi, G.; et al. Thyroid hormones induce cell proliferation and survival in ovarian granulosa cells COV434. J. Cell. Physiol. 2009, 221, 242–253. [Google Scholar] [CrossRef]
  79. Miro, C.; Nappi, A.; Cicatiello, A.G.; Di Cicco, E.; Sagliocchi, S.; Murolo, M.; Belli, V.; Troiani, T.; Albanese, S.; Amiranda, S.; et al. Thyroid hormone enhances angiogenesis and the Warburg effect in squamous cell carcinomas. Cancers 2021, 13, 2743. [Google Scholar] [CrossRef]
  80. Weingarten, C.; Jenudi, Y.; Tshuva, R.Y.; Moskovich, D.; Alfandari, A.; Hercbergs, A.; Davis, P.J.; Ellis, M.; Ashur-Fabian, O. The interplay between epithelial–mesenchymal transition (EMT) and the thyroid hormones-αvβ3 axis in ovarian cancer. Horm. Cancer 2018, 9, 22–32. [Google Scholar] [CrossRef]
  81. Chin, Y.T.; Wei, P.L.; Ho, Y.; Nana, A.W.; Changou, C.A.; Chen, Y.R.; Yang, Y.S.; Hsieh, M.T.; Hercbergs, A.; Davis, P.J.; et al. Thyroxine inhibits resveratrol-caused apoptosis by PD-L1 in ovarian cancer cells. Endocr. Relat. Cancer 2018, 25, 533–545. [Google Scholar] [CrossRef] [PubMed]
  82. Sabatino, L.; Vassalle, C.; Del Seppia, C.; Iervasi, G. Deiodinases and the three types of thyroid hormone deiodination reactions. Endocrinol. Metab. 2021, 36, 952–964. [Google Scholar] [CrossRef]
  83. Larsen, P.R.; Zavacki, A.M. The role of the iodothyronine deiodinases in the physiology and pathophysiology of thyroid hormone action. Eur. Thyroid. J. 2012, 1, 232–242. [Google Scholar] [CrossRef]
  84. Moskovich, D.; Alfandari, A.; Finkelshtein, Y.; Weisz, A.; Katzav, A.; Kidron, D.; Edelstein, E.; Veroslavski, D.; Perets, R.; Arbib, N.; et al. DIO3, the thyroid hormone inactivating enzyme, promotes tumorigenesis and metabolic reprogramming in high grade serous ovarian cancer. Cancer Lett. 2021, 501, 224–233. [Google Scholar] [CrossRef] [PubMed]
  85. Alfandari, A.; Moskovich, D.; Weisz, A.; Katzav, A.; Kidron, D.; Beiner, M.; Josephy, D.; Asali, A.; Hants, Y.; Yagur, Y.; et al. The selenoenzyme type I iodothyronine deiodinase: A new tumor suppressor in ovarian cancer. Mol. Oncol. 2024, 18, 2298–2313. [Google Scholar] [CrossRef]
  86. Mariani, A.; Wang, C.; Oberg, A.L.; Riska, S.M.; Torres, M.; Kumka, J.; Multinu, F.; Sagar, G.; Roy, D.; Jung, D.B.; et al. Genes associated with bowel metastases in ovarian cancer. Gynecol. Oncol. 2019, 154, 495–504. [Google Scholar] [CrossRef]
  87. Rohdenburg, G.L. Thyroid diabetes. Endocrinology 1920, 4, 63–70. [Google Scholar] [CrossRef]
  88. 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]
  89. Suhane, S.; Ramanujan, V.K. Thyroid hormone differentially modulates Warburg phenotype in breast cancer cells. Biochem. Biophys. Res. Commun. 2011, 414, 73–78. [Google Scholar] [CrossRef]
  90. Czarnecka, A.M.; Matak, D.; Szymanski, L.; Czarnecka, K.H.; Lewicki, S.; Zdanowski, R.; Brzezianska-Lasota, E.; Szczylik, C. Triiodothyronine regulates cell growth and survival in renal cell cancer. Int. J. Oncol. 2016, 49, 1666–1678. [Google Scholar] [CrossRef] [PubMed]
  91. Lin, H.Y.; Sun, M.; Tang, H.Y.; Lin, C.; Luidens, M.K.; Mousa, S.A.; Incerpi, S.; Drusano, G.L.; Davis, F.B.; Davis, P.J. L-Thyroxine vs. 3,5,3’-triiodo-L-thyronine and cell proliferation: Activation of mitogen-activated protein kinase and phosphatidylinositol 3-kinase. Am. J. Physiol. Cell Physiol. 2009, 296, C980–C991. [Google Scholar] [CrossRef]
  92. Ahmed, N.; Stenvers, K.L. Getting to know ovarian cancer ascites: Opportunities for targeted therapy-based translational research. Front. Oncol. 2013, 3, 256. [Google Scholar] [CrossRef]
  93. Chiarugi, P.; Giannoni, E. Anoikis: A necessary death program for anchorage-dependent cells. Biochem. Pharmacol. 2008, 76, 1352–1364. [Google Scholar] [CrossRef]
  94. Gandhi, N.; Das, G.M. Metabolic reprogramming in breast cancer and its therapeutic implications. Cells 2019, 8, 89. [Google Scholar] [CrossRef] [PubMed]
  95. Gogola-Mruk, J.; Marynowicz, W.; Krawczyk, K.; Ptak, A. Visfatin increases the invasive potential of ovarian granulosa tumor spheroids by reprogramming glucose metabolism. Reproduction 2023, 165, 521–531. [Google Scholar] [CrossRef]
  96. Li, Y.; Li, X.; Liu, K.R.; Zhang, J.N.; Liu, Y.; Zhu, Y. Visfatin derived from ascites promotes ovarian cancer cell migration through Rho/ROCK signalling-mediated actin polymerization. Eur. J. Cancer Prev. 2015, 24, 231–239. [Google Scholar] [CrossRef]
  97. Tiwari, A.; Ocon-Grove, O.M.; Hadley, J.A.; Giles, J.R.; Johnson, P.A.; Ramachandran, R. Expression of adiponectin and its receptors is altered in epithelial ovarian tumors and ascites-derived ovarian cancer cell lines. Int. J. Gynecol. Cancer 2015, 25, 399–406. [Google Scholar] [CrossRef] [PubMed]
  98. Ptak, A.; Gregoraszczuk, E.L. Bisphenol A induces leptin receptor expression, creating more binding sites for leptin, and activates the JAK/Stat, MAPK/ERK and PI3K/Akt signalling pathways in human ovarian cancer cell. Toxicol. Lett. 2012, 210, 332–337. [Google Scholar] [CrossRef]
  99. Aydogan, B.I.; Sahin, M. Adipocytokines in thyroid dysfunction. Inflamm 2013, 2013, 646271. [Google Scholar] [CrossRef]
  100. Wang, X.; Sun, J.; Li, J.; Cai, L.; Chen, Q.; Wang, Y.; Yang, Z.; Liu, W.; Lv, H.; Wang, Z. Bidirectional Mendelian randomization study of insulin-related traits and risk of ovarian cancer. Front. Endocrinol. 2023, 14, 1131767. [Google Scholar] [CrossRef]
  101. Kalli, K.R.; Falowo, O.I.; Bale, L.K.; Zschunke, M.A.; Roche, P.C.; Conover, C.A. Functional insulin receptors on human epithelial ovarian carcinoma cells: Implications for IGF-II mitogenic signalling. Endocrinology 2002, 143, 3259–3267. [Google Scholar] [CrossRef] [PubMed]
  102. Joung, K.H.; Jeong, J.W.; Ku, B.J. The association between type 2 diabetes mellitus and women cancer: The epidemiological evidence and putative mechanisms. Biomed. Res. Int. 2015, 2015, 920618. [Google Scholar] [CrossRef]
  103. O’Meara, N.M.; Blackman, J.D.; Sturis, J.; Polonsky, K.S. Alterations in the kinetics of C-peptide and insulin secretion in hyperthyroidism. J. Clin. Endocrinol. Metab. 1993, 76, 79–84. [Google Scholar]
  104. Verga Falzacappa, C.; Mangialardo, C.; Madaro, L.; Ranieri, D.; Lupoi, L.; Stigliano, A.; Torrisi, M.R.; Bouchè, M.; Toscano, V.; Misiti, S. Thyroid hormone T3 counteracts STZ induced diabetes in mouse. PLoS ONE 2011, 6, e19839. [Google Scholar] [CrossRef] [PubMed]
  105. Yang, W.; Jin, C.; Wang, H.; Lai, Y.; Li, J.; Shan, Z. Subclinical hypothyroidism increases insulin resistance in normoglycemic people. Front. Endocrinol. 2023, 14, 1106968. [Google Scholar] [CrossRef]
  106. Dimitriadis, G.; Baker, B.; Marsh, H.; Mandarino, L.; Rizza, R.; Bergman, R.; Haymond, M.; Gerich, J. Effect of thyroid hormone excess on action, secretion, and metabolism of insulin in humans. Am. J. Physiol. 1985, 248, E593–E601. [Google Scholar] [CrossRef] [PubMed]
  107. Brenta, G. Why can insulin resistance be a natural consequence of thyroid dysfunction? J. Thyroid. Res. 2011, 2011, 152850. [Google Scholar] [CrossRef]
  108. Ahrén, B.; Lundquist, I.; Hedner, P.; Valdemarsson, S.; Scherstén, B. Glucose tolerance and insulin and C-peptide responses after various insulin secretory stimuli in hyper and hypothyroid subjects before and after treatment. Diabetes Res. 1985, 2, 95–103. [Google Scholar]
  109. Spira, D.; Buchmann, N.; Dörr, M.; Markus, M.R.P.; Nauck, M.; Schipf, S.; Spranger, J.; Demuth, I.; Steinhagen-Thiessen, E.; Völzke, H.; et al. Association of thyroid function with insulin resistance: Data from two population-based studies. Eur. Thyroid. J. 2022, 11, e210063. [Google Scholar] [CrossRef] [PubMed]
  110. Sathish, R.; Mohan, V. Diabetes and thyroid diseases-a review. Int. J. Diabetes Dev. Ctries. 2003, 23, 120–123. [Google Scholar]
  111. Diaz, E.S.; Karlan, B.Y.; Li, A.J. Obesity-associated adipokines correlate with survival in epithelial ovarian cancer. Gynecol. Oncol. 2013, 129, 353–357. [Google Scholar] [CrossRef]
  112. Yaturu, S.; Prado, S.; Grimes, S.R. Changes in adipocyte hormones leptin, resistin, and adiponectin in thyroid dysfunction. J. Cell. Biochem. 2004, 93, 491–496. [Google Scholar] [CrossRef]
  113. Unal, I.; Khiavi, I.R.; Tasar, G.E.; Goksuluk, D.; Boyraz, G.; Ozgul, N.; Usubutun, A.; Yuruker, S.; Zeybek, N.D. Tumor apelin immunoreactivity is correlated with body mass index in ovarian high grade serous carcinoma. Biotech. Histochem. 2020, 95, 27–36. [Google Scholar] [CrossRef]
  114. Neelakantan, D.; Dogra, S.; Devapatla, B.; Jaiprasart, P.; Mukashyaka, M.C.; Janknecht, R.; Dwivedi, S.K.D.; Bhattacharya, R.; Husain, S.; Ding, K.; et al. Multifunctional APJ Pathway Promotes Ovarian Cancer Progression and Metastasis. Mol. Cancer Res. 2019, 17, 1378–1390. [Google Scholar] [CrossRef] [PubMed]
  115. Hoffmann, M.; Fiedor, E.; Ptak, A. Bisphenol A and its derivatives tetrabromobisphenol A and tetrachlorobisphenol A induce apelin expression and secretion in ovarian cancer cells through a peroxisome proliferator-activated receptor gamma-dependent mechanism. Toxicol. Lett. 2017, 269, 15–22. [Google Scholar] [CrossRef] [PubMed]
  116. Berta, J.; Hoda, M.A.; Laszlo, V.; Rozsas, A.; Garay, T.; Torok, S.; Grusch, M.; Berger, W.; Paku, S.; Renyi-Vamos, F.; et al. Apelin promotes lymphangiogenesis and lymph node metastasis. Oncotarget 2014, 5, 4426–4437. [Google Scholar] [CrossRef] [PubMed]
  117. Dogra, S.; Neelakantan, D.; Patel, M.M.; Griesel, B.; Olson, A.; Woo, S. Adipokine Apelin/APJ Pathway Promotes Peritoneal Dissemination of Ovarian Cancer Cells by Regulating Lipid Metabolism. Mol. Cancer Res. 2021, 19, 1534–1545. [Google Scholar] [CrossRef]
  118. Badavi, M.; Grootveld, M.; Jafari, F.; Dianat, M.; Faraji Shahrivar, F. Supplement therapy with apelin for improving the TSH level and lipid disorders in PTU-induced hypothyroid rats. Biotechnol. Appl. Biochem. 2022, 69, 668–675. [Google Scholar] [CrossRef]
  119. Taheri, S.; Murphy, K.; Cohen, M.; Sujkovic, E.; Kennedy, A.; Dhillo, W.; Dakin, C.; Sajedi, A.; Ghatei, M.; Bloom, S. The effects of centrally administered apelin-13 on food intake, water intake and pituitary hormone release in rats. Biochem. Biophys. Res. Commun. 2002, 291, 1208–1212. [Google Scholar] [CrossRef]
  120. Gürel, A.; Doğantekin, A.; Özkan, Y.; Aydın, S. Serum apelin levels in patients with thyroid dysfunction. Int. J. Clin. Exp. Med. 2015, 8, 16394–16398. [Google Scholar]
  121. Zorlu, M.; Kiskac, M.; Karatoprak, C.; Kesgin, S.; Cakirca, M.; Yildiz, K.; Ardic, C.; Cikrikcioglu, M.A.; Erkoc, R. Assessment of serum apelin and lipocalin-2 levels in patients with subclinical hypothyroidism. Ann. Endocrinol. 2014, 75, 10–14. [Google Scholar] [CrossRef]
  122. Wittamer, V.; Franssen, J.D.; Vulcano, M.; Mirjolet, J.F.; Le Poul, E.; Migeotte, I.; Brézillon, S.; Tyldesley, R.; Blanpain, C.; Detheux, M.; et al. Specific recruitment of antigen-presenting cells by chemerin, a novel processed ligand from human inflammatory fluids. J. Exp. Med. 2003, 198, 977–985. [Google Scholar] [CrossRef]
  123. Gao, C.; Shi, J.; Zhang, J.; Li, Y.; Zhang, Y. Chemerin promotes proliferation and migration of ovarian cancer cells by upregulating expression of PD-L1. J. Zhejiang Univ. Sci. B 2022, 23, 164–170. [Google Scholar] [CrossRef]
  124. Schmitt, M.; Gallistl, J.; Schüler-Toprak, S.; Fritsch, J.; Buechler, C.; Ortmann, O.; Treeck, O. Anti-Tumoral Effect of Chemerin on Ovarian Cancer Cell Lines Mediated by Activation of Interferon Alpha Response. Cancers 2022, 14, 4108. [Google Scholar] [CrossRef] [PubMed]
  125. Treeck, O.; Buechler, C.; Ortmann, O. Chemerin and Cancer. Int. J. Mol. Sci. 2019, 20, 3750. [Google Scholar] [CrossRef] [PubMed]
  126. Hoffmann, M.; Rak, A.; Ptak, A. Bisphenol A and its derivatives decrease expression of chemerin, which reverses its stimulatory action in ovarian cancer cells. Toxicol. Lett. 2018, 291, 61–69. [Google Scholar] [CrossRef]
  127. Alshaikh, E.M.; Omar, U.M.; Alsufiani, H.M.; Mansouri, R.A.; Tarbiah, N.I.; Alshaikh, A.A.; Rahimulddin, S.A.; Doghaither, H.A. The potential influence of hyperthyroidism on circulating adipokines chemerin, visfatin, and omentin. Int. J. Health Sci. 2019, 13, 44–47. [Google Scholar]
  128. Pang, L.; Chang, X. Resistin Expression in Epithelial Ovarian Cancer promotes the Proliferation and Migration of Ovarian Cancer Cells to Worsen Prognosis. J. Cancer 2021, 12, 6796–6804. [Google Scholar] [CrossRef]
  129. Zhou, L.; Song, K.; Luo, W. Association between circulating resistin levels and thyroid dysfunction: A systematic review and meta-analysis. Front. Endocrinol. 2023, 13, 1071922. [Google Scholar] [CrossRef]
  130. Iglesias, P.; Alvarez Fidalgo, P.; Codoceo, R.; Díez, J.J. Serum concentrations of adipocytokines in patients with hyperthyroidism and hypothyroidism before and after control of thyroid function. Clin. Endocrinol. 2003, 59, 621–629. [Google Scholar] [CrossRef] [PubMed]
  131. Eke Koyuncu, C.; Turkmen Yildirmak, S.; Temizel, M.; Ozpacaci, T.; Gunel, P.; Cakmak, M.; Ozbanazi, Y.G. Serum resistin and insulin-like growth factor-1 levels in patients with hypothyroidism and hyperthyroidism. J. Thyroid. Res. 2013, 2013, 306750. [Google Scholar] [CrossRef]
  132. Owecki, M.; El Ali, Z.; Waśko, R.; Nikisch, E.; Sowiński, J. Circulating resistin levels are lower in hypothyroid women but independent from thyroid hormones concentrations. Neuroendocr. Lett. 2008, 29, 131–136. [Google Scholar]
  133. Peng, Y.; Qi, Y.; Huang, F.; Chen, X.; Zhou, Y.; Ye, L.; Wang, W.; Ning, G.; Wang, S. Downregulated resistin level in consequence of decreased neutrophil counts in untreated Grave’s disease. Oncotarget 2016, 7, 78680–78687. [Google Scholar] [CrossRef]
  134. Shackelford, R.E.; Bui, M.M.; Coppola, D.; Hakam, A. Over-expression of nicotinamide phosphoribosyltransferase in ovarian cancers. Int. J. Clin. Exp. Pathol. 2010, 3, 522–527. [Google Scholar] [PubMed]
  135. Li, D.; Chen, N.N.; Cao, J.M.; Sun, W.P.; Zhou, Y.M.; Li, C.Y.; Wang, X.X. BRCA1 as a nicotinamide adenine dinucleotide (NAD)-dependent metabolic switch in ovarian cancer. Cell Cycle 2014, 13, 2564–2571. [Google Scholar] [CrossRef]
  136. Ozkaya, M.; Sahin, M.; Cakal, E.; Yuzbasioglu, F.; Sezer, K.; Kilinc, M.; Imrek, S.S. Visfatin plasma concentrations in patients with hyperthyroidism and hypothyroidism before and after control of thyroid function. J. Endocrinol. Investig. 2009, 32, 435–439. [Google Scholar] [CrossRef] [PubMed]
  137. González, C.R.; Caminos, J.E.; Vázquez, M.J.; Garcés, M.F.; Cepeda, L.A.; Angel, A.; González, A.C.; García-Rendueles, M.E.; Sangiao-Alvarellos, S.; López, M.; et al. Regulation of visceral adipose tissue-derived serine protease inhibitor by nutritional status, metformin, gender and pituitary factors in rat white adipose tissue. J. Physiol. 2009, 587, 3741–3750. [Google Scholar] [CrossRef]
  138. Handisurya, A.; Riedl, M.; Vila, G.; Maier, C.; Clodi, M.; Prikoszovich, T.; Ludvik, B.; Prager, G.; Luger, A.; Kautzky-Willer, A. Serum vaspin concentrations in relation to insulin sensitivity following RYGB-induced weight loss. Obes. Surg. 2010, 20, 198–203. [Google Scholar] [CrossRef] [PubMed]
  139. Cinar, N.; Gülçelik, N.E.; Aydín, K.; Akın, S.; Usman, A.; Gürlek, A. Serum vaspin levels in hypothyroid patients. Eur. J. Endocrinol. 2011, 165, 563–569. [Google Scholar] [CrossRef]
  140. Au-Yeung, C.L.; Yeung, T.L.; Achreja, A.; Zhao, H.; Yip, K.P.; Kwan, S.Y.; Onstad, M.; Sheng, J.; Zhu, Y.; Baluya, D.L.; et al. ITLN1 modulates invasive potential and metabolic reprogramming of ovarian cancer cells in omental microenvironment. Nat. Commun. 2020, 11, 3546. [Google Scholar] [CrossRef]
  141. Cerit, E.T.; Akturk, M.; Altinova, A.E.; Tavil, Y.; Ozkan, C.; Yayla, C.; Altay, M.; Demirtas, C.; Cakir, N. Evaluation of body composition changes, epicardial adipose tissue, and serum omentin-1 levels in overt hypothyroidism. Endocrine 2015, 49, 196–203. [Google Scholar] [CrossRef]
  142. Patil, N.; Rehman, A.; Anastasopoulou, C.; Jialal, I.; Saathoff, A.D. Hypothyroidism (Nursing); StatPearls: Treasure Island, FL, USA, 2024. [Google Scholar]
  143. Tomer, Y. Genetic susceptibility to autoimmune thyroid disease: Past, present, and future. Thyroid 2010, 20, 715–725. [Google Scholar] [CrossRef]
  144. Muderris, I.I.; Boztosun, A.; Oner, G.; Bayram, F. Effect of thyroid hormone replacement therapy on ovarian volume and androgen hormones in patients with untreated primary hypothyroidism. Ann. Saudi. Med. 2011, 31, 145–151. [Google Scholar] [CrossRef]
  145. Markman, T.M.; Markman, M.; Clark, B.W. Case of Ovarian Cancer in a Woman with Undiagnosed Graves’ Disease: A Case Report and Review of the Literature. Case Rep. Oncol. 2017, 10, 452–454. [Google Scholar] [CrossRef] [PubMed]
  146. Skarf, L.M.; Dezube, B.J.; Bryan, B.; Berkenblit, A. Ovarian carcinoma with thyroid metastases causing clinical hypothyroidism: A case report. Gynecol. Oncol. 2006, 102, 394–396. [Google Scholar] [CrossRef]
  147. Kimura, R.; Imaeda, K.; Mizuno, T.; Wakami, K.; Yamada, K.; Okayama, N.; Kamiya, Y.; Joh, T. Severe ascites with hypothyroidism and elevated CA125 concentration: A case report. Endocr. J. 2007, 54, 751–755. [Google Scholar] [CrossRef] [PubMed]
  148. Tan, A.; Stewart, C.J.; Garrett, K.L.; Rye, M.; Cohen, P.A. Novel BRAF and KRAS Mutations in Papillary Thyroid Carcinoma Arising in Struma Ovarii. Endocr. Pathol. 2015, 26, 296–301. [Google Scholar] [CrossRef] [PubMed]
  149. Grandet, P.J.; Remi, M.H. Struma ovarii with hyperthyroidism. Clin. Nucl. Med. 2000, 25, 763–765. [Google Scholar] [CrossRef]
  150. Mesquita, J.B.; Biscolla, R.P.M. Hyperthyroidism in thyroid carcinoma originating in struma ovarii. Endocrinol. Diabetes Metab. Case Rep. 2024, 2024, 24-0082. [Google Scholar] [CrossRef]
  151. Lebreton, C.; Al Ghuzlan, A.; Floquet, A.; Kind, M.; Leboulleux, S.; Godbert, Y. Cancer thyroïdien sur struma ovarii: Généralités et principes de prise en charge [Thyroid carcinoma on struma ovarii: Diagnosis and treatment]. Bull. Cancer 2018, 105, 281–289. [Google Scholar] [CrossRef]
  152. Koehler, V.F.; Keller, P.; Waldmann, E.; Schwenk, N.; Kitzberger, C.; Schmohl, K.A.; Knösel, T.; Stief, C.G.; Spitzweg, C. An unusual case of struma ovarii. Endocrinol. Diabetes Metab. Case Rep. 2021, 20-0142. [Google Scholar] [CrossRef]
  153. Carvalho, J.P.; Carvalho, F.M.; Lima de Oliveira, F.F.; Asato de Camargo, R.Y. Hypothyroidism following struma ovarii tumor resection: A case report. Rev. Hosp. Clin. 2002, 57, 112–114. [Google Scholar] [CrossRef]
Figure 1. Direct and indirect effects of thyroid dysfunction (hyperthyroidism and hypothyroidism) via alterations in thyroid hormone levels (T4 and T3) on ovarian cancer (OC). T4—thyroxine; T3—triiodothyronine; TRα—thyroid hormone receptor α; TRβ—thyroid hormone receptor β.
Figure 1. Direct and indirect effects of thyroid dysfunction (hyperthyroidism and hypothyroidism) via alterations in thyroid hormone levels (T4 and T3) on ovarian cancer (OC). T4—thyroxine; T3—triiodothyronine; TRα—thyroid hormone receptor α; TRβ—thyroid hormone receptor β.
Biomolecules 15 00870 g001
Figure 2. Flow chart review procedure.
Figure 2. Flow chart review procedure.
Biomolecules 15 00870 g002
Figure 3. Classification of ovarian cancer (OC). Epithelial ovarian cancer (EOC) is divided into type I, type II and transitional Brenner tumors. Type I EOC includes low-grade serous carcinoma (LGSC), low-grade endometrioid carcinoma (LGEC), mucinous carcinoma (MC), and clear cell carcinoma (CCC). Type II EOC includes high-grade serous carcinoma (HGSC), carcinosarcoma, and undifferentiated carcinoma. Nonepithelial OCs include germ cell tumors (GeCTs), adult granulosa cell tumors (AGCTs), juvenile granulosa cell tumors (JGCTs), sex cord-stromal tumors (SCSTs) and rare tumors (small cell carcinomas).
Figure 3. Classification of ovarian cancer (OC). Epithelial ovarian cancer (EOC) is divided into type I, type II and transitional Brenner tumors. Type I EOC includes low-grade serous carcinoma (LGSC), low-grade endometrioid carcinoma (LGEC), mucinous carcinoma (MC), and clear cell carcinoma (CCC). Type II EOC includes high-grade serous carcinoma (HGSC), carcinosarcoma, and undifferentiated carcinoma. Nonepithelial OCs include germ cell tumors (GeCTs), adult granulosa cell tumors (AGCTs), juvenile granulosa cell tumors (JGCTs), sex cord-stromal tumors (SCSTs) and rare tumors (small cell carcinomas).
Biomolecules 15 00870 g003
Figure 4. The regulation of thyroid hormone (TH) synthesis by the hypothalamus–pituitary–thyroid (HPT) axis and TH genomic and nongenomic action. TRH secreted by the hypothalamus stimulates the secretion of TSH by the pituitary. TSH stimulates the thyroid to synthesize and secrete T4 and T3 into the blood, which can enter target cells via transmembrane transporters. Inside the target cells, DIO2 converts T4 into T3. The classical genomic action of TH is mediated by binding within target cells to the nuclear TR subtypes TRα and TRβ. The nongenomic action of TH is associated with its action as a membrane thyroid hormone receptor integrin alpha V beta 3 (αvβ3) ligand, which activates specific signaling systems (trafficking and activation of mitogen-activated protein kinase (MAPK)). TH, thyroid hormone; TSH, thyroid-stimulating hormone; T4, thyroxine; T3, 3,3′,5′-triiodothyronine; HPT axis, hypothalamic–pituitary–thyroid axis; TRH, thyrotropin-releasing hormone; DIO, iodothyronine deiodinase; TRs, thyroid hormone receptors; integrin αvβ3 (integrin alpha V beta 3); MAPK, mitogen-activated protein kinase.
Figure 4. The regulation of thyroid hormone (TH) synthesis by the hypothalamus–pituitary–thyroid (HPT) axis and TH genomic and nongenomic action. TRH secreted by the hypothalamus stimulates the secretion of TSH by the pituitary. TSH stimulates the thyroid to synthesize and secrete T4 and T3 into the blood, which can enter target cells via transmembrane transporters. Inside the target cells, DIO2 converts T4 into T3. The classical genomic action of TH is mediated by binding within target cells to the nuclear TR subtypes TRα and TRβ. The nongenomic action of TH is associated with its action as a membrane thyroid hormone receptor integrin alpha V beta 3 (αvβ3) ligand, which activates specific signaling systems (trafficking and activation of mitogen-activated protein kinase (MAPK)). TH, thyroid hormone; TSH, thyroid-stimulating hormone; T4, thyroxine; T3, 3,3′,5′-triiodothyronine; HPT axis, hypothalamic–pituitary–thyroid axis; TRH, thyrotropin-releasing hormone; DIO, iodothyronine deiodinase; TRs, thyroid hormone receptors; integrin αvβ3 (integrin alpha V beta 3); MAPK, mitogen-activated protein kinase.
Biomolecules 15 00870 g004
Figure 5. THRA and THRB expression in ovarian cancer (OC). The graphs of THRA (A) and THRB (B) mRNA expression generated from GEPIA ovarian tumors (T; in red; n = 426) compared with noncancer ovarian tissues (N; in gray; n =88). * p < 0.05.
Figure 5. THRA and THRB expression in ovarian cancer (OC). The graphs of THRA (A) and THRB (B) mRNA expression generated from GEPIA ovarian tumors (T; in red; n = 426) compared with noncancer ovarian tissues (N; in gray; n =88). * p < 0.05.
Biomolecules 15 00870 g005
Figure 6. Thyroid receptor (THRA and THRB) expression levels and survival in ovarian cancer (OC) patients. Kaplan–Meier survival plots of patients with OC. Overall survival (OS) and progression-free survival (PFS) associated with the THRA expression level (A,B) and THRB expression level (C,D) are shown. The probe IDs are as follows: THRA (1316_at, 204100_at, 204760_s_at, 214883_at, 217476_at, 31637_s_at 35846_at) and THRB (207044_at, 228716_at, 229657_at). The hazard ratio (HR) is indicated, along with the 95% confidence interval (CI). The log-rank P test is displayed at the upper right corner of every graph [11].
Figure 6. Thyroid receptor (THRA and THRB) expression levels and survival in ovarian cancer (OC) patients. Kaplan–Meier survival plots of patients with OC. Overall survival (OS) and progression-free survival (PFS) associated with the THRA expression level (A,B) and THRB expression level (C,D) are shown. The probe IDs are as follows: THRA (1316_at, 204100_at, 204760_s_at, 214883_at, 217476_at, 31637_s_at 35846_at) and THRB (207044_at, 228716_at, 229657_at). The hazard ratio (HR) is indicated, along with the 95% confidence interval (CI). The log-rank P test is displayed at the upper right corner of every graph [11].
Biomolecules 15 00870 g006
Figure 7. Expression of two subunits of integrin αvβ3 in ovarian cancer (OC). Graphs of ITGAV (A) and ITGB3 (B) mRNA expression in GEPIA-identified ovarian tumors (T; red, n = 426) compared with normal tissues (N; gray, n =88).
Figure 7. Expression of two subunits of integrin αvβ3 in ovarian cancer (OC). Graphs of ITGAV (A) and ITGB3 (B) mRNA expression in GEPIA-identified ovarian tumors (T; red, n = 426) compared with normal tissues (N; gray, n =88).
Biomolecules 15 00870 g007
Figure 8. Integrin αvβ3 (subunits: ITGAV and ITGB3) expression levels and survival in ovarian cancer (OC) patients. Kaplan–Meier survival plots of patients with OC. (A) Overall survival (OS) and (B) progression-free survival (PFS) are shown; the probe IDs are as follows: ITGAV (202351_at) and ITGB3 (204625_s_at, 204626_s_at, 204628_s_at, 215240_at, 216261_at, 204627_s_at, 211579_at). The hazard ratio (HR) is indicated, along with the 95% confidence interval (CI). The log-rank P test is displayed at the upper right corner of every graph [11].
Figure 8. Integrin αvβ3 (subunits: ITGAV and ITGB3) expression levels and survival in ovarian cancer (OC) patients. Kaplan–Meier survival plots of patients with OC. (A) Overall survival (OS) and (B) progression-free survival (PFS) are shown; the probe IDs are as follows: ITGAV (202351_at) and ITGB3 (204625_s_at, 204626_s_at, 204628_s_at, 215240_at, 216261_at, 204627_s_at, 211579_at). The hazard ratio (HR) is indicated, along with the 95% confidence interval (CI). The log-rank P test is displayed at the upper right corner of every graph [11].
Biomolecules 15 00870 g008
Table 1. The effects of hyperthyroidism and hypothyroidism on OC risk and mortality.
Table 1. The effects of hyperthyroidism and hypothyroidism on OC risk and mortality.
Thyroid
Dysfunction
Incidence and MortalityHR and ClNumber of
OC Patients
Authors
HYPERTHYROIDISM
almost twice the risk of OC in women aged 20–69 years
HR:1,8; (1.0–3.0)
95% CI
767 and 1367 controls[3]
positive correlation with incidence of OC in women aged 29–50 yearsNd.12[37]
risk of OC incidence in women relative to controls even after 5 years of follow-up
HR: 1.31; (1.13–1.52) 95% CI404 and 578 controls[38]
risk of OC positively correlates with genetic predisposition to hyperthyroidism
mortality in patients with OC
HR: 1.094; (1.029–1.164) 95% CI1218 and 198,523 controls
several
[4]
OC mortality in women treated with radioactive iodine compared to women with a healthy thyroid in the control group
risk of death from OC
HR: 5.33;
(2.17–13.08)
95% Cl
HR: 1.65; (0.81–3.37) 95% Cl
5 and
143 controls
several
[5]
risk ratio for OC mortality
HR: 2.14;
95% Cl;
1501 and 69,119 control[40]
mortality associated with diagnosis of hyperthyroidism 5 years prior to OC diagnosis positive correlation with worse 5-year OC survival
association with overall OC mortality at 5 years of follow-up
HR: 1.94; (1.19 –3.18) 95% CI160[39]
risk of developing OC
HR: 0.67; (0.30–1.49)
95% CI
6 and 1052 controls[41]
risk of developing gynecological cancers, including OC
Nd44 852[42]
HYPOTHYROIDISM
slightly risk of developing OC
Nd296 872[42]
medium association with OC mortalityworse 5-year OC survivalHR: 1.16; (1.03–1.32) 95% CIseveral
624
[39]
risk of developing OC
HR: 0.84; (0.68–1.04) 95% CI92 and 1052 controls[41]
OC incidence
Nd1218 and 198,523 controls[4]
risk of death from OC
HR: 1.35;95% CI4456 and 69,119 control[40]
OC—Ovarian cancer, ↑—significant increase, ↓— significant decrease, —no association, HR—hazard risk, CI—confidence interval, Nd—no data.
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

Gogola-Mruk, J.; Sirek, A.; Kumor, I.; Wojtaszek, G.; Roszak, K.; Kulig, K.; Ptak, A. Relationship Between Thyroid Dysfunction and Ovarian Cancer. Biomolecules 2025, 15, 870. https://doi.org/10.3390/biom15060870

AMA Style

Gogola-Mruk J, Sirek A, Kumor I, Wojtaszek G, Roszak K, Kulig K, Ptak A. Relationship Between Thyroid Dysfunction and Ovarian Cancer. Biomolecules. 2025; 15(6):870. https://doi.org/10.3390/biom15060870

Chicago/Turabian Style

Gogola-Mruk, Justyna, Aleksandra Sirek, Izabela Kumor, Gabriela Wojtaszek, Klaudia Roszak, Karolina Kulig, and Anna Ptak. 2025. "Relationship Between Thyroid Dysfunction and Ovarian Cancer" Biomolecules 15, no. 6: 870. https://doi.org/10.3390/biom15060870

APA Style

Gogola-Mruk, J., Sirek, A., Kumor, I., Wojtaszek, G., Roszak, K., Kulig, K., & Ptak, A. (2025). Relationship Between Thyroid Dysfunction and Ovarian Cancer. Biomolecules, 15(6), 870. https://doi.org/10.3390/biom15060870

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