Next Article in Journal
Esophageal Lichen Planus—Contemporary Insights and Emerging Trends
Previous Article in Journal
Investigation of Serum Visfatin and Chemerin Levels in Type 2 Diabetes and Obesity Patients: Their Potential Role as Clinical and Biomarkers
Previous Article in Special Issue
Early Non-Response to Neoadjuvant Chemotherapy Will Increase the Recurrence of Cervical Cancer: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Estrogen Receptors as Key Factors in Carcinogenesis

Department of Biotechnology and Genetic Engineering, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
*
Authors to whom correspondence should be addressed.
Biomedicines 2025, 13(11), 2620; https://doi.org/10.3390/biomedicines13112620 (registering DOI)
Submission received: 26 September 2025 / Revised: 17 October 2025 / Accepted: 24 October 2025 / Published: 26 October 2025
(This article belongs to the Special Issue Current Perspectives on Gynecologic Cancers)

Abstract

Despite continuous advances in the development of methodologies for the diagnosis and therapeutic treatment of cancer, the disease remains a primary cause of mortality worldwide. A comprehensive understanding of the molecular mechanisms underlying cancer could ultimately lead to increasingly effective therapeutic interventions. One approach that could be adopted is to formulate methodologies that impede cell signalling and/or the expression of genes pivotal to carcinogenesis. A notable example of this strategy is the focus on the estrogen receptor, a key player in the development of various types of cancer. The deregulation of this receptor, and the subsequent impact on cell function, is a critical factor in the progression of these diseases. This renders it a significant therapeutic target. Furthermore, the microenvironment has been demonstrated to exert a significant influence on the development of cancers. A mounting body of evidence indicates that the abnormal physical properties of the tumour microenvironment can induce widespread changes, leading to the selection of characteristic tumour cell abilities and subsequent clonal proliferation. This process is accompanied by an increased capacity for invasive growth and, notably, the induction of multidrug resistance. The present article focuses on presenting the structure and role of the estrogen receptor in selected hormone-dependent cancers, its involvement in the formation of the tumor microenvironment, currently used therapeutic methods in the treatment of these cancers, and the challenges associated with them. Each new discovery in the field of cancer biology offers the prospect of developing new potential treatments, including targeted therapies aimed at improving the survival of patients suffering from hormone-dependent malignant tumours. Although the role of the estrogen receptor in their development is well established, further research is required to develop a detailed understanding of how its specific isoforms act in different types of cancer.

1. Introduction

On a global scale, cancer is considered to be one of the most significant health and economic challenges of the 21st century. Research has shown that cancer accounts for around one-third of premature deaths [1]. Prospective analyses indicate that, by 2040, there will be nearly 40 million new cancer cases, with mortality exceeding 15 million [2]. This will have a profound impact on millions of individuals worldwide, contributing significantly to cancer-related morbidity and mortality. A detailed analysis of cancer cases has revealed significant gender disparities in the incidence of certain types of cancer. Research has shown that this is due to hormones secreted by the gonads [3].
Human hormones are specialised chemical compounds that are produced by endocrine cells with the specific function of regulating the activity of various types of cells. These hormones are responsible for regulating numerous physiological functions within the human body. The levels of these hormones fluctuate depending on the developmental stage of the body. The balance of these elements is crucial for the optimal functioning of the body as a whole [4]. Disorders in homeostatic mechanisms can lead to adverse health consequences, manifesting as metabolic diseases, including obesity, diabetes, cardiovascular disease and cancer [5]. The role of these cells in the process of carcinogenesis is primarily to stimulate specific receptors. The activation of these receptors leads to a cascade of cellular events, the end result of which is the regulation of gene expression. The process of carcinogenesis is initiated by the activation of genes responsible for promoting cell growth and proliferation and/or inhibiting apoptosis and stimulating angiogenesis [6]. Sex hormones have been identified as playing a pivotal role in some cancers’ development [7]. These cancers are classified into a separate group referred to as hormone-dependent cancers [8]. Evidently, both androgen and estrogen hormones play a pivotal role in the initiation, development and progression of hormone-dependent cancers. This may be due to increased hormone concentrations, receptor overexpression, or constitutive stimulation caused by mutation [9]. The important role of estrogen receptors (ERs) in physiological and biological processes such as growth, development and metabolism has been demonstrated [10]. Stimulating ERs has been shown to activate transcriptional processes and signalling events, resulting in the regulation of gene expression [11,12]. This aspect of ER function is also associated with their involvement in pathological processes [12]. There is a demonstrated correlation between ER activity and various types of cancer, including prostate, breast, ovarian, endometrial and lung cancer [10]. In addition to the well-established classical molecular pathways, the estrogen receptor has also been shown to interact with the tumor microenvironment, driving carcinogenesis further [13]. Furthermore, numerous reports have emerged regarding their involvement in neurodegenerative diseases, coronary diseases, osteoporosis [10], gastrointestinal diseases and even melanoma [14].
Several forms of estrogen receptors have been described. Among these are the estrogen receptor alpha and beta, which have numerous isoforms, and the estrogen receptor coupled with G protein. The estrogen receptor alpha (ERα) was first described in the 1950s. For several years, it was considered the only receptor protein, until 1996, when the estrogen receptor beta (ERβ) was discovered, exhibiting similar properties. The most recent discovery was the G protein-coupled estrogen receptor class A (GPER) [15].
In the context of hormone-dependent cancers, estrogen receptors play a particularly important role in the tumour microenvironment (TME). The components of the tumour interact with each other to influence the development and progression of cancer. It is evident that both cancer cells and the surrounding normal cells, including immune system cells, participate in the formation of the TME. Another significant component of the TME is the extracellular matrix (ECM), which consists of active tissue components that facilitate intercellular communication, proliferation and adhesion [16]. The expression of estrogen receptors has been observed in the cells that constitute the TME. These cells influence the TME by activating pathways that stimulate microenvironment components [17].
Understanding the mechanisms by which the estrogen receptor is activated in pathways involved in cancer development has rendered ER a very promising therapeutic target [18]. Consequently, numerous therapeutic interventions have been developed for cancers that express ER. Two examples of anti-estrogens that have been developed are selective estrogen receptor modulators (SERMs) and selective estrogen receptor downregulators (SERDs). These are designed to compete with estrogen for binding to ER. The mechanism of action of selective estrogen receptor modulators involves a change in the configuration of the receptor, thereby preventing the formation of an active complex. Conversely, SERD activity ultimately results in ER degradation. In addition, there are other groups of drugs that have been demonstrated to affect gene activation by ER. For instance, aromatase inhibitors (AIs) implicated in estradiol synthesis impose constraints on the production of the ligand indispensable for ER activation, consequently engendering a diminution in its activity [19]. However, it has been demonstrated that the activity of some of these genes can be negated or reduced by a mutation in the gene encoding ER [20]. Furthermore, it has been hypothesised that certain components of the TME, notably immune cells, may facilitate the development of drug resistance in cancer cells [21]. These limitations, primarily associated with SERMs and AIs, may be surmounted by the emergence of SERD-based therapies [22]. Therapies based on the use of cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) have been developed, which are insensitive to resistance associated with mutations in the ER gene and result in prolonged progression-free survival in patients with advanced breast cancer [23].

2. Structure and Signaling of the Estrogen Receptor

It is evident that estrogen receptors alpha and beta are classified as members of the superfamily of nuclear receptors. The analysis revealed that they exhibit common structural features in several regions. However, their functional domains demonstrate significant differences. ERα consists of 595 amino acids and has a molecular weight of 67 kDa. Conversely, ERβ comprises 530 amino acids and has a molecular weight of 59 kD (Figure 1). This variation between the two proteins is due to the shorter amino domain present in the beta receptor. In humans, the alpha receptor is encoded by the ESR1 (estrogen receptor 1) gene. Its location on chromosome 6 is at locus 6q25.1 [18,24].
The letters A/B, C, D and E/F are assigned to the basic functional domains. Region A/B corresponds to the N-terminal nucleotide binding domain (NTD), which is responsible for transactivation with the gene transcription mechanism. This fragment has been found to contain zinc fingers, which are known to mediate binding to target sequences. The subsequent region, marked with the letter C, is responsible for the DNA-binding domain (DBD). It contributes to the process of dimerisation of the estrogen receptor and to the process of binding to specific sequences in chromosomes. Domain D has been identified as a hinge region, functioning as a flexible link between domains C and E. It has been demonstrated to bind to chaperone proteins and contains a nuclear localisation signal that is exposed following estrogen binding. Consequently, the receptor-ligand complex is able to enter the cell nucleus. The carboxylic terminus of the protein, designated as region E/F, has been identified as the ligand-binding domain (LBD). This region has been found to contain the estrogen binding site, in addition to binding sites for coactivators and corepressors. AF1 and AF2, located in the NTD and DBD, respectively, are two additional transcriptional activity regulators known as activation function (AF) domains [14,18,25,26].
In addition to the full-length (67 kDa) form of ERα, several isoforms have been identified that differ in length [18]. Exemplary isoforms in this category include ERα46 and ERα36, whose presence is associated with the development and progression of cancers such as breast and thyroid cancer. Some of these genes are unable to activate transcription because they lack the NTD region, and thus the functional AF-1 domain. However, the observed binding of these molecules to the full-length ERα receptor results in the formation of heterodimers, thereby inhibiting the ability of the receptor to regulate transcription. In turn, the smallest variant, ERα-36, does not contain transcription-activating domains (activation function 1, AF1, and activation function 2, AF2) in its structure [24].
The estrogen receptor beta is encoded by the ESR2 (estrogen receptor 2) gene, which is located on chromosome 14 at locus 14q23-24. The protein is classified as a nuclear receptor and is predominantly located in the cell nucleus; however, evidence suggests that it is also present in the cytoplasm and mitochondria. To date, no link has been established between cellular location and ERβ function [18]. The beta estrogen receptor occurs in five different variants. These are distinguished as ERβ1 to ERβ5, and are the result of alternative splicing. However, it is only full-length beta receptors that are capable of binding antagonistic and agonistic ligands. Furthermore, shorter forms of ERβ are unable to form homodimers and, in addition, have no intrinsic activity of their own. Conversely, they have the potential to heterodimerise with ERβ and even with ERα, leading to the induction of transcriptional activity in a ligand-dependent manner. This is particularly important in the study of the interaction and mutual regulation of related receptors [27].
A recently identified atypical estrogen receptor has been found to be coupled with a class A G protein. This G protein-coupled estrogen receptor has been identified as a membrane estrogen receptor that is not associated with the nuclear ER. It is located in the cytoplasmic membrane and has the ability to move to the endoplasmic reticulum membrane. The gene that encodes GPER is located on chromosome 7 (locus 7p22.3) [28]. Its structural configuration deviates from the two fundamental types of ERs. This receptor’s structure is typical of a G protein-coupled receptor, consisting of seven transmembrane α-helical regions, four extracellular segments, and four cytosolic segments [29]. It is evident that, owing to its divergent structural characteristics, GPER functions via an alternative mechanism to conventional nuclear receptors, such as ER α and β. The role of GPER as an estrogen mediator has been demonstrated in various physiological systems, including the nervous, immune, and cardiovascular systems. Furthermore, it has been shown to play a pivotal role in the metabolic processes of bones, kidneys and the pancreas [28,30], The structure of this receptor results in a comparatively low level of affinity for estradiol binding in comparison to other hormone receptors. However, this becomes an advantage for GPER, as it functions as a secondary messenger in rapid responses to estrogen and the activation of intracellular signalling cascades [18].
ER signalling can be initiated via either genomic or non-genomic pathways. Genomic signalling can be categorised into two distinct types: direct and indirect. The study is based on the premise that ligand-activated ER impacts DNA. In the case of direct genomic signalling, the dimerised ERα or ERβ complex is transported from the cytoplasm to the cell nucleus, where it interacts directly with the estrogen response elements sequence (EREs) within gene promoters, thereby initiating their expression. In contrast, indirect genomic signalling relies on interactions between secondary signal transducers and gene expression, which are initiated by ER activation via a ligand. The non-genomic pathway, in turn, involves the influence of activated ER on intracellular signalling pathways that initiate gene expression [18].
Activation of estrogen receptors has been shown to result in the expression of genes that regulate processes such as proliferation and cell differentiation, apoptosis and autophagy, and vasodilation (Figure 2). Consequently, imbalances in the quantity and activity of these receptors have been linked to numerous types of cancer [14].

3. Role of TME

A comprehensive understanding of the intricate interactions between the components of the tumour microenvironment is imperative for the development of future effective cancer therapies. To this end, the following presentation will summarise the latest reports on TME in hormone-dependent cancers.
Cancer-associated fibroblasts (CAFs) appear to play a key role in the tumour microenvironment. These cells are classified into multiple subpopulations, which have been observed to be associated with an increased risk of tumour development through various mechanisms. To illustrate this point, in the context of colorectal cancer (CRC), the presence of cancer-associated fibroblasts that express the melanoma cell adhesion molecule (MCAM) has been shown to correlate with a more aggressive clinical course and a poorer prognosis for patients [31]. Despite the presence of differences in subpopulations, the majority of these cells produce interleukin 6 (IL-6), a key factor in the development of immune resistance in breast cancer (BC) patients [32] and the promotion of thyroid cancer (TC) cell proliferation [33]. Its inhibition has been demonstrated to result in a weakening of interactions with the STAT3/Akt-PD-L1 pathway, thereby reversing this implication [32]. In a similar manner, CAFs have been observed to exert an influence on other TME cells. Research conducted using a mouse BC model has demonstrated elevated levels of interleukin-2 (IL-2) and interleukin-7 (IL-7), along with cytotoxic T cells and dendritic cells exhibiting antitumour activity, subsequent to the elimination of CAFs. Furthermore, a reduction in tumour-associated macrophages (TAMs), regulatory T lymphocytes (Tregs), vascular endothelial growth factor (VEGF), interleukin-4 (IL-4) and interleukin-6 (IL-6), which have immunosuppressive effects, was observed in BC [34]. Zhang et al. [35] conducted an analysis that identified seven subtypes of TAMs in primary BC samples. A significant proportion of these exhibited pro-tumour characteristics, including influencing angiogenesis via endothelial cells, maintaining inflammation, regulating Tregs and inhibiting T cell function. However, the remaining subtypes exhibited a dual or suppressive effect on BC development [35]. The recruitment of TAMs by CAFs has been linked to the promotion of programmed cell death protein-1 (PD-1) expression [36]. Conversely, the impact of TAMs on the suppression of T cell function via their influence on the programmed cell death ligand 1 (PD-L1) pathway in ovarian cancer (OC) signifies the multifaceted nature of these interactions [37]. Studies of OC have shown that TAMs are involved in forming heterotypic spheroids with high ITGA5 (integrin α5) expression in metastasis [38]. Regarding TC, studies have shown that CAFs are the only risk factor for lymph node metastasis [39]. Research conducted on TC has demonstrated that transforming growth factor beta 1 (TGFβ1), a multifaceted cytokine secreted by tumour cells, directly contributes to the influx of TAMs [40]. Additionally, a correlation has been identified between reduced autophagy in BC and resistance to tamoxifen treatment [41].
The extant research indicates that TAMs contribute to tamoxifen resistance through their activation of the PI3K/Akt/mTOR pathway [42,43]. A study conducted by Qin et al. indicates a mechanism mediated by increased cyclooxygenase-2 (COX-2) expression in TAMs [42], while Li et al. attribute this relationship to CC-chemokine ligand 2 (CCL2) [43]. Furthermore, CCL2 overexpression has been confirmed in TC [44]. The process of epithelial–mesenchymal transition (EMT), which has been demonstrated to promote BC growth and metastasis, has also been associated with tamoxifen resistance. Notch1 and Notch4 have been identified as playing a pivotal role in this process. However, Notch4 exerts a more pronounced effect on the reduction in the effectiveness of tamoxifen in BC by enhancing signal transducer and activator of transcription 3 (STAT3) [45]. EMT has also been linked to cancer stem cells (CSCs) that overexpress CD44 [46]. It is a transmembrane glycoprotein with numerous isomeric variants, which, by interacting with hyaluronic acid (HA), can enhance various further signalling pathways. It has been demonstrated that the adhesive cascade of CRC cells plays a pivotal role in metastasis [47]. Furthermore, studies indicate the expression of hyaluronan synthase 2 (HAS2) by CAFs, which drives the EMT process [48]. The interaction between CD44 and HA may therefore be a potential therapeutic target. The formation of disulphide bonds between cysteine residues is a crucial aspect of the binding of these molecules. Research has identified a particularly significant influence of the bond between Cys77 and Cys92 on cancer cell metastasis. The destabilisation of this bond through redox reactions has been shown to contribute to the therapeutic regulation of the interaction between CD44 and HA [49]. Conversely, the correlation between HA turnover in the ECM and ERα expression highlights the important role of oestrogens in the TME [50]. Advancing knowledge about epigenetic regulation of the TME has the potential to play a substantial role in the development of new treatment methods. In a study on BC, the effect of histone deacetylase 6 (HDAC6) immunosuppression in CAFs was determined by STAT3 activation and increased COX2 expression. In addition, the inhibition of its activity has been shown to reduce the number of M2 subpopulation TAMs, as well as to decrease the recruitment of myeloid-derived suppressor cells (MDSCs) and Tregs [51]. The role of collagen in the TME in cancer is also a very important factor determining the risk of malignant transformation, increased invasiveness and accelerated migration. It has been demonstrated that collagen-1, the protein responsible for increasing the ECMs density, assumes a pivotal role in these processes [52]. The ratio of different collagens is also an important factor in the development of BC. For instance, the suppression of type IV alpha 2 collagen has been demonstrated to inhibit the proliferation and migration of triple-negative breast cancer (TNBC) cells [53].

4. Role of ER in TME

A significant proportion of cancers have been observed to display various isoforms of the estrogen receptor [14]. As previously stated, ERs regulate gene expression via multiple signalling pathways. These include cyclic adenosine monophosphate (cAMP), mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT) [54,55] and Janus kinase (JAK) [56] (Figure 2). This phenomenon is consistent with the conditions created by the tumour microenvironment [57]. The TME comprises a combination of cellular and non-cellular components, including CAFs, endothelial and lymphatic cells, TAMs, tumour-associated neutrophils (TANs), Tregs, and natural killer cells (NK) [58], ECM components, and chemokines [59]. The influence of these components on the pathological development of the disease has been well documented [60]. The cells that compose the TME may contribute to disease progression by expressing and secreting factors that mediate proliferation. The suppression of the anti-tumour immune response or the induction of adaptive immunity has been demonstrated to negate its inhibitory effect on tumour development [61].
CAFs are considered to be one of the key elements of the TME. The influence of these cells on the growth, invasion, metastasis, and drug resistance of cancer cells is a result of their interaction with the cells, as well as with components of the microenvironment [55]. It has been suggested that there is a correlation between CAFs and oestrogen receptor signalling due to the function performed by CAFs [62]. The correlation is complex. A notable aspect of these interactions is revealed by the mutual activation of signalling pathways at multiple levels. For example, studies have shown that estrogen-induced transcription of IL-11 (Interleukin-11), which is, among other things, a molecule of the JAK pathway, significantly increases the migration and invasion induction abilities of CAFs [63]. Additionally, the presence of estrogen receptors in CAFs has been documented. In the context of CAFs derived from TNBC, the expression levels of the ERα36 isoform have been found to fluctuate. Cells with heightened receptor expression demonstrated an escalated production of cytokines and growth factors (e.g., interleukin-1 beta (IL-1β), hepatocyte growth factor (HGF), interleukin-8 (IL-8), VEGF), which were found to provide notable support to tumour progression [64]. In TNBC, a positive correlation was also detected between GPER activity in CAFs and their increased synthesis and secretion of glutamine into the extracellular matrix, accompanied by an increase in tumour cell absorption and metabolic utilisation of glutamine. This had a significant impact on the malignancy of the tumour under study [65]. A study investigating the impact of ERα expression in CAFs in gastric cancer confirmed increased expression of matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9). These are responsible for the remodelling of the extracellular matrix, supporting tumour invasion and metastasis [62]. Notably, there are documented accounts of the role of CAFs in regulating the expression of aromatase, a key enzyme in the conversion of androgens into estrogens by cancer cells [55]. The findings of the current studies reveal the pivotal function of the estrogen receptor in tumour progression and its interaction with CAFs. Nevertheless, these findings suggest the possibility of new therapeutic treatments being developed [65].
Pro-tumour immune cells that are part of the tumor immune microenvironment (TIME), such as Tregs and TAMs, play an important role in weakening the anti-tumour immune response and creating an immunosuppressive microenvironment [66]. TAMs can be categorised into two distinct subpopulations: classically activated M1, which exhibits anti-tumour properties, and alternatively activated M2, which demonstrates pro-tumour capabilities. M2-type TAMs primarily function by secreting various growth factors, including VEGF, PDGF, EGF, as well as MMP2 and MMP9, which are metalloproteinases found in the extracellular matrix. In addition, these cells secrete cytokines such as tumour necrosis factor alpha (TNF-α), IL-1β, and IL-8. The net effect of these secretions is to promote processes such as angiogenesis and metastasis. It has also been demonstrated that they secrete heme oxygenase-1 (HO-1) and COX-2, which are involved in the processes of angiogenesis and carcinogenesis [67]. In vitro studies have confirmed that TAMs can contribute to the development of endocrine resistance in estrogen receptor-positive (ER+) breast cancer cells by prolonging the release of TNF-α and interleukin 6 both by themselves and by the cancer cells they affect. This, in turn, resulted in the activation of nuclear factor kappa-light-chain-enhancer (NF-κB), STAT3, extracellular signal-regulated kinase 1 (ERK-1) and constitutively activated ERα through its hyperphosphorylation [68]. In the case of Tregs, these cells demonstrate a relative insensitivity to changes in the extracellular environment due to their epigenetic mode of functional regulation. Their primary function is evidently to regulate autoimmune diseases. Tregs are categorised into subsets based on the type of cytokines they secrete. However, it is important to note that all subsets contribute significantly to the development of immune tolerance in tumours [69]. Tregs are modulated by estrogens. The ERα response element, located in the forkhead box P3 (FOXP3) promoter region, is an important transcriptional factor for the immunosuppressive activity of Tregs, and controls its expression [70]. Research has shown that Treg numbers decrease in ER-positive (estrogen receptor-positive) ovarian and breast tumours in mice in response to anti-estrogen treatment. This finding indicates that the administration of anti-estrogenic agents to patients diagnosed with ER+ breast cancer may lead to a reduction in Treg influx and activity, consequently enhancing anti-tumour immune responses [71]. Another type of cell that is part of the TME and is key to the immune suppression of tumours are MDSCs. These cells constitute a heterogeneous population of myeloid cells that accumulate in the TME, where they inhibit the antitumour activity of NK cells and T lymphocytes, among other things [72]. The estrogen receptor exerts a significant influence on the accumulation of MDSC, a process that is facilitated by the expression of STAT3. It has been demonstrated that ER enhances the immunosuppressive function of MDSCs [73]. A multitude of studies have confirmed that an increase in both exogenous and endogenous estrogen concentrations increases the influx of MDSCs and enhances their immunosuppressive effects through ERα activation. This progression has been identified as a contributing factor in the development of cervical cancer [74].
The ECM, which consists of a wide variety of components including proteoglycans/glycosaminoglycans, collagens, laminin, fibronectin, macromolecules, enzymes and proteins, plays a pivotal role in the TME. The ECM is responsible for the transmission of signals between cells and provides a structural framework that facilitates cell adhesion, proliferation, differentiation and migration. It is evident that ER exerts a substantial influence on ECM remodelling, consequently leading to tumour growth and metastasis [75]. Literature reports indicate that matrix metalloproteinases (MMPs) and their inhibitors, tissue inhibitors of metalloproteinases (TIMPs), play a role in cancer cell migration. Research has demonstrated that this process is modulated by the ERα-36-STAT3 complex, which has been observed to bind to the promoters of MMP2 and MMP9 metalloproteinases, function as an enhanceosome complex, and thereby enhance their expression [76].
The ECM has been shown to play a significant role in the development of drug resistance in hormone-dependent tumours. A comprehensive review of the extant literature reveals that studies have demonstrated the involvement of insulin-like growth factor (IGF) activated ERα phosphorylation, which affects the secretion of MMP2/MMP9, which contribute to the development of resistance mechanisms to anti-estrogen therapies through ECM reorganisation [77]. In addition, collagen-1-rich ECM can alter hormonal signals in ERα-positive breast cancer, causing its development and increasing the frequency of lung metastases. An increase in collagen-1 has been shown to shift the activity of signalling cascades in tumour epithelium from signal transducer and activator of transcription 5 (STAT5), a prolactin mediator associated with a milder course of the tumour, towards ERK1/2 and AKT pathways, which are poor prognostic markers [78].
It is apparent that interactions between cancer cells, stromal cells, immune cells and extracellular molecules in the tumour microenvironment are significant for carcinogenesis and metastasis. Figure 3 shows the schematic involvement of estrogens/ER in interaction with the TME.

5. The Role of the ERs in Specific Cancers

Hormones, particularly steroids, play a key role in the development of tumours or carcinogenesis in various target tissues by influencing their physiological functions and proliferation. In the tumour microenvironment, receptors that respond to signals from the extracellular matrix play a pivotal role in tumour development. In the context of the estrogen receptor, cancers such as ovarian and breast cancer are particularly notable. However, due to their high incidence levels and coverage in the literature, this review will also include thyroid and colon cancers. Below, we present an overview of selected cancers in terms of the presence and activity of the estrogen receptor.

5.1. Ovarian Cancer

Ovarian cancer is the principal gynaecological cancer. Its incidence is constantly increasing. In terms of mortality, it is one of the lethally cancers. In 2022, there were 324,603 new cases and over 206,956 deaths [79]. Moreover, it has been determined that half of all OC cases are diagnosed in individuals aged over 65. In the USA, the average age of onset of OC is 57–59 years. However, it is important to note that only 10–15% of cases occur in patients before menopause, which occurs around the age of 51 [80]. In view of these correlations, research has been undertaken to determine the causes of the increased incidence of OC in postmenopausal women. Despite the absence of a definitive understanding of the etiology of ovarian cancer due to its multifaceted nature, several hypotheses have been postulated to explain the origin of OC in relation to hormone levels. Furthermore, the literature describes gonadotropic signalling, the direct action of progesterone and androgens, and ovulation in the progression of this type of cancer [81]. Studies have shown that 17β-estradiol (E2) can stimulate the proliferation of ovarian cells, but it can also induce neoplasia. The estrogen receptor α is present in both normal ovarian cells and cancer cells. OC is characterised based on ERα expression. Increased expression of ERα occurs in over 60% of ovarian cancer cases [82]. ERα is responsible for metastasis and OC progression through specific pathways involving VEGF and MAPK [83]. Nevertheless, this receptor has been strongly associated with improved survival in endometrioid ovarian cancer [15]. Thanks to its presence in OC, many therapeutic strategies have been developed [84].
In addition to ERα, ERβ is also present in ovarian cancer. A study by Bogush et al. confirmed the presence of both receptor forms in OC [85]. In addition to the full-length ERβ, its isoforms are also present [86]. In a normal ovary, ERβ is more abundant than ERα. However, in the case of tumour development, this ratio is reversed, with ERα mRNA levels remaining comparable to those in normal tissue [87]. This correlates with reports of the suppressive effect of ERβ on OC in both in vitro and in vivo studies [15]. Furthermore, studies involving an increase in ERβ levels or its stimulation have resulted in decreased cell motility and proliferation by reducing MMP2 and cyclin A2 (CCNA2) expression and inducing p21 and fibulin-1c ( FBLN-1C) expression [88].
In the case of GPER, the rapid and independent intracellular response to extracellular ligands via signal transduction pathways is a negative prognostic factor for the overall survival of ovarian cancer patients [89]. GPER expression levels are relatively low compared to those of ERα and ERβ [28]. However, it correlates with MMP9 expression, influencing the initiation and migration of OC cells [90]. Nevertheless, some studies have suggested an inhibitory effect of GPER on OC cell proliferation [91]. This may be due, among other things, to the epigenetic effect of GPER on H3K4me3 (Histone H3 lysine 4 trimethylation) [92].

5.2. Breast Cancer

Breast cancer is the most commonly diagnosed cancer among the global female population and represents the leading cause of mortality among women with cancer [79]. There is a great amount of evidence that links estrogen to breast cancer. Current evidence appears to confirm estrogen’s oncogenic and tumour-promoting role in breast cancer risk, recurrence and progression. However, a more detailed analysis reveals that the relationship between ostrogen and breast cancer is complex. BC is a group of diseases that present with different molecular profiles and clinical-pathological characteristics. Classification of the different molecular subtypes, among other things, on the presence of the estrogen receptor, progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) [93]. Based on their expression, four distinct types of BC can be distinguished: luminal A (ER+ and/or PR+, HER2-), luminal B (ER+ and/or PR+, HER2+/-), HER2-positive (ER-, PR-, HER2+) and TNBC (ER-, PR-, HER2-) [93,94]. The function of PR, analogous to that of ER, is to regulate gene transcription in response to ligand binding [95]. Consequently, its expression represents a significant factor in the pathogenesis of BC. Furthermore, the incidence of luminal type A breast cancer increases with age, thus emphasising the role of sex hormones in the development of BC [96]. In the case of male breast cancer (MBC), an increase in incidence relative to age can also be observed, but the cause of this phenomenon is different. In more than 40% of cases, the development of MBC is attributable to genetic mutations in the BRCA1/BRCA2 or CHEK2 genes. The majority of cases of MBC are invasive cancers [97].
This has been shown to result in a poorer prognosis, a lower 5-year survival rate and an increased risk of death when compared to the incidence of BC among the female population [98]. Risk factors for the development of MBC have been identified as including Klinefelter syndrome, liver disease or testicular abnormalities, in addition to environmental exposure resulting in an elevated oestrogen-to-androgen ratio [99]. In the context of MBC, the incidence of oestrogen markers has been shown to be comparable to that observed in the population of postmenopausal women [97]. It has been established that the vast majority of MBC cases, approximately 99%, are ESR1-positive [100]. As we mentioned estrogen receptor expression represents a core method of BC classification [101]. The presence of ERα has been documented in over 70% of BC cases in women population [102]. The prognosis for BC with ERα is more favourable than for BC without ERα [103]. In normal breast tissue, the gene encoding ERα is expressed selectively in the epithelial compartment, where the receptor is detected in only about 10% of cells. The level of expression in normal cells varies according to factors such as puberty, pregnancy and lactation. In the course of BC, ERα expression is higher than in normal cells and remains at a constant level [104]. In addition to overexpression, increased activity of this receptor may result from its post-translational modifications. This is particularly important in the context of the ligand-independent activation mechanism. An example of such dependencies is the increase in ERα activity with phosphorylated serine residues (S118, S167, S305) and tyrosine (Tyr537) [105]. The isoforms ER46α and ER36α have also been detected in BC cells. ER36α is a particularly important isoform in breast cancer because it is expressed in both ER-positive and ER-negative tumours and has been positively correlated with tumour size, stage and metastasis [106,107]. Moreover, evidence suggests that this isoform may play a role in the development of resistance to tamoxifen treatment by activating sphingosine kinase 1 (SphK1) [107]. ESR1 gene mutations have also been demonstrated to play a meaningful role in the process of carcinogenesis and BC progression. Research has shown that the majority of the numerous mutations discovered relate to the LBD region. These mutations emerged as a consequence of the response to anti-estrogenic therapy [108].
In triple-negative breast cancer, the ERβ receptor is present in 20–30% of cases, which renders it a promising therapeutic target in this aggressive subtype of breast cancer. However, there is controversy regarding its potential use in TNBC therapy. This controversy pertains to a number of aspects, including the relative inactivity of ERβ in this particular type of cancer, attributable to the low expression of AP1 (activating factor 1, a heterodimer of Fos and Jun proteins) and NF-κB [109]. Notwithstanding the extensive research conducted on the subject, a plethora of inconsistencies persist with regard to the function of ERβ in BC. These outcomes may arise from the presence of various isoforms of the receptor and their distinct capacity to influence clinical trial results [110].
In a manner analogous to ERβ, GPER has been detected in both ER+ and ER- breast cancers [105,111]. As with ERβ, there is a lack of consensus regarding the effect of GPER on the development and progression of BC. A number of studies have demonstrated a correlation between the presence of GPER and poor prognosis [111,112], while others have confirmed its suppressive effect on breast cancer [113]. However, research on BC has revealed elevated GPER levels, which have also been found to impact CAFs, thereby exacerbating tumour proliferation and angiogenesis [114]. The involvement of this receptor in the feedback loop linking the induction of IL-1β in CAFs and the expression of IL1R1 (interleukin 1 type I receptor gene) induced by hypoxia in TNBC cells has also been verified [115]. The observed variation in results may be attributable to the differential expression of GPER, which has been detected in two distinct locations within cells: the cytoplasm and the cell nucleus. Cytoplasmic expression has been associated with low-stage histological subtypes of cancer, while nuclear expression has been linked to poor differentiation, which is often indicative of malignancy [116]. A similar conclusion was drawn in a study conducted by Martin et al., where low GPER expression in the cytoplasm was found to be associated with an increased mortality rate in breast cancer patients undergoing endocrine therapy [117].

5.3. Thyroid Cancer

In 2022, thyroid cancer (TC) was one of the most prevalent cancers among adolescents [118]. Statistics show that proliferative thyroid diseases are more common in women than in men. The preponderance of TC in women is notable, with an incidence of this type of cancer that is three times higher than in men [119]. In view of the marked differences in the incidence of TC according to gender, research was initiated into the impact of hormones on the development of this particular type of cancer [120]. Once again, sex hormones play an important role, this time in the development of thyroid cancer. Estrogen is a potent growth factor for both benign and malignant thyroid cells, which may explain the gender differences in the incidence of thyroid nodules and thyroid cancer. Consequently, it was determined that both normal thyroid tissue and cancerous tissue exhibit sensitivity to changes in estrogen levels [121]. Normal thyroid tissue expresses ERβ at a higher level than ERα [122]. The presence of both ERα and ERβ has been identified in TC cells. However, in TC, the number of estrogen receptor α increases, while the number of ERβ decreases [120]. It has been demonstrated that this dysregulation may result from DNA methylation of the ERβ 5′-untranslated region [123]. Moreover, a positive effect of ERα on tumour development and a protective effect of ERβ have been observed in TC, as well as in many other types of cancer [124].
In the context of papillary thyroid carcinoma (PTC), estrogen receptor signalling, activated by estradiol, exerts a notable influence on the pathogenesis of the disease. These changes include an increased level of vimentin and MMP9 and a concomitant decrease in E-cadherin [125]. In this type of cancer, it has been observed that an increase in the ERα: ERβ expression ratio induces an increase in MMP2 expression, modulating cancer invasion [124]. Research indicates that ERα exerts a regulatory influence on ETV5 (ETS-related molecule) expression, which functions as a mediator in phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PI3KCA) activation, thereby contributing to enhanced proliferation and metastasis in TC [126].
However, the results of the studies are not clear. Some of these studies contradict the well-established positive correlation between increased ER expression and the development of PTC. Nevertheless, the role of this receptor in the carcinogenesis and progression of PTC remains unquestioned [127]. Furthermore, the presence of variations in results can be attributed to differences in the methods of receptor detection and the reagents utilised in the immunohistochemical method [128]. It is important to note that a comparable pattern is evident in the case of GPER. While the relationship between GPER levels and TC progression remains unclear, a majority of studies suggest a positive impact on the development of the disease [129]. An alternative position is posited by the analysis conducted by Bertoni’s group, which indicates a correlation between reduced GPER expression and increased lymph node metastasis in primary PTC [130]. The etiology of TC also includes genetic factors that are not associated with ER. These include mutations in the BRAF, RAS and RET/PTC genes, which lead to the activation of the oncogenic MAPK pathway [131]. In addition, the risk of TC may also increase in patients with breast cancer who harbour a CHEK2 mutation. Moreover, radiotherapy for the primary cancer in children also plays a significant role in the development of TC [132]. In addition to heavy metals, radiation and air pollution, obesity is an important environmental risk factor for thyroid cancer. In addition to other factors, the increase in thyroid-stimulating hormone (TSH) levels has been observed to result in the enhancement of thyroid cell proliferation and genetic mutations, which have been identified as significant contributors to the development of TC. In addition, obesity-induced insulin resistance is associated with an increase in insulin-like growth factor 1 (IGF-1), which leads to the activation of AKT/mTOR/PI3K or ERK/RAS/MAPK pathways promoting the development of TC. Furthermore, disturbances in adipokine levels, such as those involving adiponectin, resistin or leptin [133], and increases in insulin concentration induced by insulin resistance [134], may play an important role in the development of TC.

5.4. Colorectal Cancer

It is reported that colorectal cancer is responsible for more than half a million deaths per year [56]. Colorectal cancer is unique in its dependence on sex hormones. Studies clearly indicate that the incidence and prognosis of CRC differ depending on gender. A review of the literature on CRC cases has demonstrated sexual dimorphism with regard to incidence, mortality and tumour type [135]. As the statistics show, CRC has been shown to have a higher incidence and mortality rate among men than among women [136]. Furthermore, it has been determined that estrogens play a key protective role in CRC among the female population [135]. Pre-menopausal women demonstrating low ERα and AR levels, in conjunction with high ERβ concentrations, are observed to be the least predisposed to developing CRC [137]. Consequently, a more thorough examination of the receptors stimulated by these substances is warranted. It is well known that estrogen receptors occur naturally in the colonic epithelium. As with the aforementioned types of cancer, in CRC ERα expression increases, while ERβ decreases in relation to the amount in normal cells [138]. The quantitative ratio is reversed in comparison with normal tissue, where the predominant form of ER is the β form [139]. These changes have been demonstrated to have a substantial impact on the deterioration of the patient’s condition and the prognosis, as a result of the roles played by both forms of the receptor in CRC [138]. This assertion is corroborated by a study conducted by Topi et al., which demonstrated the impact of increased ERβ expression and decreased ERα expression on the improvement in overall survival and disease-free survival in CRC patients of both sexes [140]. The same team confirmed the involvement of ERα in metastasis and poorer prognosis in patients with its high expression, among other things through its correlation with β-catenin and CysLT1R (cysteinyl leukotriene receptor 1) [141]. Furthermore, studies conducted by Hases et al. on a mouse model established that the protective mechanism of ERβ in CRC consists in its direct binding to NF-κB activators, thereby inhibiting their activity while stimulating the expression of the NF-κB inhibitor [142]. This finding aligns with the team’s other studies, which suggest that the protective function of ERβ in CRC is based on the inhibition of NF-κB-dependent inflammatory signalling [143]. Additionally, research on ERβ in CRC has revealed a particularly high proportion of ERβ2 and ERβ5 isoforms in samples taken from patients, but their role in this type of cancer remains unclear [144]. The role of GPER in CRC remains to be fully elucidated. However, experimental analyses and clinical cases have produced contradictory results when attempting to determine whether GPER has pro- or anti-cancer properties [145,146]. A study by Abancens et al. provided confirmatory evidence for the suppressor effect of GPER, demonstrating its inhibitory effect on the JUN oncogene and the Wnt/β-catenin pathway, which is overactive in CRC [147]. The primary factors contributing to the development and progression of CRC are the accumulation of mutations in the Wnt signalling pathways, epidermal growth factor receptor (EGFR), and transforming growth factor beta (TGF-β) [148]. In addition to recognised risk factors, such as the use of stimulants and family history, diabetes has been shown to have a significant impact on CRC. It has been demonstrated that this can increase the risk of precancerous polyps by 50%, with a significant effect on the incidence of CRC, particularly among younger demographics [149]. The presence of obesity has been demonstrated to be associated with an elevated risk of colorectal cancer. Research findings have indicated that a one-unit rise in body mass index (BMI) is associated with a 2–3% elevated risk of colorectal cancer [150]. Table 1 provides a summary of the role of the different types of ER in the cancers discussed.
In general, the mechanisms associated with oestrogen receptor complex signalling are extensive and appear to be specific to a given cell type. They can also interact with various signal transduction pathways in cells. In many cases, this signalling can lead to neoplasia or promote malignant transformation. It is now known that oestrogen-dependent cancers are not limited to breast or ovarian cancer. The relationship between cancers occurring with varying frequencies depending on gender is largely related to hormone-dependent regulation. A detailed understanding of this allows for the correct diagnosis of patients and appropriate treatment or preventive measures.

6. SERMs/SERDs/AIs

Hormonal/endocrine therapy inhibits the proliferation of cancer cells through the blocking of hormone effects (use of hormone receptor antagonists or downregulation/degradation of hormone receptors) or by inhibiting hormone production [151].
Hormonal therapy is used to treat hormone-dependent cancers such as breast cancer [152], prostate cancer [153], and gynecological cancers [154], but also in the treatment of thyroid cancer [155]. In many hormone-dependent cancers, the functioning of signaling pathways mediated by estrogen receptors, among others, is disrupted [156].
Selective estrogen receptor modulators and selective estrogen receptor degraders/downregulators are used in hormonal therapy estrogen receptor-positive cancer. SERMs block/inhibit signal transmission involving ER (alpha and/or beta). Tamoxifen, toremifene, and raloxifene are selective estrogen receptor modulators currently in use [157,158,159]. Fulvestrant is selective ER downregulator/degrader (SERDs) which bind to ERα, leading to receptor downregulation/degradation and in consequence inhibition of signaling involving the ERα [160]. In addition, hormone therapy for ER-positive cancers uses aromatase inhibitors, enzyme responsible for the synthesis of estrogens from androgens. Third-generation aromatase inhibitors include letrozole, anastrozole, and exemestane [161].
Tamoxifen is one of the most commonly used drugs in adjuvant hormone therapy for ER-positive breast cancer [162]. Tamoxifen’s metabolites, afimoxifen (4-hydroxytamoxifen, 4-OHT) and endoxifen (4-hydroxy, N-desmethyl-tamoxifen) [163,164,165], a nonsteroidal ER antagonist, competitively bind to ER, causing a conformational change that promotes the release of HSP90 and ER dimerization. In the cell nucleus, the ER dimer participates in the activation of AF1 domain and the inhibition of AF2 domain, then binds to ERE in the promoter regions and stimulates AF-1-mediated gene transcription. Transcription of estrogen-responsive genes is inhibited by the tamoxifen-ER dimer because the ligand-dependent AF2 domain is inactivated and the binding of ER coactivators is restricted [166,167].
The antiestrogenic effect of tamoxifen is also achieved through the recruitment of co-repressor proteins, such as NcoR/SMRT (nuclear receptor co-repressor/ silencing mediator of retinoic acid and thyroid hormone receptors) by tamoxifen-ER dimers complex HDAC (histone deacetylases) and HDACs (HDAC3), which leads to the repression of ER-dependent gene transcription [168,169,170,171]. Tamoxifen is an estrogen antagonist in breast epithelium and is therefore used in adjuvant treatment of breast cancer. However, it acts as an agonist in other estrogen-dependent organs, including the endometrium, where it may promote endometrial proliferation disorders, including hyperplasia, and increase the risk of uterine and endometrial cancer [172].
Toremifene, a chlorinated derivative of tamoxifen, is a nonsteroidal SERM used to treat both early and late stages of metastatic ER-positive breast cancer [173]. The main active metabolite of toremifene is 4OH-NDM (N-desmethyl) -toremifene and 4-hydroxy (4-OH) toremifene [174,175] exhibit antagonistic effects to estrogens in target tissues such as breast epithelium, and partial agonistic effects in uterine and bone tissues [173,176]. The mechanism of action of toremifene is analogous to that of tamoxifen. After binding to ER, toremifene metabolites initiate ER dimerization, and the ER-toremifene complex is translocated to the nucleus, leading to activation of the AF1 domain, inactivation of the AF2 domain, and binding of the dimer to the ERE of target genes promoters. The toremifene-ER complex reduces the binding of ER coactivators, except for inactivation AF2 domain, resulting in reduced transcription of estrogens-dependent genes [167,176,177].
Raloxifene is a selective estrogen receptor modulator belonging to the benzothiophenes. Raloxifene is the only benzothiophene SERM used to reduce the risk of breast cancer. Like tamoxifen and toremifene, raloxifene also exhibits agonistic or antagonistic effects on estrogen depending on the target tissue [175,178].
Raloxifene exhibits greater affinity for ER than its main glucuronide conjugate metabolite, raloxifene-4′-β-glucuronide [179,180]. The estrogenic effect of raloxifene occurs in bone and lipid metabolism, while the antiestrogenic effect occurs in breast tissue, and a neutral effect is observed in the endometrium [181,182].
After binding to ER via the benzothiophene ring, raloxifene causes a change in the conformation of ER, which leads to the activation of the AF1 domain and inactivation of the AF2 domain of ER. The raloxifene-ER complex then binds to the ERE of target gene promoters [180]. In addition, the raloxifene-ER complex reduces the binding of ER coactivators, leading to a decrease in the transcription of estrogen-dependent genes. Furthermore, the raloxifene-ER complex recruits co-regulators to further enhance the antiestrogenic effects of raloxifene [167]. However, raloxifene has a different effect on other tissues, such as bone [183]. The raloxifene-ER complex, with the participation of various proteins (adaptors, enhancers), is able to bind to and activate a specific DNA sequence known as the Raloxifene Responding Element (RRE). The binding of the raloxifene-ER complex to RRE causes the transcription of genes involved in the synthesis of specific cellular proteins responsible for the estrogenic growth and proliferative effects of raloxifene on these tissues [184,185].
Fulvestrant is a 7α-alkylsulfinyl analogue of estradiol that competes with estrogens for binding to ER [186]. The major metabolite of fulvestrant is fulvestrant-3-sulfate conjugate [187,188].
Unlike SERMs, which exert both agonist and antagonist effects depending on the target tissue, fulvestrant exhibits exclusively antiestrogenic activity. Fulvestrant exerts its antiestrogenic effects through various mechanisms, including impairment of ER dimerization, inhibition of ER activity, and accelerated ER proteasomal degradation The antiestrogenic effect is the result of competitive binding of fulvestrant to ER monomers, which significantly impairs ER dimerization, fulvestrant-ER complex formation and translocation to the nucleus, resulting in inhibition of transcriptional activity (associated with both the AF1 and AF2 domains) of estrogen-dependent genes [189].The fulvestrant-ER complex is unstable, which may make it to accelerated proteasomal degradation of the ER compared with ER bound to estradiol or SERMs. Accumulation of the fulvestrant-ER complex in the cytoplasm may also promote its degradation, which may reduce the number of functional ERs in cells. The reduction in ER protein levels in cells caused by fulvestrant is not due to a reduction in ER mRNA levels but rather to accelerated receptor degradation due to the instability of the fulvestrant-ER complex [190,191].
AIs are drugs that lower estrogen levels by deactivating aromatase, blocking aromatization, and inhibiting estrogen synthesis [192]. Anastrozole and letrozole are nonsteroidal aromatase inhibitors, containing a triazole functional group that competitively interacts with the heme prosthetic group of aromatase, relative to androgenic substrates. These drugs can be displaced from aromatase binding sites, allowing potential recovery of enzyme activity. Exemestane is a steroidal aromatase inhibitor, which irreversibly inactivates the enzyme by forming covalent bonds with its active site [193,194]. The origin, mechanism of action, and reversibility/irreversibility of aromatase inhibitors influence not only the potency and selectivity of the inhibitors but also their side effect profiles, which are important factors in their clinical use [195].

7. Hormone Therapy Resistance

Sex hormones are ubiquitous. Estrogens are present and locally produced in many cancers. Estrogen receptors are not without importance in oncogenesis. Controlling the signalling of hormone receptors, including the oestrogen receptor (ER), inhibits the activity of cancer cells. This strategy forms the basis of hormone therapy for cancer. However, due to the complexity of sex hormone action—including interactions with the receptors themselves, induction of signal transduction through various signalling pathways, and interaction with the tumour microenvironment—a number of processes can lead to resistance to hormone therapy. Genetic, epigenetic and transcriptomic changes in cancer cells affect not only the heterogeneity of cancers, but also the selection and success of anticancer therapies [196,197,198]. Furthermore, cancers consist not only of cancer cells, but also of other cells and molecules in the surrounding TME that influence disease progression and the success of anticancer therapies [199].
The tumour microenvironment—a complex and dynamic structure comprising tumour cells, stromal components, the vascular system and infiltrating immune cells. The TME also plays an important role not only in tumour progression and evasion of immune response, but also in the acquisition of resistance to both conventional and targeted therapies by cancer cells [200,201,202].
Extracellular vesicles, including exosomes, play a key role in modulating complex interactions between cancer cells and the TME, tumour growth, angiogenesis and metastasis. Some components of exosomes, such as signalling proteins, miRNA, and lncRNA [203,204], may contribute to the development of resistance to treatment, including hormone therapy [205,206].
During anticancer hormone therapy, which brings significant benefits, intrinsic (primary, de novo) resistance or acquired resistance may occur [207]. Primary resistance is defined as disease progression within 6 months of starting hormonal treatment for metastatic breast cancer or recurrence within 2 years of starting adjuvant therapy for early breast cancer. Secondary hormone resistance, defined as progression occurring 6 or more months after the start of hormone therapy for metastatic breast cancer, ultimately develops in most patients. Recurrence during adjuvant hormone therapy, after the first 2 years or within 1 year of completion of adjuvant hormone therapy, is also commonly characterised as acquired secondary resistance [208,209].
Tamoxifen is often used in the adjuvant treatment of ER-positive breast cancer [210]. Despite the benefits of tamoxifen in patients with all stages of ERα+ positive breast cancer, drug resistance may develop [211]. Resistance to tamoxifen treatment may be caused by disruption of multiple molecular pathways: ER signalling, tyrosine kinase receptor signalling pathways (HER2, EGFR, fibroblast growth factor receptor (FGFR) and insulin-like growth factor 1 receptor (IGF1R)), the PI3K-PTEN/AKT/mTOR pathway, and NF-κB signalling [212,213,214,215].
Additional mechanisms of tamoxifen resistance include an imbalance between anabolism and catabolism of estradiol [216], altered bioavailability of tamoxifen [217], increased angiogenesis, heterogeneity of tumour cell populations, or overexpression of growth factors [218,219].
Growth factors that activate tyrosine kinases trigger signalling pathways activated by receptor tyrosine kinases (RTKs), and the non-genomic activity of cell membrane-associated estrogen receptors facilitates cross-communication between these receptors and signalling pathways [213]. Overexpression and activation of EGFR and HER2 leads to cell proliferation and survival through activation of MAPK and PI3K/AKT signalling pathways, thereby contributing to the development of resistance to endocrine therapy [220,221].
Possible causes of tamoxifen resistance associated with ER include increased signalling of growth factors and ER associated with the cell membrane [213], loss of ERα expression and acquired ERα mutations [222]. Loss of ERα expression may be the main mechanism of de novo resistance to hormone therapy and is mainly associated with epigenetic changes i.e. abnormal methylation of CpG islands and increased histone deacetylation [223,224]. ERα mutations can lead to the formation of a receptor with altered properties, which may result in altered molecular signalling. The G protein-coupled ER may also play a role in tamoxifen resistance. Signalling by GPER occurs through transactivation of EGFR and involves tyrosine kinases from the Src, MAPK/ERK and CREB/CRE families [225,226,227].
In women, adrenal and ovarian androgens are the source of pre- and post-menopausal estrogens, as they are converted into estradiol [228]. The androgen receptor (AR) is a ligand (androgen)-activated transcription factor and a member of the nuclear receptor (NR) superfamily. Tumours resistant to tamoxifen show higher levels of AR expression than tamoxifen-sensitive tumours. High AR expression may have an adverse effect on the prognosis of tamoxifen treatment for ERα-positive breast cancer, as increased AR expression may potentially enhance the agonistic properties of tamoxifen [229,230]. The ability of AR to regulate gene transcription results from its interaction with specific DNA sequences located near or within the promoter of the target gene [231]. This is a classic ligand-dependent genomic mechanism [232]. A ligand-independent mechanism triggers AR activation through signalling pathways that include JAK/STAT3, MAPK, NOTCH, and PI3K/mTOR/AKT [233,234]. AR can also rapidly initiate cytoplasmic signalling cascades, including activation of protein kinase A, protein kinase C and ERK, through a non-genomic mechanism involving binding to cytoplasmic and membrane-associated proteins such as c-Src [235].
A new molecular target in the potential treatment of tamoxifen-resistant breast cancer is the Hedgehog signalling pathway, along with the proteins patched 1 and patched 2 (PTCH1 and PTCH2), Hedgehog-interacting protein HHIP and Glioma-associated oncogene homolog 1 and 2 (GLI1/GLI2), which can trigger positive or negative feedback in this pathway [236]. Additionally, a pathway that may interact with Hedgehog signalling in breast cancer is TGF-β, which stimulates the upregulation of GLI1 and GLI2 transcription [237,238]. The PI3K/AKT pathway is also involved in the activation of Hedgehog signalling and may play a role in promoting resistance to tamoxifen [239].
Therapies targeting various receptors and individual components of signalling pathways are currently used not only to improve treatment efficacy but also to prevent the development of resistance to standard treatment. CDK4/6 inhibitors are drugs used to treat certain types of HR+/HER2- breast cancer by blocking the cyclin-dependent kinases 4 and 6 (CDK4/6) enzymes, which are crucial for cell division. Palbociclib and abemaciclib are both CDK4/6i used to treat hormone receptor-positive (HR+), HER2-negative breast cancer, often in combination with endocrine therapy. However, despite the enormous improvement achieved with the introduction of CDK4/6i in combination with hormone therapy, 10% of patients do not respond to treatment, developing de novo resistance. Conversely, others may develop acquired resistance to CDK4/6 inhibitors and disease recurrence [240,241].
Therefore, inhibitors of other important targets with potential involvement in the development of resistance, such as: mTOR inhibitors (Everolimus) [242], PI3K inhibitors (Alpelisib, Pictilib and Buparlisib) [243], AR antagonists (Enzalutamide and Bicalutamide) [244,245], histone deacetylase inhibitors (Entinostat and Vorinostat) [246,247], and MAPK pathway inhibitors (Selumetinib and Ralimetinib) [248,249,250] have been progressed, and some of them are already in clinical use.
A new generation of anti-estrogen therapies has been designed to overcome some of these resistance mechanisms and eliminate the limitations of currently available hormone therapy, e.g., the agonistic activity of tamoxifen. These therapies include variations on existing drug classes, including SERMs other than tamoxifen and new orally administered SERDs. New-generation SERDs reduce ER levels through proteasome-dependent degradation [251].
New classes of anti-estrogen drugs include complete estrogen receptor antagonists (CERANs), selective covalent estrogen receptor antagonists (SERCAs), and proteolysis-targeting chimeras (PROTACs) targeting ER. These drugs are at various stages of development and are being evaluated in both early and metastatic stages of cancer.
SERMs exhibit ER antagonist or agonist activity, depending on the cell type, through the recruitment of different coactivators and correpressors. SERMs inhibit the AF2 of ER but allow agonistic signalling through the AF1 via other signalling pathways such as PI3K-AKT-mTOR, RAS-RAF-MAPK and CDK4/6-RB-E2F. Activation of some of these pathways may be involved not only in primary or secondary resistance to hormonal therapy but also to CDK4/6i [252].
Lasofoxifene is a new-generation non-steroidal SERM with ER binding capacity comparable to 17β-estradiol. Clinical trials are evaluating the efficacy of lasofoxifene in the treatment of metastatic breast cancer in women with ESR1 mutation (compared to fulvestrant) who have progressed after prior treatment with AIs and CDK4/6i [253], and the efficacy of combining lasofoxifene with abemaciclib (a CDK4/6 inhibitor) [254].
SERDs not only act as competitive ER antagonists, they also induce proteasome-dependent degradation of the ER [255]. Fulvestrant is a SERD administered intramuscularly, which is a certain limitation. Currently, orally administered SERDs are being evaluated in clinical trials for use in metastatic, adjuvant and neoadjuvant settings. Elacestrant (RAD1901) is an orally bioavailable SERD, which is a dose-dependent mixed ER agonist/antagonist. Elacestrant at high doses acts as a direct ER antagonist as well as selective downregulator of ER. The binding of RAD1901 allows the presentation of a protein–protein interaction surface in the hinge region of the DNA and hormone-binding domain of ERα/ESR1, crucial for transcriptional activity. Moreover, increased receptor occupancy results in the receptor being targeted for degradation, which quantitatively inhibits ERα/ESR1 signalling [256,257].
Elacestrant has been approved for the treatment of advanced or metastatic breast cancer in patients with ER+, ESR1-mutated and HER2-negative (HER2-) profiles, after progression following at least one line of endocrine therapy [258]. Elacestrant in combination with abemaciclib in patients with brain metastasis from ER+, HER2-negative breast cancer is also being investigated [259].
Other orally administered SERDs include: giredestrant (GDC-9545), also used in combination with palbociclib, everolimus and in combination with CDK 4/6 inhibitors, ipatasertib, inavolisib, everolimus and samuraciclib [207]; camizestrant (AZD9833) used as monotherapy and in combination with palbociclib and in combination with abemaciclib, everolimus and capiwasertib [260] and imlunestrant (LY-3484356). Imlunestrant is an oral SERD, and synergistic effects have been observed in combination with abemaciclib, everolimus, and alpelisib [261]. SERDs include Rintodestrant (G1T48), AZD9496, ZN-c5, D-0502 (Taragarestrant) and Borestrant (ZB-716). Rintodestrant is an oral SERD that competitively binds and degrades the estrogen receptor. AZD9496 is an oral, non-steroidal, selective ER degrader and inhibitor of ER-dependent signalling pathways. Borestrant is fulvestrant modified with boronic acid, with oral bioavailability [22].
Proteolysis targeting chimerics are dual-function hybrids that simultaneously bind to a specific target protein, such as ER, and an E3 ubiquitin ligase, causing ubiquitination and degradation of the target ER protein via the ubiquitin-proteasome system [262]. Because their mechanism of action is catalytic, they are able to promote protein degradation even at low doses. The most advanced PROTAC is ARV-471 [263,264].
Complete estrogen receptor antagonists block both the AF1 and AF2 transcription activation domains of ER. The AF1 domain can be activated by signalling pathways such as mTOR, PI3K and MAPK, and the AF2 domain, which is activated by the estrogen ligand itself. Activation of AF1 and AF2 leads to gene transcription and cell proliferation. CERANs directly inhibit AF2, allowing agonistic signalling involving AF1 via other signalling pathways [265]. OP-1250 is an orally bioavailable CERAN that also acts as a SERD inducing ER degradation. OP-1250 (Palazestrant), is a complete estrogen receptor antagonist, inhibits wild-type and mutant ER-positive breast cancer models as monotherapy and in combination with other drugs [266].
The first-in-class selective covalent estrogen receptor antagonist drug, H3B-5942, covalently inactivates both wild-type and mutant ERα by targeting Cys530 and forcing a unique antagonistic conformation. H3B-5942 potently suppresses ERα function and demonstrates potent antiproliferative and antitumour activity in monotherapy and in combination with CDK4/6 inhibitors (palbociclib, ribociclib, and abemaciclib) or mTOR inhibitors (everolimus, temsirolimus, sirolimus), leading to synergistic activity [267].
Moreover, microRNAs (miRNAs) have emerged as modulators of therapy response and resistance to hormonal therapy [268]. lncRNAs (long non-coding RNAs) also play a leading role in the initiation, progression and resistance to anti-estrogens [269].
Understanding the mechanisms that lead to resistance to hormone therapy enables us to avoid ineffective therapies and propose strategies to circumvent this resistance, providing direction for the search for new drugs.

8. Summary

Hormones are one of the most important groups of substances that regulate how our body functions. At the same time, some cancers use hormones to control their growth and spread. Understanding the mechanisms of action and connections in hormonal regulation is therefore an important diagnostic and therapeutic tool. The wide range of interactions between hormone-dependent cancers and their microenvironment is a promising area of research for the development of new therapies. In addition to the pharmaceutical treatments, hormonal therapy for cancer employs techniques that involve the removal of sex hormones from the body. One such technique is castration, which primarily inhibits the development of breast and endometrial cancer in women, as well as prostate cancer in men. The main castration methods include surgical castration and pharmacological castration, which does not require the removal of the gonads. Pharmacological castration is a therapeutic intervention that involves the administration of gonadotropin-releasing hormone (GnRH) analogues. These pharmaceuticals are designed to bind to the receptor located within the pituitary gland, thereby inducing a castration effect. This therapeutic modality is predominantly employed in the management of premenopausal women diagnosed with breast cancer, particularly those below the age of 35. In male patients, this method is utilised when surgical castration is contraindicated or when estrogen therapy is not possible [270]. However, it should be noted that this treatment method is not without its potential disadvantages. In women diagnosed with breast cancer, the induction of premature menopause through the administration of GnRH agonists/antagonists has been proven to be both effective and reversible. However, this treatment is associated with a considerable deterioration in quality of life and a long-term risk of mortality [271]. This method represents the primary treatment option for metastatic prostate cancer (PC). However, it has often been observed that this results in cancer cells becoming insensitive to androgens, which in turn leads to the development of castration-resistant PC (CRPC) with a poor prognosis [272]. In addition to their role in oncological treatment, hormone therapies also play an important role in other aspects of life. Hormone replacement therapy (HRT) involves the administration of agents that induce an estrogenic or progestogenic effect. It is indicated in cases where the ovaries cease to produce these hormones naturally, as a result of cancer treatment, as well as genetic and autoimmune diseases, cardiovascular diseases and accelerated bone loss [273]. However, the potential for developing hormone-dependent cancer should not be overlooked. The type of HRT used should therefore be appropriately selected according to the patient’s age and health status in order to minimise this risk [274]. The risk of developing cancer is also influenced by the use of contraceptives. Research has indicated a correlation between the ingestion of oral contraceptives and an elevated risk of BC in women, particularly those with a mutation in the BRCA1 or BRCA2 gene, in comparison with women who do not utilise such contraceptives. This elevated risk persists for a more than 10-year period following the end of use [275]. However, in cases of OC risk in these patients, oral contraceptive pills play a chemopreventive role. This enduring effect persists for a period of up to 15 years following the discontinuation of the drug. It is estimated that the use of oral contraceptive pills reduces the risk of OC by more than 42%. This duality of action necessitates a meticulous analysis of each case by doctors, with a view to selecting the most appropriate type of substance [276].
However, it should be noted that exogenous estrogens are not only associated with therapeutic use. Humans are constantly exposed to the effects of these substances, which are present in the environment. Xenoestrogens are synthetic substances found in plastics, preservatives, pesticides, and other such products, which have an ability to bind to the ER and to activate or block its activity. For instance, bisphenol A has been demonstrated to disrupt ER activity, thereby affecting intracellular signalling and inducing an increase in ER expression. This results in increased cell proliferation, an increase in the number of cells with ER and PR, and activation of AKT and ERK pathways, which in turn leads to, among other things, an increased risk of BC, PC and bladder cancer [277].
In addition to xenoestrogens, exposure to phytoestrogens is a constant factor in the human environment. Unlike xenoestrogens, these compounds occur naturally in plants and share a chemical structure with mammalian estrogens. This enables them to exert a regulatory influence over cellular processes via ER. These properties have led to their widespread use as a treatment for menopausal symptoms without resulting in side effects. However, the ability of these compounds to modulate signalling pathways through ER activation may contribute to the development of tumours. For instance, genistein has been observed to increase the expression of ERα, AKT and NF-κB, thereby stimulating cell proliferation. However, it has been demonstrated that certain phytoestrogenic compounds possess anti-cancer properties. As demonstrated in the relevant studies, the stereospecificity of phytoestrogens plays a pivotal role in their biological activity [278]. The role of plants in providing compounds with hormonal activity extends beyond this. They have the ability to absorb estrogenic steroids present in water and soil, subsequently accumulating these chemicals within their own tissues. The consumption of these plants has been demonstrated to exert a substantial influence on estrogen levels within the human body [279].
In light of the multifaceted mechanisms of estrogen receptor ER activation exhibited in this article, the extensive cellular effects they engender, and the numerous sources of exposure to substances that interact with them, it is crucial to emphasise the pivotal role of ER in developing novel therapeutic interventions that target hormone-dependent cancers.

Author Contributions

Conceptualization: O.G. and I.A.B.; writing—original draft preparation, O.G., M.J. and I.A.B.; writing—review and editing, O.G., M.J. and I.A.B.; visualization—O.G.; supervision: I.A.B.; project administration: I.A.B.; funding acquisition: I.A.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Medical University of Silesia, Katowice, Poland; research grant number: BNW-1-091/K/4/I and BNW-1-096/N/5/I.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AFActivation function domains
AktProtein kinase B
ARAndrogen receptor
AIsAromatase inhibitors
AP1Activating factor 1
ARAromatase
BCBreast cancer
BMIBody mass index
CAFsCancer-associated fibroblasts
cAMPCyclic adenosine monophosphate
CCL2CC-chemokine ligand 2
CCNA2Cyclin A2
CDK4/6Cyclin-dependent kinases 4 and 6
CDK4/6iCyclin-dependent kinase 4/6 inhibitor
CERANsComplete estrogen receptor antagonists
COX-2Cyclooxygenase-2
CRCColorectal cancer
CREBCyclic AMP-responsive element-binding protein
CRPCCastration-resistant prostate cancer
CSCsCancer stem cells
CysLT1RCysteinyl leukotriene receptor 1
DBDDNA-binding domain
ECMExtracellular matrix
EGFEpidermal growth factor
EGFREpidermal growth factor receptor
EMTEpithelial–mesenchymal transition
EREstrogen receptor
ER+Estrogen receptor-positive
ERαEstrogen receptor alpha
ERβEstrogen receptor beta
EREEstrogen response elements
ERK-1Extracellular signal-regulated kinase 1
ER-positiveEstrogen receptor-positive
ESR1Estrogen receptor 1 gene
ESR2Estrogen receptor 2 gene
E217β-estradiol
FGFRFibroblast growth factor receptor
FBLN-1CFibulin-1C
FOXP3Forkhead box P3 gene
GLI1Glioma-associated oncogene homolog 1
GLI2Glioma-associated oncogene homolog 2
GnRHGonadotropin-releasing hormone
GPERG protein-coupled estrogen receptor
HAHyaluronic acid
HAS2Hyaluronan synthase 2
HDAC3Histone deacetylase 3
HDAC6Histone deacetylase 6
HER2Human epidermal growth factor receptor 2
HGFHepatocyte growth factor
HHIPHedgehog-interacting protein
HO-1Heme oxygenase-1
HRTHormone replacement therapy
H3K4me3Histone H3 lysine 4 trimethylation
IGFInsulin-like growth factor
IGF-1Insulin-like growth factor 1
IGF1RInsulin-like growth factor 1 receptor
IL-1βInterleukin-1 beta
IL1R1Interleukin 1 type I receptor gene
IL-2Interleukin-2
IL-4Interleukin-4
IL-6Interleukin-6
IL-7Interleukin-7
IL-8Interleukin-8
IL-11Interleukin-11
ITGA5Integrin 5α
LBDLigand-binding domain
MAPKMitogen-activated protein kinase
MBCMale breast cancer
MCAMMelanoma cell adhesion molecule
MDSCMyeloid-derived suppressor cell
MMPsMatrix metalloproteinases
MMP2Matrix metalloproteinase-2
MMP9Matrix metalloproteinase-9
mTORMammalian target of rapamycin
NF-κBNuclear factor kappa-light-chain-enhancer
NKsNatural killer cells
NRNuclear receptor
NTDN-terminal nucleotide binding domain
OCOvarian cancer
PCProstate cancer
pCREB1Phosphorylated cAMP-responsive element-binding protein 1
PD-1Programmed cell death protein-1
PDGFPlatelet-derived growth factor
PD-L1Programmed cell death-ligand 1
PIP2Phosphatidylinositol 4,5-bisphosphate
PIP3Phosphatidylinositol 3,4,5-trisphosphate
PI3KPhosphatidylinositol 3-kinase
PI3KCAPhosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha
PRProgesterone receptor
PROTACsproteolysis-targeting chimeras
PTCPapillary thyroid carcinoma
PTCH1Proteins patched 1
PTCH2Proteins patched 2
PTENPhosphatidylinositol-3,4,5-trisphosphate 3-phosphatase
RasGTPase HRas
RRERaloxifene responding element
RTKsReceptor tyrosine kinases RTKs
SERCAsSelective covalent estrogen receptor antagonists
SERDsSelective estrogen receptor downregulators
SERMsSelective estrogen receptor modulators
SrcProto-oncogene tyrosine-protein kinase
STAT3Signal transducer and activator of transcription 3
STAT5Signal transducer and activator of transcription 5
TCThyroid cancer
TregRegulatory T lymphocyte
TAMsTumour-associated macrophages
TANsTumour-associated neutrophils
TGF-βTransforming growth factor beta
TGFβ1Transforming growth factor beta 1
TIMETumor immune microenvironment
TIMPsTissue inhibitors of metalloproteinases
TMETumour microenvironment
TNBCTriple-negative breast cancer
TNF-αTumour necrosis factor alpha
TSHThyroid-stimulating hormone
VEGFVascular endothelial growth factor

References

  1. Bray, F.; Laversanne, M.; Sung, H. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  2. Dee, E.C.; Wu, J.F.; Feliciano, E.J.G.; Ting, F.I.L.; Willmann, J.; Ho, F.D.V.; Jain, B.; Jain, U.; Chen, J.; Moraes, F.Y.; et al. National Cancer System Characteristics and Global Pan-Cancer Outcomes. JAMA Oncol. 2025, 11, 650–654. [Google Scholar] [CrossRef]
  3. Jafari, H.; Hussain, S.; Campbell, M.J. Nuclear Receptor Coregulators in Hormone-Dependent Cancers. Cancers 2022, 14, 2402. [Google Scholar] [CrossRef]
  4. Hamid, A.A.; Issa, M.B.; Nizar, N.N.A. Hormones. In Preparation and Processing of Religious and Cultural Foods; Elsevier: Amsterdam, The Netherlands, 2018; pp. 253–277. [Google Scholar]
  5. Montaruli, A.; Castelli, L.; Mulè, A.; Scurati, R.; Esposito, F.; Galasso, L.; Roveda, E. Biological Rhythm and Chronotype: New Perspectives in Health. Biomolecules 2021, 11, 487. [Google Scholar] [CrossRef]
  6. Liu, Y.C.; Yeh, C.T.; Lin, K.H. Molecular Functions of Thyroid Hormone Signaling in Regulation of Cancer Progression and Anti-Apoptosis. Int. J. Mol. Sci. 2019, 20, 4986. [Google Scholar] [CrossRef]
  7. Chu, L.; Shu, X.; Huang, Y.; Chu, T.; Ge, M.; Lu, Q. Sex steroid hormones in urinary exosomes as biomarkers for the prediction of prostate cancer. Clin. Chim. Acta 2022, 531, 389–398. [Google Scholar] [CrossRef]
  8. Costa, A.R.; Lança de Oliveira, M.; Cruz, I.; Gonçalves, I.; Cascalheira, J.F.; Santos, C.R.A. The Sex Bias of Cancer. Trends Endocrinol. Metab. 2020, 31, 785–799. [Google Scholar] [CrossRef]
  9. Boye, A.; Osei, S.; Brah, A. Therapeutic prospects of sex hormone receptor signaling in hormone-responsive cancers. Biomed. Pharmacother. 2024, 180, 117473. [Google Scholar] [CrossRef]
  10. Pakdel, F. The Role of Estrogen Receptors in Health and Disease. Int. J. Mol. Sci. 2023, 24, 11354. [Google Scholar] [CrossRef] [PubMed]
  11. Pakdel, F. Molecular Pathways of Estrogen Receptor Action. Int. J. Mol. Sci. 2018, 19, 2591. [Google Scholar] [CrossRef] [PubMed]
  12. Song, D.; He, H.; Indukuri, R.; Huang, Z.; Stepanauskaite, L.; Sinha, I.; Haldosén, L.A.; Zhao, C.; Williams, C. ERα and ERβ Homodimers in the Same Cellular Context Regulate Distinct Transcriptomes and Functions. Front. Endocrinol. 2022, 13, 930227. [Google Scholar] [CrossRef]
  13. Rae, J.M.; Lippman, M.E. The role of estrogen receptor signaling in suppressing the immune response to cancer. J. Clin. Investig. 2021, 131, e155476. [Google Scholar] [CrossRef]
  14. Chen, P.; Li, B.; Ou-Yang, L. Role of estrogen receptors in health and disease. Front. Endocrinol. 2022, 13, 839005. [Google Scholar] [CrossRef] [PubMed]
  15. Langdon, S.P.; Herrington, C.S.; Hollis, R.L.; Gourley, C. Estrogen Signaling and Its Potential as a Target for Therapy in Ovarian Cancer. Cancers 2020, 12, 1647. [Google Scholar] [CrossRef]
  16. Arneth, B. Tumor Microenvironment. Medicina 2019, 56, 15. [Google Scholar] [CrossRef]
  17. Kozieł, M.J.; Piastowska-Ciesielska, A.W. Estrogens, Estrogen Receptors and Tumor Microenvironment in Ovarian Cancer. Int. J. Mol. Sci. 2023, 24, 14673. [Google Scholar] [CrossRef] [PubMed]
  18. Fuentes, N.; Silveyra, P. Estrogen receptor signaling mechanisms. Adv. Protein Chem. Struct. Biol. 2019, 116, 135–170. [Google Scholar] [PubMed]
  19. Patel, H.K.; Bihani, T. Selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs) in cancer treatment. Pharmacol. Ther. 2018, 186, 1–24. [Google Scholar] [CrossRef]
  20. Brett, J.O.; Spring, L.M.; Bardia, A.; Wander, S.A. ESR1 mutation as an emerging clinical biomarker in metastatic hormone receptor-positive breast cancer. Breast Cancer Res. 2021, 23, 85. [Google Scholar] [CrossRef]
  21. Vasan, N.; Baselga, J.; Hyman, D.M. A view on drug resistance in cancer. Nature 2019, 575, 299–309. [Google Scholar] [CrossRef]
  22. Neupane, N.; Bawek, S.; Gurusinghe, S.; Ghaffary, E.M.; Mirmosayyeb, O.; Thapa, S.; Falkson, C.; O’Regan, R.; Dhakal, A. Oral SERD, a Novel Endocrine Therapy for Estrogen Receptor-Positive Breast Cancer. Cancers 2024, 16, 619. [Google Scholar] [CrossRef]
  23. Bardia, A.; Cortés, J.; Bidard, F.C.; Neven, P.; Garcia-Sáenz, J.; Aftimos, P.; O’Shaughnessy, J.; Lu, J.; Tonini, G.; Scartoni, S.; et al. Elacestrant in ER+, HER2- Metastatic Breast Cancer with ESR1-Mutated Tumors: Subgroup Analyses from the Phase III EMERALD Trial by Prior Duration of Endocrine Therapy plus CDK4/6 Inhibitor and in Clinical Subgroups. Clin. Cancer Res. 2024, 30, 4299–4309. [Google Scholar] [PubMed]
  24. Gong, Z.; Yang, S.; Wei, M.; Vlantis, A.C.; Chan, J.Y.K.; van Hasselt, C.A.; Li, D.; Zeng, X.; Xue, L.; Tong, M.C.F.; et al. The Isoforms of Estrogen Receptor Alpha and Beta in Thyroid Cancer. Front. Oncol. 2022, 12, 916804. [Google Scholar] [CrossRef]
  25. Belluti, S.; Imbriano, C.; Casarini, L. Nuclear Estrogen Receptors in Prostate Cancer: From Genes to Function. Cancers 2023, 15, 4653. [Google Scholar] [CrossRef]
  26. Arao, Y.; Korach, K.S. The physiological role of estrogen receptor functional domains. Essays Biochem. 2021, 65, 867–875. [Google Scholar] [CrossRef]
  27. Bano, A.; Stevens, J.H.; Modi, P.S.; Gustafsson, J.Å.; Strom, A.M. Estrogen Receptor β4 Regulates Chemotherapy Resistance and Induces Cancer Stem Cells in Triple Negative Breast Cancer. Int. J. Mol. Sci. 2023, 24, 5867. [Google Scholar] [CrossRef]
  28. Tirado-Garibay, A.C.; Falcón-Ruiz, E.A.; Ochoa-Zarzosa, A.; López-Meza, J.E. GPER: An Estrogen Receptor Key in Metastasis and Tumoral Microenvironments. Int. J. Mol. Sci. 2023, 24, 14993. [Google Scholar] [CrossRef]
  29. Barton, M.; Filardo, E.J.; Lolait, S.J.; Thomas, P.; Maggiolini, M.; Prossnitz, E.R. Twenty years of the G protein-coupled estrogen receptor GPER: Historical and personal perspectives. J. Steroid Biochem. Mol. Biol. 2018, 176, 4–15. [Google Scholar] [CrossRef] [PubMed]
  30. Périan, S.; Vanacker, J.M. GPER as a Receptor for Endocrine-Disrupting Chemicals (EDCs). Front. Endocrinol. 2020, 11, 545. [Google Scholar] [CrossRef] [PubMed]
  31. Kobayashi, H.; Gieniec, K.A.; Lannagan, T.R.M.; Wang, T.; Asai, N.; Mizutani, Y.; Iida, T.; Ando, R.; Thomas, E.M.; Sakai, A.; et al. The Origin and Contribution of Cancer-Associated Fibroblasts in Colorectal Carcinogenesis. Gastroenterology 2022, 162, 890–906. [Google Scholar] [CrossRef]
  32. Chuangchot, N.; Jamjuntra, P.; Yangngam, S.; Luangwattananun, P.; Thongchot, S.; Junking, M.; Thuwajit, P.; Yenchitsomanus, P.T.; Thuwajit, C. Enhancement of PD-L1-attenuated CAR-T cell function through breast cancer-associated fibroblasts-derived IL-6 signaling via STAT3/AKT pathways. Breast Cancer Res. 2023, 25, 86. [Google Scholar] [CrossRef]
  33. Shin, E.; Koo, J.S. Cell Component and Function of Tumor Microenvironment in Thyroid Cancer. Int. J. Mol. Sci. 2022, 23, 12578. [Google Scholar] [CrossRef] [PubMed]
  34. Hu, D.; Li, Z.; Zheng, B.; Lin, X.; Pan, Y.; Gong, P.; Zhuo, W.; Hu, Y.; Chen, C.; Chen, L.; et al. Cancer-associated fibroblasts in breast cancer: Challenges and opportunities. Cancer Commun. 2022, 42, 401–434. [Google Scholar] [CrossRef]
  35. Zhang, Y.; Zhong, F.; Liu, L. Single-cell transcriptional atlas of tumor-associated macrophages in breast cancer. Breast Cancer Res. 2024, 26, 129. [Google Scholar] [CrossRef]
  36. Gok Yavuz, B.; Gunaydin, G.; Gedik, M.E.; Kosemehmetoglu, K.; Karakoc, D.; Ozgur, F.; Guc, D. Cancer associated fibroblasts sculpt tumour microenvironment by recruiting monocytes and inducing immunosuppressive PD-1+ TAMs. Sci. Rep. 2019, 9, 3172. [Google Scholar] [CrossRef]
  37. Wang, Y.; Ma, C.; Li, X.; Yang, F.; Wang, N.; Ji, G.; Liu, Q.; Zhu, H.; Xu, S.; Li, H. Unraveling the role of M2 TAMs in ovarian cancer dynamics: A systematic review. J. Transl. Med. 2025, 23, 623. [Google Scholar] [CrossRef] [PubMed]
  38. Gao, Q.; Yang, Z.; Xu, S.; Li, X.; Yang, X.; Jin, P.; Liu, Y.; Zhou, X.; Zhang, T.; Gong, C.; et al. Heterotypic CAF-tumor spheroids promote early peritoneal metastatis of ovarian cancer. J. Exp. Med. 2019, 216, 688–703. [Google Scholar] [CrossRef]
  39. Cho, J.G.; Byeon, H.K.; Oh, K.H.; Baek, S.K.; Kwon, S.Y.; Jung, K.Y.; Woo, J.S. Clinicopathological significance of cancer-associated fibroblasts in papillary thyroid carcinoma: A predictive marker of cervical lymph node metastasis. Eur. Arch. Otorhinolaryngol. 2018, 275, 2355–2361. [Google Scholar] [CrossRef]
  40. Jaroszewski, A.; Geysels, R.C.; Volpini, X.; Pellizas, C.G.; Motran, C.C.; Stempin, C.C.; Nicola, J.P.; Cheng, S.Y.; Fozzatti, L. Anaplastic thyroid cancer cell-secreted TGFβ1 plays a key role in inducing macrophage polarization of human monocytes. Am. J. Cancer Res. 2024, 14, 3626–3638. [Google Scholar] [CrossRef] [PubMed]
  41. Bandyopadhyay, A.; Sinha, S.; Roy, R.; Biswas, N. Autophagy mediated immune response regulation and drug resistance in cancer. Mol. Biol. Rep. 2025, 52, 492. [Google Scholar] [CrossRef]
  42. Qin, Q.; Ji, H.; Li, D.; Zhang, H.; Zhang, Z.; Zhang, Q. Tumor-associated macrophages increase COX-2 expression promoting endocrine resistance in breast cancer via the PI3K/Akt/mTOR pathway. Neoplasma 2021, 68, 938–946. [Google Scholar] [CrossRef] [PubMed]
  43. Li, D.; Ji, H.; Niu, X.; Yin, L.; Wang, Y.; Gu, Y.; Wang, J.; Zhou, X.; Zhang, H.; Zhang, Q. Tumor-associated macrophages secrete CC-chemokine ligand 2 and induce tamoxifen resistance by activating PI3K/Akt/mTOR in breast cancer. Cancer Sci. 2020, 111, 47–58. [Google Scholar] [CrossRef] [PubMed]
  44. Febrero, B.; Ruiz-Manzanera, J.J.; Ros-Madrid, I.; Hernández, A.M.; Orenes-Piñero, E.; Rodríguez, J.M. Tumor microenvironment in thyroid cancer: Immune cells, patterns, and novel treatments. Head Neck. 2024, 46, 1486–1499. [Google Scholar] [CrossRef]
  45. Mirzaei, M.; Sheikholeslami, S.A.; Jalili, A.; Bereimipour, A.; Sharbati, S.; Kaveh, V.; Salari, S. Investigating the molecular mechanisms of Tamoxifen on the EMT pathway among patients with breast cancer. J. Med. Life 2022, 15, 835–844. [Google Scholar] [CrossRef]
  46. Hassn Mesrati, M.; Syafruddin, S.E.; Mohtar, M.A.; Syahir, A. CD44: A Multifunctional Mediator of Cancer Progression. Biomolecules 2021, 11, 1850. [Google Scholar] [CrossRef]
  47. Soliman, F.; Ye, L.; Jiang, W.; Hargest, R. Targeting Hyaluronic Acid and Peritoneal Dissemination in Colorectal Cancer. Clin. Color. Cancer. 2022, 21, e126–e134. [Google Scholar] [CrossRef]
  48. Affo, S.; Nair, A.; Brundu, F.; Ravichandra, A.; Bhattacharjee, S.; Matsuda, M.; Chin, L.; Filliol, A.; Wen, W.; Song, X.; et al. Promotion of cholangiocarcinoma growth by diverse cancer-associated fibroblast subpopulations. Cancer Cell 2021, 39, 866–882.e11. [Google Scholar] [CrossRef] [PubMed]
  49. Kellett-Clarke, H.; Stegmann, M.; Barclay, A.N.; Metcalfe, C. CD44 Binding to Hyaluronic Acid Is Redox Regulated by a Labile Disulfide Bond in the Hyaluronic Acid Binding Site. PLoS ONE 2015, 10, e0138137. [Google Scholar] [CrossRef]
  50. Hanoux, V.; Eguida, J.; Fleurot, E.; Levallet, J.; Bonnamy, P.J. Increase in hyaluronic acid degradation decreases the expression of estrogen receptor alpha in MCF7 breast cancer cell line. Mol. Cell. Endocrinol. 2018, 476, 185–197. [Google Scholar] [CrossRef]
  51. Trnkova, L.; Buocikova, V.; Mego, M.; Cumova, A.; Burikova, M.; Bohac, M.; Miklikova, S.; Cihova, M.; Smolkova, B. Epigenetic deregulation in breast cancer microenvironment: Implications for tumor progression and therapeutic strategies. Biomed. Pharmacother. 2024, 174, 116559. [Google Scholar] [CrossRef]
  52. Li, X.; Jin, Y.; Xue, J. Unveiling Collagen’s Role in Breast Cancer: Insights into Expression Patterns, Functions and Clinical Implications. Int. J. Gen. Med. 2024, 17, 1773–1787. [Google Scholar] [CrossRef]
  53. JingSong, H.; Hong, G.; Yang, J.; Duo, Z.; Li, F.; Wei Cai, C.; Xue Ying, L.; You Sheng, M.; Yi Wen, O.; Yue, P.; et al. siRNA-mediated suppression of collagen type iv alpha 2 (COL4A2) mRNA inhibits triple-negative breast cancer cell proliferation and migration. Oncotarget 2017, 8, 2585–2593. [Google Scholar] [CrossRef] [PubMed]
  54. Pellegrino, A.; Tiidus, P.M.; Vandenboom, R. Mechanisms of Estrogen Influence on Skeletal Muscle: Mass, Regeneration, and Mitochondrial Function. Sports Med. 2022, 52, 2853–2869. [Google Scholar] [CrossRef]
  55. Inoue, C.; Miki, Y.; Suzuki, T. New Perspectives on Sex Steroid Hormones Signaling in Cancer-Associated Fibroblasts of Non-Small Cell Lung Cancer. Cancers 2023, 15, 3620. [Google Scholar] [CrossRef] [PubMed]
  56. Reid, S.E.; Pantaleo, J.; Bolivar, P.; Bocci, M.; Sjölund, J.; Morsing, M.; Cordero, E.; Larsson, S.; Malmberg, M.; Seashore-Ludlow, B.; et al. Cancer-associated fibroblasts rewire the estrogen receptor response in luminal breast cancer, enabling estrogen independence. Oncogene 2024, 43, 1113–1126. [Google Scholar] [CrossRef] [PubMed]
  57. Sun, P.; Meng, L.H. Emerging roles of class I PI3K inhibitors in modulating tumor microenvironment and immunity. Acta Pharmacol. Sin. 2020, 41, 1395–1402. [Google Scholar] [CrossRef]
  58. Fabris, L.; Sato, K.; Alpini, G.; Strazzabosco, M. The Tumor Microenvironment in Cholangiocarcinoma Progression. Hepatology 2021, 73 (Suppl. S1), 75–85. [Google Scholar] [CrossRef]
  59. Basak, U.; Sarkar, T.; Mukherjee, S.; Chakraborty, S.; Dutta, A.; Dutta, S.; Nayak, D.; Kaushik, S.; Das, T.; Sa, G. Tumor-associated macrophages: An effective player of the tumor microenvironment. Front. Immunol. 2023, 14, 1295257. [Google Scholar] [CrossRef]
  60. Swanton, C.; Bernard, E.; Abbosh, C.; André, F.; Auwerx, J.; Balmain, A.; Bar-Sagi, D.; Bernards, R.; Bullman, S.; DeGregori, J.; et al. Embracing cancer complexity: Hallmarks of systemic disease. Cell 2024, 187, 1589–1616. [Google Scholar] [CrossRef]
  61. Harris, M.A.; Savas, P.; Virassamy, B.; O’Malley, M.M.R.; Kay, J.; Mueller, S.N.; Mackay, L.K.; Salgado, R.; Loi, S. Towards targeting the breast cancer immune microenvironment. Nat. Rev. Cancer 2024, 24, 554–577. [Google Scholar] [CrossRef]
  62. Bae, W.J.; Kim, S.; Ahn, J.M.; Han, J.H.; Lee, D. Estrogen-responsive cancer-associated fibroblasts promote invasive property of gastric cancer in a paracrine manner via CD147 production. FASEB J. 2022, 36, e22597. [Google Scholar] [CrossRef]
  63. Cirillo, F.; Pellegrino, M.; Talia, M.; Perrotta, I.D.; Rigiracciolo, D.C.; Spinelli, A.; Scordamaglia, D.; Muglia, L.; Guzzi, R.; Miglietta, A.M.; et al. Estrogen receptor variant ERα46 and insulin receptor drive in primary breast cancer cells growth effects and interleukin 11 induction prompting the motility of cancer-associated fibroblasts. Clin. Transl. Med. 2021, 11, e516. [Google Scholar] [CrossRef]
  64. Nagel, A.; Popeda, M.; Muchlinska, A.; Sadej, R.; Szade, J.; Zielinski, J.; Skokowski, J.; Niemira, M.; Kretowski, A.; Markiewicz, A.; et al. ERα36-High Cancer-Associated Fibroblasts as an Unfavorable Factor in Triple-Negative Breast Cancer. Cancers 2022, 14, 2005. [Google Scholar] [CrossRef]
  65. He, C.; Peng, M.; Zeng, X.; Dong, H.; Sun, Z.; Xu, J.; Liu, M.; Liu, L.; Huang, Y.; Peng, Z.; et al. Microenvironmental G protein-coupled estrogen receptor-mediated glutamine metabolic coupling between cancer-associated fibroblasts and triple-negative breast cancer cells governs tumour progression. Clin. Transl. Med. 2024, 14, e70131. [Google Scholar] [CrossRef] [PubMed]
  66. Lv, B.; Wang, Y.; Ma, D.; Cheng, W.; Liu, J.; Yong, T.; Chen, H.; Wang, C. Immunotherapy: Reshape the Tumor Immune Microenvironment. Front. Immunol. 2022, 13, 844142. [Google Scholar] [CrossRef] [PubMed]
  67. Gao, J.; Liang, Y.; Wang, L. Shaping Polarization of Tumor-Associated Macrophages in Cancer Immunotherapy. Front. Immunol. 2022, 13, 888713. [Google Scholar] [CrossRef] [PubMed]
  68. Castellaro, A.M.; Rodriguez-Baili, M.C.; Di Tada, C.E.; Gil, G.A. Tumor-Associated Macrophages Induce Endocrine Therapy Resistance in ER+ Breast Cancer Cells. Cancers 2019, 11, 189. [Google Scholar] [CrossRef]
  69. Jayshree, R.S. The Immune Microenvironment in Human Papilloma Virus-Induced Cervical Lesions-Evidence for Estrogen as an Immunomodulator. Front. Cell. Infect. Microbiol. 2021, 11, 649815. [Google Scholar]
  70. Sharpe, M.A.; Baskin, D.S.; Jenson, A.V.; Baskin, A.M. Hijacking Sexual Immuno-Privilege in GBM-An Immuno-Evasion Strategy. Int. J. Mol. Sci. 2021, 22, 10983. [Google Scholar] [CrossRef]
  71. O’Leary, K.A.; Bates, A.M.; Jin, W.J.; Burkel, B.M.; Sriramaneni, R.N.; Emma, S.E.; Nystuen, E.J.; Sumiec, E.G.; Ponik, S.M.; Morris, Z.S.; et al. Estrogen receptor blockade and radiation therapy cooperate to enhance the response of immunologically cold ER+ breast cancer to immunotherapy. Breast Cancer Res. 2023, 25, 68. [Google Scholar] [CrossRef]
  72. Lasser, S.A.; Ozbay Kurt, F.G.; Arkhypov, I.; Utikal, J.; Umansky, V. Myeloid-derived suppressor cells in cancer and cancer therapy. Nat. Rev. Clin. Oncol. 2024, 21, 147–164. [Google Scholar] [CrossRef] [PubMed]
  73. Ma, Z.; Zhen, Y.; Hu, C.; Yi, H. Myeloid-Derived Suppressor Cell-Derived Arginase-1 Oppositely Modulates IL-17A and IL-17F Through the ESR/STAT3 Pathway During Colitis in Mice. Front. Immunol. 2020, 11, 687. [Google Scholar] [CrossRef] [PubMed]
  74. Kozasa, K.; Mabuchi, S.; Matsumoto, Y.; Kuroda, H.; Yokoi, E.; Komura, N.; Kawano, M.; Takahashi, R.; Sasano, T.; Shimura, K.; et al. Estrogen stimulates female cancer progression by inducing myeloid-derived suppressive cells: Investigations on pregnant and non-pregnant experimental models. Oncotarget 2019, 10, 1887–1902. [Google Scholar] [CrossRef]
  75. Mangani, S.; Piperigkou, Z.; Koletsis, N.E.; Ioannou, P.; Karamanos, N.K. Estrogen receptors and extracellular matrix: The critical interplay in cancer development and progression. FEBS J. 2025, 292, 1558–1572. [Google Scholar] [CrossRef] [PubMed]
  76. Xiang, Y.; Li, J.P.; Guo, W.; Wang, D.Q.; Yao, A.; Zhang, H.M.; Huang, F.; Li, H.H.; Dai, Z.T.; Zhang, Z.; et al. Novel interactions between ERα-36 and STAT3 mediate breast cancer cell migration. Cell Commun. Signal. 2019, 17, 93. [Google Scholar] [CrossRef]
  77. Piperigkou, Z.; Karamanos, N.K. Estrogen receptor-mediated targeting of the extracellular matrix network in cancer. Semin. Cancer Biol. 2020, 62, 116–124. [Google Scholar] [CrossRef]
  78. Barcus, C.E.; O’Leary, K.A.; Brockman, J.L.; Rugowski, D.E.; Liu, Y.; Garcia, N.; Yu, M.; Keely, P.J.; Eliceiri, K.W.; Schuler, L.A. Elevated collagen-I augments tumor progressive signals, intravasation and metastasis of prolactin-induced estrogen receptor alpha positive mammary tumor cells. Breast Cancer Res. 2017, 19, 9. [Google Scholar] [CrossRef]
  79. Filho, A.M.; Laversanne, M.; Ferlay, J.; Colombet, M.; Piñeros, M.; Znaor, A.; Parkin, D.M.; Soerjomataram, I.; Bray, F. The GLOBOCAN 2022 cancer estimates: Data sources, methods, and a snapshot of the cancer burden worldwide. Int. J. Cancer 2025, 156, 1336–1346. [Google Scholar] [CrossRef]
  80. Trabert, B.; Coburn, S.B.; Falk, R.T.; Manson, J.E.; Brinton, L.A.; Gass, M.L.; Kuller, L.H.; Rohan, T.E.; Pfeiffer, R.M.; Qi, L.; et al. Circulating estrogens and postmenopausal ovarian and endometrial cancer risk among current hormone users in the Women’s Health Initiative Observational Study. Cancer Causes Control 2019, 30, 1201–1211. [Google Scholar] [CrossRef]
  81. Li, H.; Liu, Y.; Wang, Y.; Zhao, X.; Qi, X. Hormone therapy for ovarian cancer: Emphasis on mechanisms and applications (Review). Oncol. Rep. 2021, 46, 223. [Google Scholar] [CrossRef]
  82. Smolarz, B.; Biernacka, K.; Łukasiewicz, H.; Samulak, D.; Piekarska, E.; Romanowicz, H.; Makowska, M. Ovarian Cancer-Epidemiology, Classification, Pathogenesis, Treatment, and Estrogen Receptors’ Molecular Backgrounds. Int. J. Mol. Sci. 2025, 26, 4611. [Google Scholar] [CrossRef]
  83. Salvati, A.; Gigantino, V.; Nassa, G.; Giurato, G.; Alexandrova, E.; Rizzo, F.; Tarallo, R.; Weisz, A. The Histone Methyltransferase DOT1L Is a Functional Component of Estrogen Receptor Alpha Signaling in Ovarian Cancer Cells. Cancers 2019, 11, 1720. [Google Scholar] [CrossRef] [PubMed]
  84. Matsumura, S.; Ohta, T.; Yamanouchi, K.; Liu, Z.; Sudo, T.; Kojimahara, T.; Seino, M.; Narumi, M.; Tsutsumi, S.; Takahashi, T.; et al. Activation of estrogen receptor α by estradiol and cisplatin induces platinum-resistance in ovarian cancer cells. Cancer Biol. Ther. 2017, 18, 730–739. [Google Scholar] [CrossRef] [PubMed]
  85. Bogush, T.A.; Basharina, A.A.; Bogush, E.A.; Ryabinina, O.M.; Tjulandina, A.S.; Tjulandin, S.A. Estrogen Receptors alpha and beta in Ovarian Cancer: Expression Level and Prognosis. Dokl. Biochem. Biophys. 2018, 482, 249–251. [Google Scholar] [CrossRef]
  86. Chauvin, S.; Cohen-Tannoudji, J.; Guigon, C.J. Estradiol Signaling at the Heart of Folliculogenesis: Its Potential Deregulation in Human Ovarian Pathologies. Int. J. Mol. Sci. 2022, 23, 512. [Google Scholar] [CrossRef]
  87. Andreinie, R.; Nita, S.; Saleh, I. Relationship of estrogen beta (ERβ) receptor genes polymorphism with epithelial ovarian cancer. J. Phys. 2019, 1246, 012005. [Google Scholar]
  88. Sajeev, A.; BharathwajChetty, B.; Manickasamy, M.K.; Alqahtani, M.S.; Abbas, M.; Shakibaei, M.; Sethi, G.; Ma, Z.; Kunnumakkara, A.B. Nuclear receptors in ovarian cancer: Changing paradigms in cancer therapeutics. Front. Oncol. 2024, 14, 1383939. [Google Scholar] [CrossRef]
  89. Reichenbach, J.; Fraungruber, P.; Mayr, D.; Buschmann, C.; Kraus, F.B.T.; Topalov, N.E.; Chelariu-Raicu, A.; Kolben, T.; Burges, A.; Mahner, S.; et al. Nuclear receptor co-repressor NCOR2 and its relation to GPER with prognostic impact in ovarian cancer. J. Cancer Res. Clin. Oncol. 2023, 149, 8719–8728. [Google Scholar] [CrossRef] [PubMed]
  90. Jung, J. Role of G Protein-Coupled Estrogen Receptor in Cancer Progression. Toxicol. Res. 2019, 35, 209–214. [Google Scholar] [CrossRef]
  91. Schüler-Toprak, S.; Skrzypczak, M.; Ignatov, T.; Ignatov, A.; Ortmann, O.; Treeck, O. G protein-coupled estrogen receptor 1 (GPER-1) and agonist G-1 inhibit growth of ovarian cancer cells by activation of anti-tumoral transcriptome responses: Impact of GPER-1 mRNA on survival. J. Cancer Res. Clin. Oncol. 2020, 146, 3175–3188. [Google Scholar] [CrossRef]
  92. Han, N.; Heublein, S.; Jeschke, U.; Kuhn, C.; Hester, A.; Czogalla, B.; Mahner, S.; Rottmann, M.; Mayr, D.; Schmoeckel, E.; et al. The G-Protein-Coupled Estrogen Receptor (GPER) Regulates Trimethylation of Histone H3 at Lysine 4 and Represses Migration and Proliferation of Ovarian Cancer Cells In Vitro. Cells 2021, 10, 619. [Google Scholar] [CrossRef]
  93. Toàn, N.M. Novel Molecular Classification of Breast Cancer with PET Imaging. Medicina 2024, 60, 2099. [Google Scholar] [CrossRef]
  94. Zhang, X. Molecular Classification of Breast Cancer: Relevance and Challenges. Arch. Pathol. Lab. Med. 2023, 147, 46–51. [Google Scholar] [CrossRef]
  95. Vang, A.; Salem, K.; Fowler, A.M. Progesterone Receptor Gene Polymorphisms and Breast Cancer Risk. Endocrinology 2023, 164, bqad020. [Google Scholar] [CrossRef]
  96. Gosain, R.; Pollock, Y.; Jain, D. Age-related Disparity: Breast Cancer in the Elderly. Curr. Oncol. Rep. 2016, 18, 69. [Google Scholar] [CrossRef] [PubMed]
  97. Giordano, S.H. Breast Cancer in Men. N. Engl. J. Med. 2018, 378, 2311–2320. [Google Scholar] [CrossRef] [PubMed]
  98. Fox, S.; Speirs, V.; Shaaban, A.M. Male breast cancer: An update. Virchows Arch. 2022, 480, 85–93. [Google Scholar] [CrossRef] [PubMed]
  99. Gao, Y.; Heller, S.L. Breast Cancer Screening in Men. J. Breast Imaging 2023, 5, 104–111. [Google Scholar] [CrossRef]
  100. Loibl, S.; Poortmans, P.; Morrow, M.; Denkert, C.; Curigliano, G. Breast cancer. Lancet 2021, 397, 1750–1769. [Google Scholar] [CrossRef]
  101. Orrantia-Borunda, E.; Anchondo-Nuñez, P.; Acuña-Aguilar, L.E.; Gómez-Valles, F.O.; Ramírez-Valdespino, C.A. Subtypes of Breast Cancer. In Breast Cancer; Mayrovitz, H.N., Ed.; Exon Publications: Brisbane, Australia, 2022. [Google Scholar]
  102. Tecalco-Cruz, A.C.; Ramírez-Jarquín, J.O.; Cruz-Ramos, E. Estrogen Receptor Alpha and its Ubiquitination in Breast Cancer Cells. Curr. Drug Targets 2019, 20, 690–704. [Google Scholar] [CrossRef]
  103. Rugo, H.S.; Rumble, R.B.; Macrae, E.; Barton, D.L.; Connolly, H.K.; Dickler, M.N.; Fallowfield, L.; Fowble, B.; Ingle, J.N.; Jahanzeb, M.; et al. Endocrine Therapy for Hormone Receptor-Positive Metastatic Breast Cancer: American Society of Clinical Oncology Guideline. J. Clin. Oncol. 2016, 34, 3069–3103. [Google Scholar] [CrossRef] [PubMed]
  104. Porras, L.; Ismail, H.; Mader, S. Positive Regulation of Estrogen Receptor Alpha in Breast Tumorigenesis. Cells 2021, 10, 2966. [Google Scholar] [CrossRef]
  105. Clusan, L.; Ferrière, F.; Flouriot, G.; Pakdel, F. A Basic Review on Estrogen Receptor Signaling Pathways in Breast Cancer. Int. J. Mol. Sci. 2023, 24, 6834. [Google Scholar] [CrossRef]
  106. Pagano, M.T.; Ortona, E.; Dupuis, M.L. A Role for Estrogen Receptor alpha36 in Cancer Progression. Front. Endocrinol. 2020, 11, 506. [Google Scholar] [CrossRef]
  107. Maczis, M.A.; Maceyka, M.; Waters, M.R.; Newton, J.; Singh, M.; Rigsby, M.F.; Turner, T.H.; Alzubi, M.A.; Harrell, J.C.; Milstien, S.; et al. Sphingosine kinase 1 activation by estrogen receptor α36 contributes to tamoxifen resistance in breast cancer. J. Lipid Res. 2018, 59, 2297–2307. [Google Scholar] [CrossRef]
  108. Clusan, L.; Le Goff, P.; Flouriot, G.; Pakdel, F. A Closer Look at Estrogen Receptor Mutations in Breast Cancer and Their Implications for Estrogen and Antiestrogen Responses. Int. J. Mol. Sci. 2021, 22, 756. [Google Scholar] [CrossRef]
  109. Wu, W.; Warner, M.; Wang, L.; He, W.W.; Zhao, R.; Guan, X.; Botero, C.; Huang, B.; Ion, C.; Coombes, C.; et al. Drivers and suppressors of triple-negative breast cancer. Proc. Natl. Acad. Sci. USA 2021, 118, e2104162118. [Google Scholar] [CrossRef]
  110. Choi, Y. Estrogen Receptor β Expression and Its Clinical Implication in Breast Cancers: Favorable or Unfavorable? J. Breast Cancer 2022, 25, 75–93. [Google Scholar] [CrossRef] [PubMed]
  111. Xu, T.; Ma, D.; Chen, S.; Tang, R.; Yang, J.; Meng, C.; Feng, Y.; Liu, L.; Wang, J.; Luo, H.; et al. High GPER expression in triple-negative breast cancer is linked to pro-metastatic pathways and predicts poor patient outcomes. npj Breast Cancer 2022, 8, 100. [Google Scholar] [CrossRef] [PubMed]
  112. Cirillo, F.; Talia, M.; Santolla, M.F.; Pellegrino, M.; Scordamaglia, D.; Spinelli, A.; De Rosis, S.; Giordano, F.; Muglia, L.; Zicarelli, A.; et al. GPER deletion triggers inhibitory effects in triple negative breast cancer (TNBC) cells through the JNK/c-Jun/p53/Noxa transduction pathway. Cell Death Discov. 2023, 9, 353. [Google Scholar] [CrossRef]
  113. Chen, Z.J.; Wei, W.; Jiang, G.M.; Liu, H.; Wei, W.D.; Yang, X.; Wu, Y.M.; Liu, H.; Wong, C.K.; Du, J.; et al. Activation of GPER suppresses epithelial mesenchymal transition of triple negative breast cancer cells via NF-κB signals. Mol. Oncol. 2016, 10, 775–788. [Google Scholar] [CrossRef]
  114. Kampa, M.; Lappano, R.; Grande, F.; Rizzuti, B.; Maggiolini, M.; Castanas, E.; Jacquot, Y. Promising Perspectives of the Antiproliferative GPER Inverse Agonist ERα17p in Breast Cancer. Cells 2023, 12, 653. [Google Scholar] [CrossRef]
  115. Lappano, R.; Talia, M.; Cirillo, F.; Rigiracciolo, D.C.; Scordamaglia, D.; Guzzi, R.; Miglietta, A.M.; De Francesco, E.M.; Belfiore, A.; Sims, A.H.; et al. The IL1β-IL1R signaling is involved in the stimulatory effects triggered by hypoxia in breast cancer cells and cancer-associated fibroblasts (CAFs). J. Exp. Clin. Cancer Res. 2020, 39, 153. [Google Scholar] [CrossRef] [PubMed]
  116. Ramírez-López, I.G.; Ramírez de Arellano, A.; Jave-Suárez, L.F.; Hernández-Silva, C.D.; García-Chagollan, M.; Hernández-Bello, J.; Lopez-Pulido, E.I.; Macias-Barragan, J.; Montoya-Buelna, M.; Muñoz-Valle, J.F.; et al. Interaction between 17β-estradiol, prolactin and human papillomavirus induce E6/E7 transcript and modulate the expression and localization of hormonal receptors. Cancer Cell Int. 2019, 19, 227. [Google Scholar] [CrossRef] [PubMed]
  117. Martin, S.G.; Lebot, M.N.; Sukkarn, B.; Ball, G.; Green, A.R.; Rakha, E.A.; Ellis, I.O.; Storr, S.J. Low expression of G protein-coupled oestrogen receptor 1 (GPER) is associated with adverse survival of breast cancer patients. Oncotarget 2018, 9, 25946–25956. [Google Scholar] [CrossRef]
  118. Li, W.; Liang, H.; Wang, W.; Liu, J.; Liu, X.; Lao, S.; Liang, W.; He, J. Global cancer statistics for adolescents and young adults: Population based study. J. Hematol. Oncol. 2024, 17, 99. [Google Scholar] [CrossRef] [PubMed]
  119. Pizzato, M.; Li, M.; Vignat, J.; Laversanne, M.; Singh, D.; La Vecchia, C.; Vaccarella, S. The epidemiological landscape of thyroid cancer worldwide: GLOBOCAN estimates for incidence and mortality rates in 2020. Lancet Diabetes Endocrinol. 2022, 10, 264–272. [Google Scholar] [CrossRef]
  120. Liu, J.; Xu, T.; Ma, L.; Chang, W. Signal Pathway of Estrogen and Estrogen Receptor in the Development of Thyroid Cancer. Front. Oncol. 2021, 11, 593479. [Google Scholar] [CrossRef]
  121. Wang, H.; Li, S.; Shi, J.; Feng, C.; Wang, Y.; Zhang, F. Unbalanced bidirectional causal association between thyroid cancer and ER-positive breast cancer: Should we recommend screening for thyroid cancer in breast cancer patients? BMC Genom. 2023, 24, 762. [Google Scholar] [CrossRef]
  122. Qiu, Y.B.; Liao, L.Y.; Jiang, R.; Xu, M.; Xu, L.W.; Chen, G.G.; Liu, Z.M. PES1 promotes the occurrence and development of papillary thyroid cancer by upregulating the ERα/ERβ protein ratio. Sci. Rep. 2019, 9, 1032. [Google Scholar] [CrossRef]
  123. Xu, L.W.; Gou, X.; Yang, J.Y.; Jiang, R.; Jiang, X.; Chen, G.G.; Liu, Z.M. Methylation of ERβ 5′-untranslated region attenuates its inhibitory effect on ERα gene transcription and promotes the initiation and progression of papillary thyroid cancer. FASEB J. 2021, 35, e21516. [Google Scholar] [CrossRef] [PubMed]
  124. Rubio, G.A.; Catanuto, P.; Glassberg, M.K.; Lew, J.I.; Elliot, S.J. Estrogen receptor subtype expression and regulation is altered in papillary thyroid cancer after menopause. Surgery 2018, 163, 143–149. [Google Scholar] [CrossRef] [PubMed]
  125. Dong, W.; Zhang, H.; Li, J.; Guan, H.; He, L.; Wang, Z.; Shan, Z.; Teng, W. Estrogen Induces Metastatic Potential of Papillary Thyroid Cancer Cells through Estrogen Receptor α and β. Int. J. Endocrinol. 2013, 2013, 941568. [Google Scholar] [CrossRef] [PubMed]
  126. Meng, D.; Li, Z.; Ma, X.; Wu, L.; Fu, L.; Qin, G. ETV5 overexpression contributes to tumor growth and progression of thyroid cancer through PIK3CA. Life Sci. 2020, 253, 117693. [Google Scholar] [CrossRef]
  127. Chou, C.K.; Chi, S.Y.; Hung, Y.Y.; Yang, Y.C.; Fu, H.C.; Wang, J.H.; Chen, C.C.; Kang, H.Y. Decreased Expression of Estrogen Receptors Is Associated with Tumorigenesis in Papillary Thyroid Carcinoma. Int. J. Mol. Sci. 2022, 23, 1015. [Google Scholar] [CrossRef]
  128. Ahn, H.Y.; Song, R.Y.; Ahn, H.S.; Kim, H.S. Expression of Estrogen and Progesterone Receptors in Papillary Thyroid Carcinoma in Korea. Cancer Res. Treat. 2021, 53, 1204–1212. [Google Scholar] [CrossRef]
  129. Manfroi, P.A.; Bertoni, A.P.S.; Furlanetto, T.W. GPER1 in the thyroid: A systematic review. Life Sci. 2020, 241, 117112. [Google Scholar] [CrossRef]
  130. Bertoni, A.P.S.; Manfroi, P.A.; Tomedi, J.; Assis-Brasil, B.M.; de Souza Meyer, E.L.; Furlanetto, T.W. The gene expression of GPER1 is low in fresh samples of papillary thyroid carcinoma (PTC), and in silico analysis. Mol. Cell. Endocrinol. 2021, 535, 111397. [Google Scholar] [CrossRef]
  131. Laha, D.; Nilubol, N.; Boufraqech, M. New Therapies for Advanced Thyroid Cancer. Front. Endocrinol. 2020, 11, 82. [Google Scholar] [CrossRef]
  132. Yuan, Y.; Sun, P.; Xiao, H.; Li, L.; Li, J.; Ai, X. Risk of second primary thyroid cancer in cancer survivors. Sci. Rep. 2024, 14, 12478. [Google Scholar] [CrossRef]
  133. Forma, A.; Kłodnicka, K.; Pająk, W.; Flieger, J.; Teresińska, B.; Januszewski, J.; Baj, J. Thyroid Cancer: Epidemiology, Classification, Risk Factors, Diagnostic and Prognostic Markers, and Current Treatment Strategies. Int. J. Mol. Sci. 2025, 26, 5173. [Google Scholar] [CrossRef]
  134. Wu, K.; Chen, Y.; Guo, R.; Zeng, Q.; Yu, Y. Leptin and insulin synergize with PIK3CA mutation to enhance PD-L1 mediated immunosuppression in thyroid cancer. Exp. Cell Res. 2024, 442, 114229. [Google Scholar] [CrossRef]
  135. Abancens, M.; Bustos, V.; Harvey, H.; McBryan, J.; Harvey, B.J. Sexual Dimorphism in Colon Cancer. Front. Oncol. 2020, 10, 607909. [Google Scholar] [CrossRef]
  136. Siegel, R.L.; Wagle, N.S.; Cercek, A.; Smith, R.A.; Jemal, A. Colorectal cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 233–254. [Google Scholar] [CrossRef] [PubMed]
  137. Refaat, B.; Aslam, A.; Idris, S.; Almalki, A.H.; Alkhaldi, M.Y.; Asiri, H.A.; Almaimani, R.A.; Mujalli, A.; Minshawi, F.; Alamri, S.A.; et al. Profiling estrogen, progesterone, and androgen receptors in colorectal cancer in relation to gender, menopausal status, clinical stage, and tumour sidedness. Front. Endocrinol. 2023, 14, 1187259. [Google Scholar] [CrossRef] [PubMed]
  138. Maingi, J.W.; Tang, S.; Liu, S.; Ngenya, W.; Bao, E. Targeting estrogen receptors in colorectal cancer. Mol. Biol. Rep. 2020, 47, 4087–4091. [Google Scholar] [CrossRef]
  139. Ditonno, I.; Losurdo, G.; Rendina, M.; Pricci, M.; Girardi, B.; Ierardi, E.; Di Leo, A. Estrogen Receptors in Colorectal Cancer: Facts, Novelties and Perspectives. Curr. Oncol. 2021, 28, 4256–4263. [Google Scholar] [CrossRef]
  140. Topi, G.; Ghatak, S.; Satapathy, S.R.; Ehrnström, R.; Lydrup, M.L.; Sjölander, A. Combined Estrogen Alpha and Beta Receptor Expression Has a Prognostic Significance for Colorectal Cancer Patients. Front. Med. 2022, 9, 739620. [Google Scholar] [CrossRef] [PubMed]
  141. Topi, G.; Satapathy, S.R.; Ghatak, S.; Hellman, K.; Ek, F.; Olsson, R.; Ehrnström, R.; Lydrup, M.L.; Sjölander, A. High Oestrogen receptor alpha expression correlates with adverse prognosis and promotes metastasis in colorectal cancer. Cell Commun. Signal. 2024, 22, 198. [Google Scholar] [CrossRef]
  142. Hases, L.; Indukuri, R.; Birgersson, M.; Nguyen-Vu, T.; Lozano, R.; Saxena, A.; Hartman, J.; Frasor, J.; Gustafsson, J.Å.; Katajisto, P.; et al. Intestinal estrogen receptor beta suppresses colon inflammation and tumorigenesis in both sexes. Cancer Lett. 2020, 492, 54–62. [Google Scholar] [CrossRef]
  143. Indukuri, R.; Hases, L.; Archer, A.; Williams, C. Estrogen Receptor Beta Influences the Inflammatory p65 Cistrome in Colon Cancer Cells. Front. Endocrinol. 2021, 12, 650625. [Google Scholar] [CrossRef] [PubMed]
  144. Božović, A.; Mandušić, V.; Todorović, L.; Krajnović, M. Estrogen Receptor Beta: The Promising Biomarker and Potential Target in Metastases. Int. J. Mol. Sci. 2021, 22, 1656. [Google Scholar] [CrossRef] [PubMed]
  145. Lu, L.; Zhang, Q.; Shen, X.; Zhen, P.; Marin, A.; Garcia-Milian, R.; Roper, J.; Khan, S.A.; Johnson, C.H. Asparagine synthetase and G-protein coupled estrogen receptor are critical responders to nutrient supply in KRAS mutant colorectal cancer. bioRxiv 2023. [Google Scholar] [CrossRef]
  146. Qiu, Y.A.; Xiong, J.; Yu, T. Role of G Protein-Coupled Estrogen Receptor in Digestive System Carcinomas: A Minireview. OncoTargets Ther. 2021, 14, 2611–2622. [Google Scholar] [CrossRef]
  147. Abancens, M.; Harvey, B.J.; McBryan, J. GPER Agonist G1 Prevents Wnt-Induced JUN Upregulation in HT29 Colorectal Cancer Cells. Int. J. Mol. Sci. 2022, 23, 12581. [Google Scholar] [CrossRef]
  148. Ionescu, V.A.; Gheorghe, G.; Bacalbasa, N.; Chiotoroiu, A.L.; Diaconu, C. Colorectal Cancer: From Risk Factors to Oncogenesis. Medicina 2023, 59, 1646. [Google Scholar] [CrossRef]
  149. Sninsky, J.A.; Shore, B.M.; Lupu, G.V.; Crockett, S.D. Risk Factors for Colorectal Polyps and Cancer. Gastrointest. Endosc. Clin. N. Am. 2022, 32, 195–213. [Google Scholar] [CrossRef] [PubMed]
  150. Kuipers, E.J.; Grady, W.M.; Lieberman, D.; Seufferlein, T.; Sung, J.J.; Boelens, P.G.; van de Velde, C.J.; Watanabe, T. Colorectal cancer. Nat. Rev. Dis. Primers 2015, 1, 15065. [Google Scholar] [CrossRef]
  151. Hormone Therapy for Brest Cancer. Available online: https://www.cancer.gov/types/breast/breast-hormone-therapy-fact-sheet (accessed on 22 September 2025).
  152. Lobo-Martins, S.; Arecco, L.; Cabral, T.P.; Agostinetto, E.; Dauccia, C.; Franzoi, M.A.; Del Mastro, L.; Lambertini, M.; Piccart, M.; de Azambuja, E. Extended adjuvant endocrine therapy in early breast cancer: Finding the individual balance. ESMO Open 2025, 10, 105057. [Google Scholar] [CrossRef]
  153. Homewood, D.; Fu, M.H.; Sathianathen, N.; La Bianca, S.; Tran, B.; Corcoran, N.M. Evolution of hormonal therapy for prostate cancer. Aust. J. Gen. Pract. 2024, 53, 291–300. [Google Scholar] [CrossRef]
  154. Mitra, S.; Lami, M.S.; Ghosh, A.; Das, R.; Tallei, T.E.; Fatimawali Islam, F.; Dhama, K.; Begum, M.Y.; Aldahish, A.; Chidambaram, K.; et al. Hormonal Therapy for Gynecological Cancers: How Far Has Science Progressed toward Clinical Applications? Cancers 2022, 14, 759. [Google Scholar] [CrossRef] [PubMed]
  155. Biondi, B.; Filetti, S.; Schlumberger, M. Thyroid-hormone therapy and thyroid cancer: A reassessment. Nat. Clin. Pract. Endocrinol. Metab. 2005, 1, 32–40. [Google Scholar] [CrossRef] [PubMed]
  156. Jin, P.; Duan, X.; Huang, Z.; Dong, Y.; Zhu, J.; Guo, H.; Tian, H.; Zou, C.G.; Xie, K. Nuclear receptors in health and disease: Signaling pathways, biological functions and pharmaceutical interventions. Signal Transduct. Target. Ther. 2025, 10, 228. [Google Scholar] [CrossRef] [PubMed]
  157. Pickar, J.H.; MacNeil, T.; Ohleth, K. SERMs: Progress and future perspectives. Maturitas 2010, 67, 129–138. [Google Scholar] [CrossRef]
  158. Mirkin, S.; Pickar, J.H. Selective estrogen receptor modulators (SERMs): A review of clinical data. Maturitas 2015, 80, 52–57. [Google Scholar] [CrossRef]
  159. Pickar, J.H.; Boucher, M.; Morgenstern, D. Tissue selective estrogen complex (TSEC): A review. Menopause 2018, 25, 1033–1045. [Google Scholar] [CrossRef]
  160. Lumachi, F.; Santeufemia, D.A.; Basso, S.M. Current medical treatment of estrogen receptor-positive breast cancer. World J. Biol. Chem. 2015, 6, 231–239. [Google Scholar] [CrossRef]
  161. Zhang, Y.; Zhao, L.; Liu, Y.; Zhang, J.; Zheng, L.; Zheng, M. Adverse Event Profiles of the Third-Generation Aromatase Inhibitors: Analysis of Spontaneous Reports Submitted to FAERS. Biomedicines 2024, 12, 1708. [Google Scholar] [CrossRef]
  162. Davies, C.; Pan, H.; Godwin, J.; Gray, R.; Arriagada, R.; Raina, V.; Abraham, M.; Medeiros Alencar, V.H.; Badran, A.; Bonfill, X.; et al. Adjuvant Tamoxifen: Longer Against Shorter (ATLAS) Collaborative Group. Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet 2013, 381, 805–816. [Google Scholar] [CrossRef]
  163. Available online: https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=2817 (accessed on 22 September 2025).
  164. Maximov, P.Y.; McDaniel, R.E.; Fernandes, D.J.; Korostyshevskiy, V.R.; Bhatta, P.; Mürdter, T.E.; Flockhart, D.A.; Jordan, V.C. Simulation with cells in vitro of tamoxifen treatment in premenopausal breast cancer patients with different CYP2D6 genotypes. Br. J. Pharmacol. 2014, 171, 5624–5635. [Google Scholar] [CrossRef]
  165. Rahmawati, D.R.; Nurrochmad, A.; Meiyanto, E.; Jenie, R.I. Persistent synergistic antiproliferative and apoptosis induction effects of PGV-1 and tamoxifen or 4-hydroxytamoxifen on ER-positive breast cancer cells. J. Appl. Pharm. Sci. 2025, 15, 205–215. [Google Scholar] [CrossRef]
  166. Martinkovich, S.; Shah, D.; Planey, S.L.; Arnott, J.A. Selective estrogen receptor modulators: Tissue specificity and clinical utility. Clin. Interv. Aging 2014, 9, 1437–1452. [Google Scholar] [CrossRef] [PubMed]
  167. Brufsky, A.M. Understanding the estrogen receptor signaling pathway: Focus on current endocrine agents for breast cancer in postmenopausal women. Community Oncol. 2011, 8, 343–352. [Google Scholar] [CrossRef]
  168. Wong, M.M.; Guo, C.; Zhang, J. Nuclear receptor corepressor complexes in cancer: Mechanism, function and regulation. Am. J. Clin. Exp. Urol. 2014, 2, 169–187. [Google Scholar]
  169. Shang, Y.; Brown, M. Molecular determinants for the tissue specificity of SERMs. Science 2002, 295, 2465–2468. [Google Scholar] [CrossRef]
  170. Ali, S.; Rasool, M.; Chaoudhry, H.; NPushparaj, P.; Jha, P.; Hafiz, A.; Mahfooz, M.; Abdus Sami, G.; Azhar Kamal, M.; Bashir, S.; et al. Molecular mechanisms and mode of tamoxifen resistance in breast cancer. Bioinformation 2016, 12, 135–139. [Google Scholar] [CrossRef]
  171. Légaré, S.; Basik, M. Minireview: The Link Between ERα Corepressors and Histone Deacetylases in Tamoxifen Resistance in Breast Cancer. Mol. Endocrinol. 2016, 30, 965–976. [Google Scholar] [CrossRef]
  172. Ghanavati, M.; Khorshidi, Y.; Shadnoush, M.; Akbari, M.E.; Ardehali, S.H.; Chavarri-Guerra, Y.; Akbari, A.; Barragan-Carrillo, R.; Amin Amlashi, M.; Javid, Z.; et al. Tamoxifen use and risk of endometrial cancer in breast cancer patients: A systematic review and dose-response meta-analysis. Cancer Rep. 2023, 6, e1806. [Google Scholar] [CrossRef]
  173. Mustonen, M.V.; Pyrhönen, S.; Kellokumpu-Lehtinen, P.L. Toremifene in the treatment of breast cancer. World J. Clin. Oncol. 2014, 5, 393–405. [Google Scholar] [CrossRef]
  174. Berthou, F.; Dreano, Y.; Belloc, C.; Kangas, L.; Gautier, J.C.; Beaune, P. Involvement of cytochrome P450 3A enzyme family in the major metabolic pathways of toremifene in human liver microsomes. Biochem. Pharmacol. 1994, 47, 1883–1895. [Google Scholar] [CrossRef]
  175. Kim, J.; Coss, C.C.; Barrett, C.M.; Mohler, M.L.; Bohl, C.E.; Li, C.M.; He, Y.; Veverka, K.A.; Dalton, J.T. Role and pharmacologic significance of cytochrome P-450 2D6 in oxidative metabolism of toremifene and tamoxifen. Int. J. Cancer 2013, 132, 1475–1485. [Google Scholar] [CrossRef]
  176. Wiseman, L.R.; Goa, K. L Toremifene: A review of its pharmacological properties and clinical efficacy in the management of advanced breast cancer. Drugs 1997, 54, 141–160. [Google Scholar] [CrossRef]
  177. Kangas, L. Biochemical and pharmacological effects of toremifene metabolites. Cancer Chemother. Pharmacol. 1990, 27, 8–12. [Google Scholar] [CrossRef] [PubMed]
  178. Provinciali, N.; Suen, C.; Dunn, B.K.; DeCensi, A. Raloxifene hydrochloride for breast cancer risk reduction in postmenopausal women. Expert. Rev. Clin. Pharmacol. 2016, 9, 1263–1272. [Google Scholar] [CrossRef] [PubMed]
  179. Sun, D.; Jones, N.R.; Manni, A.; Lazarus, P. Characterization of raloxifene glucuronidation: Potential role of UGT1A8 genotype on raloxifene metabolism in vivo. Cancer Prev. Res. 2013, 6, 719–730. [Google Scholar] [CrossRef] [PubMed]
  180. Dodge, J.A.; Lugar, C.W.; Cho, S.; Short, L.L.; Sato, M.; Yang, N.N.; Spangle, L.A.; Martin, M.J.; Phillips, D.L.; Glasebrook, A.L.; et al. Evaluation of the major metabolites of raloxifene as modulators of tissue selectivity. J. Steroid Biochem. Mol. Biol. 1997, 61, 97–106. [Google Scholar] [CrossRef]
  181. Bryant, H.U.; Dere, W.H. Selective estrogen receptor modulators: An alternative to hormone replacement therapy. Proc. Soc. Exp. Biol. Med. 1998, 217, 45–52. [Google Scholar] [CrossRef]
  182. Snyder, K.R.; Sparano, N.; Malinowski, J.M. Raloxifene hydrochloride. Am. J. Health Syst. Pharm. 2000, 57, 1669–1678. [Google Scholar] [CrossRef]
  183. Rey, J.R.; Cervino, E.V.; Rentero, M.L.; Crespo, E.C.; Alvaro, A.O.; Casillas, M. Raloxifene: Mechanism of action, effects on bone tissue, and applicability in clinical traumatology practice. Open Orthop. J. 2009, 3, 14–21. [Google Scholar] [CrossRef]
  184. Yang, N.N.; Venugopalan, M.; Hardikar, S.; Glasebrook, A. Identification of an estrogen response element activated by metabolites of 17beta-estradiol and raloxifene. Science 1996, 273, 1222–1225. [Google Scholar] [CrossRef]
  185. Han, K.O.; Kang, Y.S.; Hwang, C.S.; Moon, I.G.; Yim, C.H.; Chung, H.Y.; Jang, H.C.; Yoon, H.K.; Han, I.K.; Choi, Y.K. Identification of a mutation in the human raloxifene response element of the transforming growth factor-beta 3 gene. J. Korean Med. Sci. 2001, 16, 549–552. [Google Scholar] [CrossRef]
  186. Curran, M.; Wiseman, L. Fulvestrant. Drugs 2001, 61, 807–814. [Google Scholar] [CrossRef]
  187. Kim, N.; Lukong, K.E. Treating ER-positive breast cancer: A review of the current FDA-approved SERMs and SERDs and their mechanisms of action. Oncol. Rev. 2025, 19, 1564642. [Google Scholar] [CrossRef]
  188. Robertson, J.F.; Harrison, M. Fulvestrant: Pharmacokinetics and pharmacology. Br. J. Cancer 2004, 90 (Suppl. S1), S7–S10. [Google Scholar] [CrossRef]
  189. Carlson, R.W. The history and mechanism of action of fulvestrant. Clin. Breast Cancer. 2005, 6 (Suppl. S1), S5–S8. [Google Scholar] [CrossRef] [PubMed]
  190. Nicholson, R.I.; Gee, J.M.; Manning, D.L.; Wakeling, A.E.; Montano, M.M.; Katzenellenbogen, B.S. Responses to pure antiestrogens (ICI 164384, ICI 182780) in estrogen-sensitive and -resistant experimental and clinical breast cancer. Ann. N. Y. Acad. Sci. 1995, 761, 148–163. [Google Scholar] [CrossRef] [PubMed]
  191. Long, X.; Nephew, K.P. Fulvestrant (ICI 182,780)-dependent interacting proteins mediate immobilization and degradation of estrogen receptor-alpha. J. Biol. Chem. 2006, 281, 9607–9615. [Google Scholar] [CrossRef]
  192. Chumsri, S.; Howes, T.; Bao, T.; Sabnis, G.; Brodie, A. Aromatase, aromatase inhibitors, and breast cancer. J. Steroid Biochem. Mol. Biol. 2011, 125, 13–22. [Google Scholar] [CrossRef]
  193. Hong, Y.; Chen, S. Aromatase inhibitors: Structural features and biochemical characterization. Ann. N. Y. Acad. Sci. 2006, 1089, 237–251. [Google Scholar] [CrossRef]
  194. Bhutani, K.; Vishwakarma, S.; Yadav, P.; Yadav, M.K. The current landscape of aromatase inhibitors for the treatment of estrogen receptor-positive breast carcinoma. J. Steroid Biochem. Mol. Biol. 2025, 250, 106729. [Google Scholar] [CrossRef] [PubMed]
  195. Moharrer Navaei, N.; Moharrer Navaei, N. Unlocking the Potential of Aromatase Inhibitors: Recent Advances in Drug Design, Synthesis, Docking Activity, and in Vitro Bioactivity Evaluations. Synth. Sinter. 2023, 3, 234–240. [Google Scholar] [CrossRef]
  196. Sinn, H.P.; Kreipe, H. A Brief Overview of the WHO Classification of Breast Tumors, 4th Edition, Focusing on Issues and Updates from the 3rd Edition. Breast Care 2013, 8, 149–154. [Google Scholar] [CrossRef] [PubMed]
  197. Guo, L.; Kong, D.; Liu, J.; Zhan, L.; Luo, L.; Zheng, W.; Zheng, Q.; Chen, C.; Sun, S. Breast cancer heterogeneity and its implication in personalized precision therapy. Exp. Hematol. Oncol. 2023, 12, 3. [Google Scholar] [CrossRef]
  198. Testa, U.; Castelli, G.; Pelosi, E. Breast Cancer: A Molecularly Heterogenous Disease Needing Subtype-Specific Treatments. Med. Sci. 2020, 8, 18. [Google Scholar] [CrossRef]
  199. Ferreira Almeida, C.; Correia-da-Silva, G.; Teixeira, N.; Amaral, C. Influence of tumor microenvironment on the different breast cancer subtypes and applied therapies. Biochem. Pharmacol. 2024, 223, 116178. [Google Scholar] [CrossRef]
  200. Hanahan, D.; Coussens, L.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef]
  201. Akinpelu, A.; Akinsipe, T.; Avila, L.A.; Arnold, R.D.; Mistriotis, P. The impact of tumor microenvironment: Unraveling the role of physical cues in breast cancer progression. Cancer Metastasis Rev. 2024, 43, 823–844. [Google Scholar] [CrossRef]
  202. Deepak, K.G.K.; Vempati, R.; Nagaraju, G.P.; Dasari, V.R.; Nagini, S.; Rao, D.N.; Malla, R.R. Tumor microenvironment: Challenges and opportunities in targeting metastasis of triple negative breast cancer. Pharmacol. Res. 2020, 153, 104683. [Google Scholar] [CrossRef] [PubMed]
  203. Dai, J.; Su, Y.; Zhong, S.; Cong, L.; Liu, B.; Yang, J.; Tao, Y.; He, Z.; Chen, C.; Jiang, Y. Exosomes: Key players in cancer and potential therapeutic strategy. Signal Transduct. Target. Ther. 2020, 5, 145. [Google Scholar] [CrossRef]
  204. Xie, F.; Zhou, X.; Fang, M.; Li, H.; Su, P.; Tu, Y.; Zhang, L.; Zhou, F. Extracellular Vesicles in Cancer Immune Microenvironment and Cancer Immunotherapy. Adv. Sci. 2019, 6, 1901779. [Google Scholar] [CrossRef] [PubMed]
  205. Liu, J.; Zhu, S.; Tang, W.; Huang, Q.; Mei, Y.; Yang, H. Exosomes from tamoxifen-resistant breast cancer cells transmit drug resistance partly by delivering miR-9-5p. Cancer Cell Int. 2021, 21, 55. [Google Scholar] [CrossRef]
  206. La Camera, G.; Gelsomino, L.; Caruso, A.; Panza, S.; Barone, I.; Bonofiglio, D.; Andò, S.; Giordano, C.; Catalano, S. The Emerging Role of Extracellular Vesicles in Endocrine Resistant Breast Cancer. Cancers 2021, 13, 1160. [Google Scholar] [CrossRef]
  207. Patel, R.; Klein, P.; Tiersten, A.; Sparano, J.A. An emerging generation of endocrine therapies in breast cancer: A clinical perspective. npj Breast Cancer 2023, 9, 20. [Google Scholar] [CrossRef]
  208. Hanker, A.B.; Sudhan, D.R.; Arteaga, C.L. Overcoming Endocrine Resistance in Breast Cancer. Cancer Cell 2020, 37, 496–513. [Google Scholar] [CrossRef] [PubMed]
  209. Cardoso, F.; Paluch-Shimon, S.; Senkus, E.; Curigliano, G.; Aapro, M.S.; André, F.; Barrios, C.H.; Bergh, J.; Bhattacharyya, G.S.; Biganzoli, L.; et al. 5th ESO-ESMO international consensus guidelines for advanced breast cancer (ABC 5). Ann. Oncol. 2020, 31, 1623–1649. [Google Scholar] [CrossRef]
  210. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Extending the duration of endocrine treatment for early breast cancer: Patient-level meta-analysis of 12 randomised trials of aromatase inhibitors in 22,031 postmenopausal women already treated with at least 5 years of endocrine therapy. Lancet 2025, 406, 603–614. [Google Scholar] [CrossRef]
  211. Eapen, J.V.; George, P.; Thomas, S.; Antony, J. Integrative Strategies in Breast Cancer Therapy: Conventional Approaches, Emerging Advances, and Future Challenges. ASPET Discov. 2025, 1, 100012. [Google Scholar] [CrossRef]
  212. Berry, D.A.; Muss, H.B.; Thor, A.D.; Dressler, L.; Liu, E.T.; Broadwater, G.; Budman, D.R.; Henderson, I.C.; Barcos, M.; Hayes, D.; et al. HER-2/neu and p53 expression versus tamoxifen resistance in estrogen receptor-positive, node-positive breast cancer. J. Clin. Oncol. 2000, 18, 3471–3479. [Google Scholar] [CrossRef] [PubMed]
  213. Parisot, J.P.; Hu, X.F.; DeLuise, M.; Zalcberg, J.R. Altered expression of the IGF-1 receptor in a tamoxifen-resistant human breast cancer cell line. Br. J. Cancer 1999, 79, 693–700. [Google Scholar] [CrossRef] [PubMed]
  214. Ojo, D.; Wei, F.; Liu, Y.; Wang, E.; Zhang, H.; Lin, X.; Wong, N.; Bane, A.; Tang, D. Factors Promoting Tamoxifen Resistance in Breast Cancer via Stimulating Breast Cancer Stem Cell Expansion. Curr. Med. Chem. 2015, 22, 2360–2374. [Google Scholar] [CrossRef] [PubMed]
  215. Pietras, R.J.; Arboleda, J.; Reese, D.M.; Wongvipat, N.; Pegram, M.D.; Ramos, L.; Gorman, C.M.; Parker, M.G.; Sliwkowski, M.X.; Slamon, D.J. HER-2 tyrosine kinase pathway targets estrogen receptor and promotes hormone-independent growth in human breast cancer cells. Oncogene 1995, 10, 2435–2446. [Google Scholar] [PubMed]
  216. Girault, I.; Bièche, I.; Lidereau, R. Role of estrogen receptor alpha transcriptional coregulators in tamoxifen resistance in breast cancer. Maturitas 2006, 54, 342–351. [Google Scholar] [CrossRef]
  217. Nass, N.; Kalinski, T. Tamoxifen resistance: From cell culture experiments towards novel biomarkers. Pathol. Res. Pract. 2015, 211, 189–197. [Google Scholar] [CrossRef]
  218. Miller, D.L.; Kern, F.G. Growth factor signal transduction and hormone independence in breast cancer. Adv. Oncobiol. 1999, 2, 11–80. [Google Scholar]
  219. Gago, F.E.; Fanelli, M.A.; Ciocca, D.R. Co-expression of steroid hormone receptors (estrogen receptor alpha and/or progesterone receptors) and Her2/neu (c-erbB-2) in breast cancer: Clinical outcome following tamoxifen-based adjuvant therapy. J. Steroid Biochem. Mol. Biol. 2006, 98, 36–40. [Google Scholar] [CrossRef] [PubMed]
  220. Arpino, G.; Wiechmann, L.; Osborne, C.K.; Schiff, R. Crosstalk between the estrogen receptor and the HER tyrosine kinase receptor family: Molecular mechanism and clinical implications for endocrine therapy resistance. Endocr. Rev. 2008, 29, 217–233. [Google Scholar] [CrossRef]
  221. Pietras, R.J.; Márquez-Garbán, D.C. Membrane-associated estrogen receptor signaling pathways in human cancers. Clin. Cancer Res. 2007, 13, 4672–4676. [Google Scholar] [CrossRef] [PubMed]
  222. Yang, X.; Phillips, D.L.; Ferguson, A.T.; Nelson, W.G.; Herman, J.G.; Davidson, N.E. Synergistic activation of functional estrogen receptor (ER)-alpha by DNA methyltransferase and histone deacetylase inhibition in human ER-alpha-negative breast cancer cells. Cancer Res. 2001, 61, 7025–7029. [Google Scholar]
  223. Fuqua, S.A.; Wiltschke, C.; Zhang, Q.X.; Borg, A.; Castles, C.G.; Friedrichs, W.E.; Hopp, T.; Hilsenbeck, S.; Mohsin, S.; O’Connell, P.; et al. A hypersensitive estrogen receptor-alpha mutation in premalignant breast lesions. Cancer Res. 2000, 60, 4026–4029. [Google Scholar]
  224. Parl, F.F. Multiple mechanisms of estrogen receptor gene repression contribute to ER-negative breast cancer. Pharmacogenom. J. 2003, 3, 251–253. [Google Scholar] [CrossRef]
  225. Prossnitz, E.R.; Barton, M. The G-protein-coupled estrogen receptor GPER in health and disease. Nat. Rev. Endocrinol. 2011, 7, 715–726. [Google Scholar] [CrossRef] [PubMed]
  226. Filardo, E.J.; Quinn, J.A.; Bland, K.I.; Frackelton, A.R., Jr. Estrogen-induced activation of Erk-1 and Erk-2 requires the G protein-coupled receptor homolog, GPR30, and occurs via trans-activation of the epidermal growth factor receptor through release of HB-EGF. Mol. Endocrinol. 2000, 14, 1649–1660. [Google Scholar] [CrossRef]
  227. Revankar, C.M.; Cimino, D.F.; Sklar, L.A.; Arterburn, J.B.; Prossnitz, E.R. A transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science 2005, 307, 1625–1630. [Google Scholar] [CrossRef]
  228. Bryś, M. Androgens and androgen receptor: Do they play a role in breast cancer? Med. Sci. Monit. 2000, 6, 433–438. [Google Scholar]
  229. De Amicis, F.; Thirugnansampanthan, J.; Cui, Y.; Selever, J.; Beyer, A.; Parra, I.; Weigel, N.L.; Herynk, M.H.; Tsimelzon, A.; Lewis, M.T.; et al. Androgen receptor overexpression induces tamoxifen resistance in human breast cancer cells. Breast Cancer Res. Treat. 2010, 121, 1–11. [Google Scholar] [CrossRef]
  230. Ciupek, A.; Rechoum, Y.; Gu, G.; Gelsomino, L.; Beyer, A.R.; Brusco, L.; Covington, K.R.; Tsimelzon, A.; Fuqua, S.A. Androgen receptor promotes tamoxifen agonist activity by activation of EGFR in ERα-positive breast cancer. Breast Cancer Res. Treat. 2015, 154, 225–237. [Google Scholar] [CrossRef]
  231. Chang, C.; Saltzman, A.; Yeh, S.; Young, W.; Keller, E.; Lee, H.J.; Wang, C.; Mizokami, A. Androgen receptor: An overview. Crit. Rev. Eukaryot. Gene Expr. 1995, 5, 97–125. [Google Scholar] [CrossRef]
  232. Higa, G.M.; Fell, R.G. Sex hormone receptor repertoire in breast cancer. Int. J. Breast Cancer 2013, 2013, 284036. [Google Scholar] [CrossRef] [PubMed]
  233. Tarulli, G.A.; Butler, L.M.; Tilley, W.D.; Hickey, T.E. Bringing androgens up a NOTCH in breast cancer. Endocr. Relat. Cancer 2014, 21, T183–T202. [Google Scholar] [CrossRef]
  234. Cuenca-López, M.D.; Montero, J.C.; Morales, J.C.; Prat, A.; Pandiella, A.; Ocana, A. Phospho-kinase profile of triple negative breast cancer and androgen receptor signaling. BMC Cancer. 2014, 14, 302. [Google Scholar] [CrossRef] [PubMed]
  235. Heinlein, C.A.; Chang, C. The roles of androgen receptors and androgen-binding proteins in nongenomic androgen actions. Mol. Endocrinol. 2002, 16, 2181–2187. [Google Scholar] [CrossRef] [PubMed]
  236. Stecca, B.; Ruiz i Altaba, A. Context-dependent regulation of the GLI code in cancer by HEDGEHOG and non-HEDGEHOG signals. J. Mol. Cell Biol. 2010, 2, 84–95. [Google Scholar] [CrossRef] [PubMed]
  237. Johnson, R.W.; Nguyen, M.P.; Padalecki, S.S.; Grubbs, B.G.; Merkel, A.R.; Oyajobi, B.O.; Matrisian, L.M.; Mundy, G.R.; Sterling, J.A. TGF-beta promotion of Gli2-induced expression of parathyroid hormone-related protein, an important osteolytic factor in bone metastasis, is independent of canonical Hedgehog signaling. Cancer Res. 2011, 71, 822–831. [Google Scholar] [CrossRef] [PubMed]
  238. Javelaud, D.; Alexaki, V.I.; Dennler, S.; Mohammad, K.S.; Guise, T.A.; Mauviel, A. TGF-β/SMAD/GLI2 signaling axis in cancer progression and metastasis. Cancer Res. 2011, 71, 5606–5610. [Google Scholar] [CrossRef]
  239. Ramaswamy, B.; Lu, Y.; Teng, K.Y.; Nuovo, G.; Li, X.; Shapiro, C.L.; Majumder, S. Hedgehog signaling is a novel therapeutic target in tamoxifen-resistant breast cancer aberrantly activated by PI3K/AKT pathway. Cancer Res. 2012, 72, 5048–5059. [Google Scholar] [CrossRef]
  240. Wang, X.; Zhao, S.; Xin, Q.; Zhang, Y.; Wang, K.; Li, M. Recent progress of CDK4/6 inhibitors′ current practice in breast cancer. Cancer Gene Ther. 2024, 31, 1283–1291. [Google Scholar] [CrossRef]
  241. Gallanis, G.T.; Sharif, G.M.; Schmidt, M.O.; Friedland, B.N.; Battina, R.; Rahhal, R.; Davis, J.E.; Khan, I.S., Jr.; Wellstein, A.; Riegel, A.T. Stromal Senescence following Treatment with the CDK4/6 Inhibitor Palbociclib Alters the Lung Metastatic Niche and Increases Metastasis of Drug-Resistant Mammary Cancer Cells. Cancers 2023, 15, 1908. [Google Scholar] [CrossRef]
  242. Roberto, M.; Astone, A.; Botticelli, A.; Carbognin, L.; Cassano, A.; D’Auria, G.; Fabbri, A.; Fabi, A.; Gamucci, T.; Krasniqi, E.; et al. CDK4/6 Inhibitor Treatments in Patients with Hormone Receptor Positive, Her2 Negative Advanced Breast Cancer: Potential Molecular Mechanisms, Clinical Implications and Future Perspectives. Cancers 2021, 13, 332. [Google Scholar] [CrossRef]
  243. André, F.; Ciruelos, E.; Rubovszky, G.; Campone, M.; Loibl, S.; Rugo, H.S.; Iwata, H.; Conte, P.; Mayer, I.A.; Kaufman, B.; et al. Alpelisib for PIK3CA-mutated, hormone receptor–positive advanced breast cancer. N. Engl. J. Med. 2019, 380, 1929–1940. [Google Scholar] [CrossRef]
  244. Schwartzberg, L.S.; Yardley, D.A.; Elias, A.D.; Patel, M.; LoRusso, P.; Burris, H.A.; Gucalp, A.; Peterson, A.C.; Blaney, M.E.; Steinberg, J.L.; et al. A Phase I/Ib Study of Enzalutamide Alone and in Combination with Endocrine Therapies in Women with Advanced Breast Cancer. Clin. Cancer Res. 2017, 23, 4046–4054. [Google Scholar] [CrossRef]
  245. Lu, Q.; Xia, W.; Lee, K.; Zhang, J.; Yuan, H.; Yuan, Z.; Shi, Y.; Wang, S.; Xu, F. Bicalutamide plus Aromatase Inhibitor in Patients with Estrogen Receptor-Positive/Androgen Receptor-Positive Advanced Breast Cancer. Oncologist 2020, 25, 21–e15. [Google Scholar] [CrossRef] [PubMed]
  246. Wang, C.; Lin, Y.; Zhu, H.; Zhou, Y.; Mao, F.; Huang, X.; Sun, Q.; Li, C. Efficacy and Safety Profile of Histone Deacetylase Inhibitors for Metastatic Breast Cancer: A Meta-Analysis. Front. Oncol. 2022, 12, 901152. [Google Scholar] [CrossRef] [PubMed]
  247. Wawruszak, A.; Borkiewicz, L.; Okon, E.; Kukula-Koch, W.; Afshan, S.; Halasa, M. Vorinostat (SAHA) and Breast Cancer: An Overview. Cancers 2021, 13, 4700. [Google Scholar] [CrossRef] [PubMed]
  248. Patnaik, A.; Haluska, P.; Tolcher, A.W.; Erlichman, C.; Papadopoulos, K.P.; Lensing, J.L.; Beeram, M.; Molina, J.R.; Rasco, D.W.; Arcos, R.R.; et al. A First-in-Human Phase I Study of the Oral p38 MAPK Inhibitor, Ralimetinib (LY2228820 Dimesylate), in Patients with Advanced Cancer. Clin. Cancer Res. 2016, 22, 1095–1102. [Google Scholar] [CrossRef] [PubMed]
  249. Zaman, K.; Winterhalder, R.; Mamot, C.; Hasler-Strub, U.; Rochlitz, C.; Mueller, A.; Berset, C.; Wiliders, H.; Perey, L.; Rudolf, C.B.; et al. Fulvestrant with or without selumetinib, a MEK 1/2 inhibitor, in breast cancer progressing after aromatase inhibitor therapy: A multicentre randomised placebo-controlled double-blind phase II trial, SAKK 21/08. Eur. J. Cancer 2015, 51, 1212–1220. [Google Scholar] [CrossRef]
  250. Kaboli, P.J.; Imani, S.; Jomhori, M.; Ling, K.H. Chemoresistance in breast cancer: PI3K/Akt pathway inhibitors vs the current chemotherapy. Am. J. Cancer Res. 2021, 11, 5155–5183. [Google Scholar]
  251. McDonnell, D.P.; Wardell, S.E.; Chang, C.Y.; Norris, J.D. Next-Generation Endocrine Therapies for Breast Cancer. J. Clin. Oncol. 2021, 39, 1383–1388. [Google Scholar] [CrossRef]
  252. Razavi, P.; Chang, M.T.; Xu, G.; Bandlamudi, C.; Ross, D.S.; Vasan, N.; Cai, Y.; Bielski, C.M.; Donoghue, M.T.A.; Jonsson, P.; et al. The Genomic Landscape of Endocrine-Resistant Advanced Breast Cancers. Cancer Cell 2018, 34, 427–438.e6. [Google Scholar] [CrossRef]
  253. Goetz, M.P.; Bagegni, N.A.; Batist, G.; Brufsky, A.; Cristofanilli, M.A.; Damodaran, S.; Daniel, B.R.; Fleming, G.F.; Gradishar, W.J.; Graff, S.L.; et al. Lasofoxifene versus fulvestrant for ER+/HER2- metastatic breast cancer with an ESR1 mutation: Results from the randomized, phase II ELAINE 1 trial. Ann. Oncol. 2023, 34, 1141–1151. [Google Scholar] [CrossRef]
  254. Goetz, M.P.; Wander, S.A.; Bachelot, T.; de Nonneville, A.; Gal-Yam, E.N.; Sammons, S.L.; Shen, S.; Twelves, C.; Boruta, G.; Portman, D.J.; et al. ELAINE 3: Phase 3 study of lasofoxifene plus abemaciclib to treat ER+/HER2-, ESR1-mutated, metastatic breast cancer. Future Oncol. 2025, 21, 1317–1324. [Google Scholar] [CrossRef]
  255. McDonnell, D.P.; Wardell, S.E. The molecular mechanisms underlying the pharmacological actions of ER modulators: Implications for new drug discovery in breast cancer. Curr. Opin. Pharmacol. 2010, 10, 620–628. [Google Scholar] [CrossRef] [PubMed]
  256. Wardell, S.E.; Nelson, E.R.; Chao, C.A.; Alley, H.M.; McDonnell, D.P. Evaluation of the pharmacological activities of RAD1901, a selective estrogen receptor degrader. Endocr. Relat. Cancer. 2015, 22, 713–724. [Google Scholar] [CrossRef]
  257. Beumer, J.H.; Foldi, J. Pharmacology and pharmacokinetics of elacestrant. Cancer Chemother. Pharmacol. 2023, 92, 157–163. [Google Scholar] [CrossRef]
  258. Sarfraz, A.; Sarfraz, M.; Javad, F.; Khalid, M.; Shah, B.; Gul, A.; Ganiyani, M.A.; Ismail, A.; Cheema, K. Elacestrant in hormone receptor-positive metastatic breast cancer: A post-hoc analysis. Explor. Target. Antitumor Ther. 2025, 6, 1002293. [Google Scholar] [CrossRef]
  259. Ciruelos, E.M.; Hamilton, E.P.; Kim, S.-B.; Im, Y.-H.; Segui Solis, E.; García Saenz., J.Á.; Di Sanzo, A.; Domínguez Lizarbe, M.; Wasserman, T.; Theall, K.P.; et al. 364P Elacestrant in combination with abemaciclib in patients (pts) with brain metastasis (mets) from estrogen receptor-positive (ER+), HER2-negative (HER2-) breast cancer: Preliminary data from ELECTRA, an open-label, multicenter, phase Ib/II study. Ann. Oncol. 2024, 35, S370. [Google Scholar] [CrossRef]
  260. Oliveira, M.; Hamilton, E.P.; Incorvati, J.; de la Heras, B.B.; Calvo, E.; Garcia-Corbacho, J.; Ruiz-Borrego, M.; Vaklavas, C.; Turner, N.C.; Ciruelos, E.M.; et al. Serena-1: Updated analyses from a phase 1 study (parts C/D) of the next-generation oral SERD camizestrant (AZD9833) in combination with palbociclib, in women with ER-positive, HER2-negative advanced breast cancer. J. Clin. Oncol. 2022, 40, 1032. [Google Scholar] [CrossRef]
  261. Bhagwat, S.V.; Zhao, B.; Shen, W.; Mur, C.; Barr, R.; Kindler, L.J.; Rubio, A.; Bastian, J.A.; Cohen, J.D.; Mattioni, B.E.; et al. Preclinical characterization of LY3484356, a novel, potent and orally bioavailable selective estrogen receptor degrader (SERD). Cancer Res. 2021, 81, 1236. [Google Scholar] [CrossRef]
  262. Bondeson, D.P.; Mares, A.; Smith, I.E.; Ko, E.; Campos, S.; Miah, A.H.; Mulholland, K.E.; Routly, N.; Buckley, D.L.; Gustafson, J.L.; et al. Catalytic in vivo protein knockdown by small-molecule PROTACs. Nat. Chem. Biol. 2015, 11, 611–617. [Google Scholar] [CrossRef] [PubMed]
  263. Lin, X.; Xiang, H.; Luo, G. Targeting estrogen receptor α for degradation with PROTACs: A promising approach to overcome endocrine resistance. Eur. J. Med. Chem. 2020, 206, 112689. [Google Scholar] [CrossRef]
  264. Flanagan, J.J.; Qian, Y.; Gough, S.M.; Andreoli, M.; Bookbinder, M.; Cadelina, G.; Bradley, J.; Rousseau, E.; Willard, R.; Pizzano, J.; et al. Abstract P5-04-18: ARV-471, an oral estrogen receptor PROTAC degrader for breast cancer. Cancer Res. 2019, 79 (Suppl. S4), P5-04-18. [Google Scholar] [CrossRef]
  265. Hodges-Gallaghe, L.; Sun, R.; Myles, D.C.; Harmon, C.L.; Kushner, P.J. Abstract P5-05-02: Preclinical development of OP-1250, an oral complete estrogen receptor antagonist (CERAN) that shrinks ER-positive breast tumors in xenograft models. Cancer Res. 2020, 80 (Suppl. S4), P5-05-02. [Google Scholar] [CrossRef]
  266. Parisian, A.D.; Barratt, S.A.; Hodges-Gallagher, L.; Ortega, F.E.; Peña, G.; Sapugay, J.; Robello, B.; Sun, R.; Kulp, D.; Palanisamy, G.S.; et al. Palazestrant (OP-1250), A Complete Estrogen Receptor Antagonist, Inhibits Wild-type and Mutant ER-positive Breast Cancer Models as Monotherapy and in Combination. Mol. Cancer Ther. 2024, 23, 285–300. [Google Scholar] [CrossRef] [PubMed]
  267. Puyang, X.; Furman, C.; Zheng, G.Z.; Wu, Z.J.; Banka, D.; Aithal, K.; Agoulnik, S.; Bolduc, D.M.; Buonamici, S.; Caleb, B.; et al. Discovery of Selective Estrogen Receptor Covalent Antagonists for the Treatment of ERαWT and ERαMUT Breast Cancer. Cancer Discov. 2018, 8, 1176–1193. [Google Scholar] [CrossRef] [PubMed]
  268. Al Hashami, Z.S.; van der Vegt, B.; Mourits, M.J.E.; Kluiver, J.; van den Berg, A. miRNA-dependent resistance mechanisms to anti-hormonal therapies in estrogen receptor-positive breast cancer patients. Mol. Ther. Oncol. 2025, 33, 200941. [Google Scholar] [CrossRef]
  269. Huang, L.; Liang, G.; Zhang, Q.; Zhao, W. The Role of Long Noncoding RNAs in Antiestrogen Resistance in Breast Cancer: An Overview and Update. J. Breast Cancer 2020, 23, 129–140. [Google Scholar] [CrossRef]
  270. Leppert, W.; Strąg-Lemanowicz, A. Rola leczenia hormonalnego u pacjentów z zaawansowaną chorobą nowotworową. Med. Paliatywna Prakt. 2015, 9, 30–38. [Google Scholar]
  271. Nourmoussavi, M.; Pansegrau, G.; Popesku, J.; Hammond, G.L.; Kwon, J.S.; Carey, M.S. Ovarian ablation for premenopausal breast cancer: A review of treatment considerations and the impact of premature menopause. Cancer Treat. Rev. 2017, 55, 26–35. [Google Scholar] [CrossRef]
  272. Castellón, E.A.; Indo, S.; Contreras, H.R. Cancer Stemness/Epithelial-Mesenchymal Transition Axis Influences Metastasis and Castration Resistance in Prostate Cancer: Potential Therapeutic Target. Int. J. Mol. Sci. 2022, 23, 14917. [Google Scholar] [CrossRef]
  273. Deli, T.; Orosz, M.; Jakab, A. Hormone Replacement Therapy in Cancer Survivors-Review of the Literature. Pathol. Oncol. Res. 2020, 26, 63–78. [Google Scholar] [CrossRef]
  274. Rozenberg, S.; Di Pietrantonio, V.; Vandromme, J.; Gilles, C. Menopausal hormone therapy and breast cancer risk. Best Pract. Res. Clin. Endocrinol. Metab. 2021, 35, 101577. [Google Scholar] [CrossRef]
  275. van Bommel, M.H.D.; IntHout, J.; Veldmate, G.; Kets, C.M.; de Hullu, J.A.; van Altena, A.M.; Harmsen, M.G. Contraceptives and cancer risks in BRCA1/2 pathogenic variant carriers: A systematic review and meta-analysis. Hum. Reprod. Update 2023, 29, 197–217. [Google Scholar] [CrossRef]
  276. Loizzi, V.; Cerbone, M.; Arezzo, F.; Silvestris, E.; Damiani, G.R.; Cazzato, G.; Cicinelli, E.; Cormio, G. Contraception as chemoprevention of ovarian cancer in BRCA1 and BRCA2 women. Hormones 2024, 23, 277–286. [Google Scholar] [CrossRef]
  277. Wang, X.; Ha, D.; Yoshitake, R.; Chan, Y.S.; Sadava, D.; Chen, S. Exploring the Biological Activity and Mechanism of Xenoestrogens and Phytoestrogens in Cancers: Emerging Methods and Concepts. Int. J. Mol. Sci. 2021, 22, 8798. [Google Scholar] [CrossRef] [PubMed]
  278. Tanwar, A.K.; Dhiman, N.; Kumar, A.; Jaitak, V. Engagement of phytoestrogens in breast cancer suppression: Structural classification and mechanistic approach. Eur. J. Med. Chem. 2021, 213, 113037. [Google Scholar] [CrossRef] [PubMed]
  279. Adeel, M.; Zain, M.; Shakoor, N.; Azeem, I.; Hussain, M.; Ahmad, M.A.; Chaudhary, S.; Zaheer, U.; Aziz, M.A.; Ahmar, S.; et al. Estrogens in plants and emerging risks to human health. Environ. Int. 2023, 178, 107985. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The structural organisation of alpha and beta estrogen receptors. NTD: gene transcription mechanism; DBD: DNA-binding domain; LBD: ligand-binding domain; AF-1: activation function 1; AF-2: activation function 2.
Figure 1. The structural organisation of alpha and beta estrogen receptors. NTD: gene transcription mechanism; DBD: DNA-binding domain; LBD: ligand-binding domain; AF-1: activation function 1; AF-2: activation function 2.
Biomedicines 13 02620 g001
Figure 2. The role of the ER in the activation of various signaling pathways in cells. ER: estrogen receptor; Ras: GTPase HRas; MAPK: mitogen-activated protein kinase; P: phosphorylated form; Src: proto-oncogene tyrosine-protein kinase; pCREB1: phosphorylated cAMP-responsive element-binding protein 1; cAMP: cyclic adenosine monophosphate; PI3K: phosphatidylinositol 3-kinase; AKT: protein kinase B; mTOR: mammalian target of rapamycin; PTEN: phosphatidylinositol-3,4,5-trisphosphate 3-phosphatase; PIP2: phosphatidylinositol 4,5-bisphosphate; PIP3: phosphatidylinositol 3,4,5-trisphosphate.
Figure 2. The role of the ER in the activation of various signaling pathways in cells. ER: estrogen receptor; Ras: GTPase HRas; MAPK: mitogen-activated protein kinase; P: phosphorylated form; Src: proto-oncogene tyrosine-protein kinase; pCREB1: phosphorylated cAMP-responsive element-binding protein 1; cAMP: cyclic adenosine monophosphate; PI3K: phosphatidylinositol 3-kinase; AKT: protein kinase B; mTOR: mammalian target of rapamycin; PTEN: phosphatidylinositol-3,4,5-trisphosphate 3-phosphatase; PIP2: phosphatidylinositol 4,5-bisphosphate; PIP3: phosphatidylinositol 3,4,5-trisphosphate.
Biomedicines 13 02620 g002
Figure 3. A schematic diagram illustrating ER-mediated signaling in the TME. ERs: estrogen receptors; CAFs: cancer-associated fibroblasts; IL-1β: interleukin-1 beta; IL-8: IL-1β: interleukin-8; VEGF: vascular endothelial growth factor; PDGF: platelet-derived growth factor; EGF: epidermal growth factor; MAPK: mitogen-activated protein kinase; PI3K: phosphatidylinositol 3-kinase; cAMP: cyclic adenosine monophosphate; MDSCs: myeloid-derived suppressor cells; TAMs: tumour-associated macrophages; NKs: natural killer cells; MMP2: matrix metalloproteinase-2; MMP9: Matrix metalloproteinase-9; ↑: increase; ↓: decrease.
Figure 3. A schematic diagram illustrating ER-mediated signaling in the TME. ERs: estrogen receptors; CAFs: cancer-associated fibroblasts; IL-1β: interleukin-1 beta; IL-8: IL-1β: interleukin-8; VEGF: vascular endothelial growth factor; PDGF: platelet-derived growth factor; EGF: epidermal growth factor; MAPK: mitogen-activated protein kinase; PI3K: phosphatidylinositol 3-kinase; cAMP: cyclic adenosine monophosphate; MDSCs: myeloid-derived suppressor cells; TAMs: tumour-associated macrophages; NKs: natural killer cells; MMP2: matrix metalloproteinase-2; MMP9: Matrix metalloproteinase-9; ↑: increase; ↓: decrease.
Biomedicines 13 02620 g003
Table 1. A comparison of the role of different types of ER in selected cancers.
Table 1. A comparison of the role of different types of ER in selected cancers.
Estrogen Receptor ExpressionEstrogen Receptor Level Compared to Normal TissuePoor Prognosis
Connection
References
Ovarian cancerERα+
ERβ+
GPER+
ERα ˜
Erβ↓
GPER ˜
Erβ↓
ERβ ND
GPER↑/↓
[85,86,87,89,90,91]
Breast cancerERα+/-
ERβ+/-
GPER+
ERα↑
ERβ ND
GPER ND
Erα↓
ERβ ND
GPER↑/↓
[104,105,106,107,109,111,112,113]
Thyroid cancerERα+
ERβ+
GPER+
Erα↑
Erβ↓
GPER ND
Erα↑
Erβ↓
GPER↑/↓
[120,124,129,130]
Colorectal cancerERα+
ERβ+
GPER+
Erα↑
Erβ↓
GPER ND
Erα↑
Erβ↓
GPER↑/↓
[138,139,140,145,146,147]
+: expression; -: non expression; ↑: increase; ↓: decrease; ˜: similar level; ND: not data avalible; ERα: estrogen receptor alpha; ERβ: estrogen receptor beta; GPER: G protein-coupled estrogen receptor.
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

Gruszka, O.; Jurzak, M.; Bednarek, I.A. Estrogen Receptors as Key Factors in Carcinogenesis. Biomedicines 2025, 13, 2620. https://doi.org/10.3390/biomedicines13112620

AMA Style

Gruszka O, Jurzak M, Bednarek IA. Estrogen Receptors as Key Factors in Carcinogenesis. Biomedicines. 2025; 13(11):2620. https://doi.org/10.3390/biomedicines13112620

Chicago/Turabian Style

Gruszka, Oliwia, Magdalena Jurzak, and Ilona Anna Bednarek. 2025. "Estrogen Receptors as Key Factors in Carcinogenesis" Biomedicines 13, no. 11: 2620. https://doi.org/10.3390/biomedicines13112620

APA Style

Gruszka, O., Jurzak, M., & Bednarek, I. A. (2025). Estrogen Receptors as Key Factors in Carcinogenesis. Biomedicines, 13(11), 2620. https://doi.org/10.3390/biomedicines13112620

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