Next Article in Journal / Special Issue
Maternal MicroRNA Profile Changes When LH Is Added to the Ovarian Stimulation Protocol: A Pilot Study
Previous Article in Journal
Current Approaches to Epigenetic Therapy
Previous Article in Special Issue
Linc2function: A Comprehensive Pipeline and Webserver for Long Non-Coding RNA (lncRNA) Identification and Functional Predictions Using Deep Learning Approaches
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The ErbB Signaling Network and Its Potential Role in Endometrial Cancer

by
Georgios Androutsopoulos
1,2,*,
Ioanna Styliara
2,
Evgenia Zarogianni
2,
Nadia Lazurko
2,
George Valasoulis
3,4,
Georgios Michail
2 and
Georgios Adonakis
2
1
Gynaecological Oncology Unit, Department of Obstetrics and Gynaecology, School of Medicine, University of Patras, 26504 Rion, Greece
2
Department of Obstetrics and Gynaecology, School of Medicine, University of Patras, 26504 Rion, Greece
3
Department of Obstetrics and Gynaecology, Medical School, University of Thessaly, 41334 Larisa, Greece
4
Hellenic National Public Health Organization—ECDC, 15123 Athens, Greece
*
Author to whom correspondence should be addressed.
Epigenomes 2023, 7(4), 24; https://doi.org/10.3390/epigenomes7040024
Submission received: 17 August 2023 / Revised: 24 September 2023 / Accepted: 26 September 2023 / Published: 1 October 2023
(This article belongs to the Collection Feature Papers in Epigenomes)

Abstract

:
Endometrial cancer (EC) is the second most common malignancy of the female reproductive system worldwide. The updated EC classification emphasizes the significant role of various signaling pathways such as PIK3CA-PIK3R1-PTEN and RTK/RAS/β-catenin in EC pathogenesis. Some of these pathways are part of the EGF system signaling network, which becomes hyperactivated by various mechanisms and participates in cancer pathogenesis. In EC, the expression of ErbB receptors is significantly different, compared with the premenopausal and postmenopausal endometrium, mainly because of the increased transcriptional activity of ErbB encoding genes in EC cells. Moreover, there are some differences in ErbB-2 receptor profile among EC subgroups that could be explained by the alterations in pathophysiology and clinical behavior of various EC histologic subtypes. The fact that ErbB-2 receptor expression is more common in aggressive EC histologic subtypes (papillary serous and clear cell) could indicate a future role of ErbB-targeted therapies in well-defined EC subgroups with overexpression of ErbB receptors.

1. Introduction

Endometrial cancer (EC) is the second most common malignancy of the female reproductive system worldwide [1]. It is more prevalent in wealthy and more developed regions (North America, Europe, Australia, New Zealand), compared with less developed ones (Central and South America, Asia, Africa) [1]. However, mortality rates are considerably higher in less developed areas (northern Africa, Melanesia) [2,3]. The disease usually affects postmenopausal women, with an average annual incidence reaching 2.1% [1]. Although the vast majority of EC patients are postmenopausal, approximately 14% of them are premenopausal and almost 4% are below 40 years of age [4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22].
Current evidence does not support any screening methodology for early detection of EC, as cervical cytology performs poorly [23]. However, the fluorescence in situ hybridization test (FISH) in vaginal swab specimens has shown promising results in EC detection [24]. Artificial intelligence and machine learning algorithms have been proposed to assist in the discrimination between benign and malignant endometrial nuclei, obtained via image analysis and measured from liquid-based cytology slides and lesions so far. These show promising results, as their performance appears to be similar to that of traditional regression models in EC [25,26,27,28]. Moreover, biospectroscopy has several applications in biomedical science, from detecting toxins and pollutants in the human body to identifying areas of stem cells in human tissue. It can effectively identify biomarkers of disease states at many organ sites without the need for staining or isotopic labeling [29,30]. Apart from that, pelvic ultrasound scans or saline infusion sonograms can be offered on a 1–2-yearly basis in the context of routine gynecological examination, except for individuals undergoing close follow-up for hereditary, non-polyposis colon cancer (HNPCC) [31].
In the past, the sporadic classification of EC cases was based on clinical, metabolic, endocrine and pathological features [32,33]. More recently, genomic data including somatic mutation rates, frequency of copy number alterations and MSI status have been used to create an updated EC classification, reflecting the increased impact of molecular biology in disease progression and patients’ outcome [34,35]. Moreover, the updated EC classification emphasizes the significant role of various signaling pathways, such as PIK3CA-PIK3R1-PTEN and RTK/RAS/β-catenin, in EC pathogenesis [34,35,36].
Some of these pathways are part of the EGF system signaling network, which becomes hyperactivated with various mechanisms (gain of function mutations, genomic amplification, chromosomal rearrangements and autocrine activation) and participates in cancer pathogenesis [37,38,39,40,41,42].
Our aim is to provide an update on current knowledge of the signaling network of ErbB receptors and their participation in cancer pathogenesis, as well as their potential clinical role in EC cases.

2. Physiology of ErbB Receptors

The EGF system is present in various human organs and plays a significant role in cell proliferation, differentiation, migration and apoptosis during embryogenesis and postnatal development [39,43,44].

2.1. ErbB Receptors

ErbB receptors are members of the subclass I superfamily of receptor tyrosine kinases (RTKs) [37,39,45]. In humans, the EGF system consists of the following ErbB receptors: epidermal growth factor receptor (EGFR), ErbB-2, ErbB-3 and ErbB-4 [37,39,44,45,46]. These receptors are trans-membrane glycoproteins that catalyze the transferring of γ phosphate of ATP to hydroxyl groups of tyrosines in target proteins [47]. However, ErbB-3 has no intrinsic tyrosine kinase activity, and it depends on another ErbB receptor (usually ErbB-2) for intracellular signaling [45,48].
Regarding their structure, ErbB receptors have an extracellular ligand-binding domain, a transmembrane domain, a short juxtamembrane section, an intracellular bilobed tyrosine kinase domain and a tyrosine-containing C-terminal tail (Figure 1) [44,45,46,49].
The extracellular ligand-binding domain is divided into four subdomains: L1 (or I), CR1 (or II), L2 (or III) and CR2 (or IV) [44,46,49]. The leucine-rich subdomains L1 and L2 participate in ligand binding [44,46,49]. The cysteine-rich subdomains CR1 and CR2 participate in disulfide bond formation, while subdomain CR1 contains a β-hairpin loop and participates in ErbB receptors’ homodimerization and heterodimerization [44,46,49]. Moreover, the intracellular tyrosine kinase domain is subdivided into two lobes, N and C [44,46].

2.2. ErbB Ligands

In humans, the EGF system has the following ErbB peptide mediators (ligands): EGF, transforming growth factor-a (TGF-a), amphiregulin (AR), heparin-binding growth factor (HB-EGF), betacellulin (BTC), epigen, epiregulin (EPR), neuregulin-1 (NRG-1), neuregulin-2 (NRG-2), neuregulin-3 (NRG-3), neuregulin-4 (NRG-4), neuroglycan C and tomoregulin [39,44,45,46]. Ligand binding to the extracellular domain of the ErbB receptor results in conformational changes and induces homodimerization and heterodimerization of receptors [37,44,45,46]. However, the ErbB-2 receptor fails to bind any ligands [37,44,45,46].
Based on their affinity for one or more receptors, ErbB ligands could be further classified into the following subgroups:
1. Ligands with binding specificity for EGFR only: EGF, TGF-a and AR [44,45,46].
2. Ligands with dual binding specificity for EGFR and ErbB4: HB-EGF, BTC and EPR [44,45,46].
3. Ligands with binding specificity for ErbB-3 only: neuroglycan C [44,45,46].
4. Ligands with binding specificity for ErbB-4 only: NRG-3, NRG-4 and tomoregulin [44,45,46].
5. Ligands with dual binding specificity for ErbB-3 and ErbB-4: NRG-1, NRG-2 [44,45,46].
All these data are presented in detail in Table 1.
It should be emphasized that ErbB ligands usually act a short distance from the cells producing them [46,50]. Overall, ErbB ligands may act either on the same cell (autocrine signaling), on an adjacent cell (juxtacrine signaling) or on a nearby cell (paracrine signaling) [46,50].

2.3. Receptor Homodimerization and Heterodimerization

There are two distinct conformations of the extracellular ligand-binding domain, based on the activation status of EGFR, ErbB-3 and ErbB-4 receptors:
1. Closed conformation. When ErbB receptors are inactive, there are intramolecular interactions between the cysteine-rich subdomains CR1 and CR2, causing closed conformation of the extracellular ligand-binding domain [44,45,46,51,52].
2. Open conformation. When ErbB receptors become active, the leucine-rich subdomains L1 and L2 create a ligand-binding pocket, allowing interactions with a single ligand, while the extracellular ligand-binding domain takes an open conformation and the β-hairpin loop dimerization arm of subdomain CR1 is exposed [44,45,46,51,52].
It seems that there is equilibrium between both conformations of the extracellular ligand-binding domain, related directly to ligand presence and subsequent ligand binding [51,52,53]. More specifically, ligand binding to the leucine-rich subdomains L1 and L2 stabilises the extracellular ligand-binding domain to an open conformation, exposes the β-hairpin loop dimerization arm of subdomain CR1 and allows receptor homodimerization and heterodimerization [44,45,46,52,53,54]. Subsequently, ErbB receptor dimerization induces conformational changes of the intracellular bilobed tyrosine kinase domain [44,45,46,55,56].
In contrast, the extracellular ligand-binding domain of the ErbB-2 receptor has an extended conformation that is not suitable for ligand binding, as there is close proximity of the leucine-rich subdomains L1 and L2, abolishing the ligand-binding site [44,45,46,57,58,59]. However, the extended conformation of the ErbB-2 receptor is necessary for interaction with other ErbB receptors and subsequent ligand-independent heterodimerization and signaling [44,45,46,57,58,59]. Moreover, abnormal overexpression of the ErbB-2 receptor permits ligand-independent receptor homodimerization [44,46,58].
Overall, homodimerization and heterodimerization of ErbB receptors represents an essential part in the pathophysiology of the EGF system signaling network [44,45,46,55,56]. Furthermore, the ErbB-2 and ErbB-3 heterodimer is the most transforming and mitogenic receptor complex [60].

2.4. Intracellular Tyrosine Kinase Activation

Following homodimerization and heterodimerization of ErbB receptors, conformational changes of the intracellular tyrosine kinase domain take place, which in turn cause tyrosine kinase activation and phosphorylation of the tyrosine-containing C-terminal tail [44,45,46,55,56].
As already mentioned, the intracellular tyrosine kinase domain has a bilobed structure, with ATP binding between the N and C lobes [44,45,46,56]. More specifically, the C-lobe of an intracellular tyrosine kinase domain (activator) allosterically interacts with the N-lobe of another intracellular tyrosine kinase domain (receiver) within the same dimerization pair [44,45,46,56]. This interaction induces conformational changes in the N-lobe of the receiver tyrosine kinase and finally causes its activation [44,45,46,56]. Subsequently, the activated receiver tyrosine kinase catalyzes phosphorylation of tyrosine residues in the tyrosine-containing C-terminal tail of the activator tyrosine kinase [44,45,46,56]. These phosphorylated tyrosine residues serve as docking sites for adaptor proteins, enzymes and various signaling molecules containing Src homology 2 (SH2) and phosphotyrosine binding (PTB) domains [38,44,45,46,56,61,62].

3. Signaling Pathways

Activation and subsequent autophosphorylation of ErbB receptors enables recruitment of various signaling molecules containing the SH2 and PTB domains, that result in downstream signaling via several pathways (Figure 2) [38,42,44,45,46,56,61,62,63]:

3.1. Ras/Raf/MAPK Pathway

The Ras/Raf/mitogen-activated protein kinase (MAPK) pathway has a fundamental role in cell biology, mainly as a transducer of extracellular signals to cellular responses [63,64]. Ιt is actively involved in cell cycle regulation (proliferation, differentiation, migration and apoptosis), integrin signaling, tissue repair and angiogenesis [64,65,66,67].
Following ErbB receptor activation and phosphorylation of the tyrosine-containing C-terminal tail, the activated ErbB receptor recruits, directly or indirectly (through the Shc adaptor protein), an adaptor protein named growth factor receptor binding protein 2 (Grb2) via its SH2 domain (Src homology 2) [64,68,69,70,71,72]. Subsequently, the SH3 domain of Grb2 interacts with the proline-rich C-terminal domain of Son of Sevenless (Sos) in order to create an ErbB receptor–Grb2–Sos complex [64,68,71,72]. This leads to Sos translocation to the cell membrane and enables its interaction with Ras [72,73,74].
The interaction between Sos and Ras causes conformational changes and allosteric activation of Sos through a rotation of its REM domain [72,73,74,75]. The allosteric activation of Sos allows Ras binding and promotes replacement of GDP with GTP in Ras that leads to Ras activation (Ras-GTP) and initiation of the Ras pathway [72,73,75,76].
More specifically, Ras–GTP recruits and dimerizes Raf-1 protein kinase on the inner side of the cell membrane, in order to activate it through tyrosine phosphorylation [72,77,78]. Subsequently, activated Raf-1 interacts and activates MAPK/ERK kinase (MEK1 and MEK2), which in turn phosphorylates, activates and anchors to the cytoplasm downstream proteins such as extracellular signal-regulated kinases (ERK1 and ERK2) [64,72]. Then, activated ERK1 and ERK2 translocate to the nucleus in order to phosphorylate and activate various nuclear transcription factors involved in cell proliferation, differentiation and migration [63,64,72].
Overall, the Ras/Raf/MAPK pathway is implicated in a wide variety of cellular biological functions, but is also related to tumorigenesis [64,72].

3.2. PI3K/Akt Pathway

The phosphatidylinositol 3-kinase (PI3K)/Akt pathway has an essential role in cell biology, mainly in transduction of extracellular signals to intracellular messages [79]. Ιt is actively involved in cell cycle regulation (proliferation, migration and apoptosis) and cytoskeletal rearrangement [80].
Following ErbB receptor activation and phosphorylation of the tyrosine-containing C-terminal tail, the activated ErbB receptor directly recruits the PI3K (subclass IA) regulatory subunit via its SH2 domain and causes allosteric activation of the PI3K catalytic subunit [79,81,82]. Subsequently, activated PI3K catalyzes conversion of phosphatidylinositol (4, 5) bisphosphate (PIP2) to phosphatidylinositol (3, 4, 5) trisphosphate (PIP3) at the cell membrane [79,81]. Then PIP3 provides docking sites for signaling proteins with pleckstrin homology (PH) domains, including 3-phosphoinositide-dependent kinase 1 (PDK1) and serine-threonine protein kinase Akt (protein kinase B (PKB)) [79,81,82].
In particular, PIP3 directly recruits Akt to the cell membrane via its PH domain and this results in Akt conformational changes and exposure of two crucial amino-acid residues (Thr308 and Ser473) [79,80,83]. Thr308 is phosphorylated by PDK1, while Ser473 is phosphorylated by PDK2 [79,80,81,83,84]. Both phosphorylation events are necessary for full Akt activation, which in turn phosphorylates many cytoplasmic and nuclear proteins and regulates a wide range of cellular processes involved in protein synthesis, cell cycle progression and cell survival [79,80,81,82,83,84].
It is interesting to note that specific docking sites for the PI3K (subclass IA) regulatory subunit are present on the ErbB-3 receptor, while they are absent on the EGFR receptor [63,85]. Moreover, EGFR-dependent PI3K activation occurs either through EGFR and ErbB-3 dimerization or through a Gab-1 docking protein [63,86].
Overall, the PI3K/Akt pathway is implicated in various cellular processes and plays an important role in carcinogenesis [63,80].

3.3. STAT Pathway

The signal transducers and activators of transcription (STAT) pathway has a principal role in cell biology, mainly as a transducer of extracellular cytokine signals to cellular responses [87,88,89]. Ιt is actively involved in cell cycle regulation (proliferation, differentiation, migration and apoptosis) [87,88,89].
Following ErbB receptor activation and phosphorylation of the tyrosine-containing C-terminal tail, the activated ErbB receptor can cause JAK—independent tyrosine phosphorylation of STAT proteins, probably via the Src kinase [87,88,89,90]. Phosphorylated and activated STAT proteins create dimers via SH2 domain interactions and translocate to the nucleus, where they bind to specific DNA sequences in gene promoters and regulate gene transcription [87,88,89,91].
Overall, the STAT pathway is implicated in various developmental and homeostatic processes, but also related to tumorigenesis [87,88,89,91].

3.4. Src Kinase Pathway

The Src kinase pathway has a critical role in cell biology, especially as a transducer of extracellular signals to cellular responses [92]. Ιt is actively involved in cell cycle regulation (proliferation, adhesion, migration and apoptosis), integrin signaling and angiogenesis [91,92].
Following ErbB receptor activation and phosphorylation of the tyrosine-containing C-terminal tail, the activated ErbB receptor recruits Src kinase via its SH2 domain and causes Src activation [91,93]. Subsequently, activated Src acts as signal transducer and enhancer of ErbB receptor activation [63,94,95].
More specifically, Src activates many downstream proteins (p130Cas, FAK, PI3K, VEGF, HIF1α and STAT) through tyrosine phosphorylation [91]. Furthermore, Src regulates various cell cycle proteins (c-Myc, cyclin D and p21) through transcriptional and post-translational mechanisms [91].
Overall, the Src kinase pathway is implicated in many cellular processes and plays an important role in carcinogenesis [63,91,96,97].

3.5. PLCγ/PKC Pathway

The phospholipase Cγ (PLCγ)/protein kinase C (PKC) pathway has an essential role in cell biology, mainly in transduction of extracellular signals to intracellular messages [46,98]. Ιt is actively involved in cell cycle regulation (proliferation, differentiation and migration) and angiogenesis [46,98].
Following ErbB receptor activation and phosphorylation of the tyrosine-containing C-terminal tail, the activated ErbB receptor recruits PLCγ via its SH2 domain and causes PLCγ phosphorylation and activation [98,99,100]. Subsequently, activated PLCγ catalyzes hydrolysis of phosphatidylinositol (4, 5) bisphosphate (PIP2) to inositol (1, 4, 5) trisphosphate (IP3) and (1, 2) diacylglycerol (DAG) [95,98,101]. IP3 has significant role in intracellular calcium release, while DAG is cofactor in protein kinase C (PKC) activation [46,63,95]. Activated PKC catalyzes phosphorylation and activation of several transcription factors [46]. Moreover, PKC is actively involved in multiple signaling components, including MAPK and JNK pathways [46,63,95,102,103].
Overall, the PLCγ/PKC pathway is implicated in many cellular processes and plays an important role in carcinogenesis [46,98].

4. Epigenetic Regulation of ErbB Signaling

As already mentioned, the activation and subsequent autophosphorylation of ErbB receptors enables recruitment of various signaling molecules and results in downstream signaling via several pathways [38,42,44,45,46,56,61,62,63].
However, heritable changes in gene function without alterations in the DNA sequence (epigenetic changes) could possibly affect ErbB—mediated signal transduction and gene transcription via several mechanisms [104,105]:

4.1. DNA Methylation

DNA methylation is an extensively studied mechanism of epigenetic alterations [106]. DNA methylation patterns (methylation and demethylation) are regulated by specific enzymes and subsequently affect gene transcription [106,107].
More specifically, DNA methylation is catalyzed by the family of DNA methyltransferase (DNMT) enzymes, which transfer methyl groups from S-adenosyl-L-methionine (SAM) to cytosine residues and form 5-methylcytosine (5-mC) [106,108]. The majority of DNA methylation occurs in CpG islands, in which cytosine is followed by a guanine [106]. Most CpG islands are present in promoters and their methylation leads to transcriptional silencing [106,108]. Especially in ErbB signaling, PTEN promoter hypermethylation suppresses PTEN expression and activity, with a direct effect on PI3K/Akt pathway signaling [104,109].
Likewise, DNA demethylation is achieved either by active enzymatic demethylation or by passive replication—dependent on the dilution of methylation [108]. Particularly in active enzymatic demethylation, 5-mC undergoes a series of oxidation reactions catalyzed by the methylcytosine dioxygenases Ten-Eleven-Translocation (TET) enzymes [108,110]. The 5-hydroxymethylcytosine (5hmC) is the first intermediate of active DNA demethylation [108]. Enrichment of 5hmc in promoter regions is often associated with activation of gene expression [108]. In this way, DNA demethylation links to genomic instability [106,108]. Especially in ErbB signaling, Ras promoter hypomethylation enhances Ras expression and activity, with a direct effect on signaling of the Ras/Raf/MAPK and PI3K/Akt pathways [104].

4.2. Histone Modification

Histone modifications represent another mechanism of epigenetic alterations [106]. They affect lysine and arginine residues on histone tails, which are targets of covalent post-transcriptional modifications (acetylation, methylation, phosphorylation and ubiquitylation) [106,108].
More specifically, histone acetylation occurs through the addition of an acetyl group to the lysine residues in histone tails [106]. Histone acetyltransferases (HATs) add acetyl groups and are associated with active gene transcription at promoter and enhancer sites [106]. In contrast, histone deacetylases (HDACs) remove acetyl groups and are associated with gene silencing and transcriptional repression [106]. Especially in ErbB signaling, EGFR acetylation by CREB-binding protein (CBP) acetyltransferase affects receptor phosphorylation and subsequent activation [104,111].
Likewise, histone methylation occurs through the addition of methyl groups to the arginine or lysine residues in histone tails [106]. Histone methyltransferases (HMTs) add methyl groups and are associated with both active gene transcription and gene repression [106]. In contrast, histone demethylases (HDMs) remove methyl groups [106].

4.3. Non-Coding RNAs

Non-coding RNAs (ncRNAs) are functional RNA molecules that occupy a large fraction of the genome, but they are not translated into proteins [108,112,113,114]. They have key roles in the regulation of gene expression at transcriptional and translational levels [113,114]. Moreover, they can be divided into the following categories: microRNAs (miRNAs), long noncoding RNAs (lncRNAs), small interfering RNAs (siRNAs), small nuclear RNAs (snRNAs), small nucleolar RNAs (snoRNAs), ribosomal RNAs (rRNAs), transfer RNAs (tRNAs), circular RNAs (circRNAs) and PIWI-interacting RNAs (piRNAs) [108,112,113,114]. Among these, miRNAs and lncRNAs have crucial roles in cancer epigenetics [113,114].
More specifically, miRNAs are small ncRNAs, approximately 19 to 22 nucleotides in length, that regulate gene expression by posttranscriptional silencing [115,116]. They usually bind to the 3′-untranslated region (3′-UTR) of target messenger RNA (mRNA) molecules, resulting in either translational inhibition or mRNA degradation [113,114,115,117].
In contrast, lncRNAs are larger ncRNAs, more than 200 nucleotides in length, that regulate gene expression at transcriptional, post-transcriptional and epigenetic levels [113,118,119]. In particular, guide lncRNAs act by recruiting or rejecting epigenetic regulators (chromatin modifying complexes and chromatin remodeling complexes) onto specific chromosomal loci [120]. Architect lncRNAs act by modifying the three-dimensional chromatin conformation [120]. Enhancer lncRNAs regulate gene transcription through enhancer-like functions [120]. Moreover, lncRNAs regulate DNA methylation status by recruiting or inhibiting DNA methyltransferases and demethylases [120]. Furthermore, lncRNAs regulate mRNA stability, protein—protein interactions and post-translational protein modifications [113,121,122,123,124].

5. EGF Dysregulation and Carcinogenesis

Dysregulation of the EGF system signaling network participates in the pathogenesis of various diseases (diabetes, autoimmune, inflammatory, cardiovascular and nervous system disorders), as well as in cancer [37,38,39,41]. Moreover, constitutive EGF system activation and uncontrolled ErbB signaling may disrupt the balance in cell cycle regulation (proliferation, differentiation, migration and apoptosis), sensitize cells to oncogenic transformation and trigger ErbB-induced oncogenesis [37,38,39,41,42].
More specifically, in malignant transformation, the EGF system becomes hyperactivated with the following four main mechanisms: gain of function mutations, genomic amplification, chromosomal rearrangements and autocrine activation [40,41,42].

5.1. Gain of Function Mutations

The gain of function (GOF) mutations may have a crucial role in carcinogenesis, as they generate novel protein isoforms with new and important functions [125]. Based on their consequences for cancer development, GOF mutations could be further subclassified into driver and passenger mutations [125,126]. Driver mutations provide a selective cell growth advantage and promote cancer development, while passenger mutations do not confer any cell growth advantage and do not contribute to carcinogenesis [126,127].
Especially in the EGF system, GOF mutations could possibly affect most domains of an ErbB receptor and lead to aberrant downstream signaling [42]. More specifically, a GOF mutation usually involves the bilobed tyrosine kinase domain of an ErbB receptor and causes tyrosine kinase hyperactivation and aberrant downstream signaling, as well as conferring oncogenic properties [42]. However, GOF mutations could also affect various ErbB receptor domains (the extracellular ligand-binding domain, transmembrane domain and short juxtamembrane section) and cause receptor activation using alternative mechanisms [42].

5.2. Genomic Amplification

Genomic amplification is the copy number increase in a specific region of the genome and is associated with overexpression of the amplified genes [128,129]. It usually occurs during development and carcinogenesis and may be promoted by common chromosomal fragile sites, errors in DNA replication or telomere dysfunction [129,130]. Amplified sequences can be organized as extrachromosomal elements, repeated units at a single locus or interspersed throughout the genome [128,129].
Especially in the EGF system, genomic amplification and subsequent ErbB receptor overexpression leads to increased receptor local concentration, constitutive receptor activation, avoidance of receptor regulatory mechanisms and aberrant downstream signaling [42,131,132]. More specifically, ErbB-2 overexpression causes constitutive ErbB-2 activation as well as EGFR ligand-independent activation [131]. Moreover, ErbB-2 overexpression inhibits down-regulation mechanisms of ErbB-2 and EGFR [131].

5.3. Chromosomal Rearrangements

Chromosomal rearrangements have important roles in carcinogenesis and include deletions, duplications, inversions and translocations [133]. They are mainly caused by either defective DNA double strand break repair or faulty DNA replication [134]. Based on their effect on chromosomes, they could be further subclassified into simple and complex [134]. Simple chromosomal rearrangement results from a single fusion that preserves genetic information but sometimes disrupts regulation of the genes involved [134]. In contrast, complex chromosomal rearrangement results from multiple fusions at a single locus that cause changes in genetic content and in chromosomal linear structure [134]. Overall, chromosomal rearrangements lead to either hybrid gene formation or gene dysregulation [133,134].
Especially in the EGF system, chromosomal rearrangements cause the formation of fusion oncoproteins, consisting partly of the ErbB receptor and partly of the fusion partner [42,135]. These fusion oncoproteins have remarkable structural similarities, can be membrane bound or cytoplasmic, and contain an activated tyrosine kinase domain [42,135].

5.4. Autocrine Activation

Autocrine activation is a type of self-stimulation in which a cell secretes a hormone-like factor that binds functional receptors on the same cell [136]. This type of cell signaling has a significant role in carcinogenesis, particularly in cases of constitutive autocrine activation [136,137,138].
Especially in the EGF system, autocrine activation of ErbB receptors is a well described phenomenon that leads to downstream signaling via several pathways and may confer oncogenic properties [42,139,140].

6. ErbB Receptors in Endometrial Cancer

During the menstrual cycle, there is a wide variation in the profile of ErbB receptors, indicating a central role of the EGF system in the regulation of endometrial cyclical growth and shedding [141,142].
In EC, the expression of ErbB receptors is significantly different, compared with the premenopausal and postmenopausal endometrium [141,143,144]. This is mainly because of the increased transcriptional activity of ErbB encoding genes in EC cells [144].

6.1. Profile of ErbB Receptors in Endometrial Cancer

Overall, EGFR overexpression is reported in 43–67% of unselected EC cases [144,145,146,147,148,149,150,151,152,153,154,155]. EGFR overexpression is present in approximately 46% of type I EC (endometrioid) cases [149,151,156]. EGFR overexpression is observed in 34–50% of type II EC (papillary serous, clear cell, undifferentiated) cases [149,151,156,157,158,159].
ErbB-2 overexpression and ΕrbB-2 gene amplification represents a very rare event in unselected EC cases [144,149,153,154,155]. However, ΕrbB-2 overexpression and ΕrbB-2 gene amplification are present in only 8–15% and 3% of type I EC cases, respectively [144,149,156,160,161,162,163]. In contrast, ΕrbB-2 overexpression and ErbB-2 gene amplification are more common in type II EC cases [149,151,156,157,158,159].
Moreover, the exact frequency of ErbB-2 overexpression and ΕrbB-2 gene amplification in type II EC remains controversial, as there are many racial differences [149,151,157,164,165]. More specifically, ErbB-2 overexpression and ΕrbB-2 gene amplification are more common in African—American patients with type II EC, when compared with Caucasian individuals [164,165].
Likewise, ErbB-2 overexpression and ΕrbB-2 gene amplification have significant variations among different histologic subtypes of type II EC [149,151,157,160,165,166,167]. ErbB-2 overexpression and ΕrbB-2 gene amplification are reported in 18–80% and 17–47% of papillary serous EC cases, respectively [149,151,160,163,165,166,167], and 33% and 16–50% of clear cell EC cases, respectively [149,151,160,167].
ErbB-3 overexpression is reported in 30% of unselected EC cases [141,153]. More specifically, ErbB-3 overexpression is more common in well differentiated tumors when compared with moderately and poorly differentiated ones [141].
Similarly, ErbB-4 overexpression is reported in 15% of unselected EC cases [141,153].
Overall, there are some differences in ErbB-2 receptor profile in selected EC patients (EC histologic subtypes and racial—ethnic subgroups) [143,150,157,164]. ErbB-2 receptor expression is more common in papillary serous and clear cell EC cases [143,150,157]. This is mainly based on differences in the pathophysiology and clinical behavior of various EC histologic subtypes [143,150,157].

6.2. Clinical Role in Endometrial Cancer

The relationship of the ErbB receptors profile with disease stage, tumor grade and response to treatment remains controversial in EC cases [149,153].
In particular, the clinical role of EGFR overexpression has not been studied thoroughly in EC patients [149,153]. Some studies demonstrate an association between EGFR overexpression and poor clinical outcome, while others report otherwise [145,146,147,148]. It seems that EGFR overexpression may have a dual role in EC cases [149]. EGFR overexpression in type I EC is associated with less aggressive disease and more favorable outcomes [149,151,153,157]. In contrast, EGFR overexpression in type II EC is associated with more aggressive disease and adverse clinical outcomes [149,151,153,157].
However, the clinical significance of ΕrbB-2 overexpression and ΕrbB-2 gene amplification has been studied extensively in EC patients [151,157,160,165,166,168,169,170]. ΕrbB-2 overexpression and ΕrbB-2 gene amplification are indicators of a more aggressive disease with reduced response to treatment and less favorable outcomes, especially in patients with type II EC [151,153,157,160,165,166,168,169,170,171].
Furthermore, the clinical role of ErbB-3 and ErbB-4 overexpression has not been studied extensively in patients with EC [141,143,150,151,152,153,157].
It becomes apparent that ErbB-2 receptor expression is more common in aggressive EC histologic subtypes (papillary serous and clear cell) [143,150,157]. This possibly indicates a future role of ErbB-targeted therapies in well-defined EC subgroups with overexpression of ErbB receptors [150,157,172].

7. Conclusions

Overall, the EGF system signaling network becomes hyperactivated with various mechanisms and possibly participates in EC pathogenesis via several signaling pathways [37,38,39,40,41,42]. There are some differences in ErbB-2 receptor profile among EC subgroups that could be explained by the differences in pathophysiology and clinical behavior of various EC histologic subtypes [143,150,157].
The fact that ErbB-2 receptor expression is more common in aggressive EC histologic subtypes (papillary serous and clear cell), might indicate a future role of ErbB-targeted therapies in well-defined EC subgroups with overexpression of ErbB receptors [150,157,172]. In this context, future studies are needed in order to evaluate thoroughly the effectiveness of ErbB-targeted therapies as single agents or adjuvant treatment in well-defined EC subgroups with overexpression of ErbB receptors [7,21,172,173,174,175,176,177,178].

Author Contributions

Conceptualization, G.A. (Georgios Androutsopoulos) and G.A. (Georgios Adonakis); data curation, G.A. (Georgios Androutsopoulos), I.S., E.Z., N.L., G.V. and G.M.; writing—original draft preparation, G.A. (Georgios Androutsopoulos), I.S., E.Z., N.L., G.V. and G.M.; writing—review and editing, G.A. (Georgios Androutsopoulos), I.S., E.Z., N.L., G.V., G.M. and G.A. (Georgios Adonakis). All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
  2. WHO Globocan. Estimated Cancer Incidence, Mortality and Prevalence Worldwide in 2020; International Agency for Research on Cancer: Lyon, France, 2020.
  3. Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, E359–E386. [Google Scholar] [CrossRef]
  4. Gitsch, G.; Hanzal, E.; Jensen, D.; Hacker, N.F. Endometrial cancer in premenopausal women 45 years and younger. Obstet. Gynecol. 1995, 85, 504–508. [Google Scholar] [CrossRef] [PubMed]
  5. Duska, L.R.; Garrett, A.; Rueda, B.R.; Haas, J.; Chang, Y.; Fuller, A.F. Endometrial Cancer in Women 40 Years Old or Younger. Gynecol. Oncol. 2001, 83, 388–393. [Google Scholar] [CrossRef] [PubMed]
  6. Erkanli, S.; Ayhan, A. Fertility-sparing therapy in young women with endometrial cancer: 2010 update. Int. J. Gynecol. Cancer 2010, 20, 1170–1187. [Google Scholar] [CrossRef] [PubMed]
  7. Androutsopoulos, G. Current treatment options in patients with endometrial cancer. J. Community Med. Health Educ. 2012, 2, e113. [Google Scholar] [CrossRef]
  8. Androutsopoulos, G.; Decavalas, G. Management of endometrial cancer. Int. J. Transl. Community Dis. 2013, 1, 101. [Google Scholar]
  9. Koufopoulos, N.; Carrer, D.; Koureas, N.; Sofopoulos, M.; Paraoulakis, I.; Androutsopoulos, G.; Arnogiannaki, N.; Zygouris, D.; Derdelis, G.; Terzakis, E. Pathological data on 19 cases of endometrioid carcinoma of the endometrium in women of reproductive age. Int. J. Gynecol. Cancer 2013, 23 (Suppl. 1), 322. [Google Scholar]
  10. Androutsopoulos, G.; Decavalas, G. Endometrial cancer: Current treatment strategies. World J. Oncol. Res. 2014, 1, 1–4. [Google Scholar]
  11. Androutsopoulos, G.; Michail, G.; Adonakis, G.; Decavalas, G. Current treatment approach of endometrial cancer. Int. J. Clin. Ther. Diagn. 2015, S1, 8–11. [Google Scholar]
  12. Androutsopoulos, G.; Adonakis, G.; Decavalas, G. Present and future in endometrial cancer treatment. Obstet. Gynecol. Int. J. 2015, 2, 00031. [Google Scholar] [CrossRef]
  13. ACOG. ACOG practice bulletin No. 149: Endometrial cancer. Obstet. Gynecol. 2015, 125, 1006–1026. [Google Scholar] [CrossRef] [PubMed]
  14. Colombo, N.; Creutzberg, C.; Amant, F.; Bosse, T.; Gonzalez-Martin, A.; Ledermann, J.; Marth, C.; Nout, R.; Querleu, D.; Mirza, M.R.; et al. ESMO-ESGO-ESTRO Consensus Conference on Endometrial Cancer: Diagnosis, treatment and follow-up. Ann. Oncol. 2016, 27, 16–41. [Google Scholar] [CrossRef] [PubMed]
  15. Androutsopoulos, G.; Michail, G.; Decavalas, G. New insights in endometrial cancer treatment. Clin. Oncol. Endometrial Cancer 2016, 1, 1040. [Google Scholar]
  16. Androutsopoulos, G.; Decavalas, G. Standard and novel therapies in endometrial cancer. J. Gynecol. Women’s Health 2016, 1, 555564. [Google Scholar] [CrossRef]
  17. Androutsopoulos, G.; Kotsopoulos, I.; Decavalas, G. Fertility preservation in young patients with endometrial cancer. World J. Oncol. Res. 2016, 3, 36–39. [Google Scholar] [CrossRef]
  18. Sundar, S.; Balega, J.; Crosbie, E.; Drake, A.; Edmondson, R.; Fotopoulou, C.; Gallos, I.; Ganesan, R.; Gupta, J.; Johnson, N.; et al. BGCS uterine cancer guidelines: Recommendations for practice. Eur. J. Obstet. Gynecol. Reprod. Biol. 2017, 213, 71–97. [Google Scholar] [CrossRef]
  19. Androutsopoulos, G.; Kotsopoulos, I.; Korompelis, P.; Michail, G.; Adonakis, G.; Decavalas, G. Systematic lymphadenectomy or sentinel lymph node dissection in endometrial cancer: A clinical dilemma. Hell. J. Obst. Gynecol. 2017, 16, 14–19. [Google Scholar]
  20. Androutsopoulos, G.; Kotsopoulos, I.; Adonakis, G.; Decavalas, G. Conservative management of young patients with early stage endometrial cancer. J. Gynecol. Women’s Health 2017, 2, 555586. [Google Scholar] [CrossRef]
  21. Androutsopoulos, G.; Adonakis, G.; Decavalas, G. ErbB targeted therapy in endometrial cancer. In Endometrial Cancer: Current Epidemiology, Detection and Management; Farghaly, S., Ed.; Nova Science Publishers: Hauppauge, NY, USA, 2014. [Google Scholar]
  22. Androutsopoulos, G.; Kotsopoulos, I.; Korompelis, P.; Michail, G.; Adonakis, G.; Decavalas, G. Conservative therapeutic approach in young patients with endometrial cancer: Is it really possible? Hell. J. Obst. Gynecol. 2017, 16, 7–23. [Google Scholar]
  23. Michail, G.D. Endometrial Cancer-Diagnosis. Int. J. Clin. Ther. Diagn. 2015, 1, 17–27. [Google Scholar]
  24. Weimer, J.; Hüttmann, M.; Nusilati, A.; Andreas, S.; Röseler, J.; Tribian, N.; Rogmans, C.; Stope, M.B.; Dahl, E.; Mustea, A.; et al. Fluorescence in situ hybridization test for detection of endometrial carcinoma cells by non-invasive vaginal swab. J. Cell. Mol. Med. 2023, 27, 379–391. [Google Scholar] [CrossRef] [PubMed]
  25. Pouliakis, A.; Damaskou, V.; Margari, N.; Karakitsou, E.; Pergialiotis, V.; Valasoulis, G.; Michail, G.; Chrelias, C.; Chrelias, G.; Sioulas, V.; et al. Artificial Intelligence and Image Analysis for the Identification of Endometrial Malignancies: A Comparative Study. In Research Anthology on Medical Informatics in Breast and Cervical Cancer; I.R. Management Association, Ed.; IGI Global: Hershey, PA, USA, 2023; pp. 1–30. [Google Scholar]
  26. Pouliakis, A.; Damaskou, V.; Margari, N.; Karakitsou, E.; Pergialiotis, V.; Valasoulis, G.; Michail, G.; Chrelias, C.; Chrelias, G.; Sioulas, V.; et al. Artificial Intelligence and Image Analysis for the Identification of Endometrial Malignancies: A Comparative Study. In Quality Assurance in the Era of Individualized Medicine; Moumtzoglou, A.S., Ed.; IGI Global: Hershey, PA, USA, 2020; pp. 110–146. [Google Scholar]
  27. Pouliakis, A.; Margari, N.; Karakitsou, E.; Valasoulis, G.; Koufopoulos, N.; Koureas, N.; Alamanou, E.; Pergialiotis, V.; Damaskou, V.; Panayiotides, I.G. Artificial Intelligence via Competitive Learning and Image Analysis for Endometrial Malignancies: Discriminating Endometrial Cells and Lesions. Int. J. Reliab. Qual. E Healthc. 2019, 8, 38–54. [Google Scholar] [CrossRef]
  28. Piedimonte, S.; Rosa, G.; Gerstl, B.; Sopocado, M.; Coronel, A.; Lleno, S.; Vicus, D. Evaluating the use of machine learning in endometrial cancer: A systematic review. Int. J. Gynecol. Cancer 2023, 33, 1383–1393. [Google Scholar] [CrossRef]
  29. Purandare, N.C.; Trevisan, J.; Patel, I.I.; Gajjar, K.; Mitchell, A.L.; Theophilou, G.; Valasoulis, G.; Martin, M.; von Bünau, G.; Kyrgiou, M.; et al. Exploiting biospectroscopy as a novel screening tool for cervical cancer: Towards a framework to validate its accuracy in a routine clinical setting. Bioanalysis 2013, 5, 2697–2711. [Google Scholar] [CrossRef]
  30. Theophilou, G.; Morais, C.L.M.; Halliwell, D.E.; Lima, K.M.G.; Drury, J.; Martin-Hirsch, P.L.; Stringfellow, H.F.; Hapangama, D.K.; Martin, F.L. Synchrotron- and focal plane array-based Fourier-transform infrared spectroscopy differentiates the basalis and functionalis epithelial endometrial regions and identifies putative stem cell regions of human endometrial glands. Anal. Bioanal. Chem. 2018, 410, 4541–4554. [Google Scholar] [CrossRef]
  31. Jacobs, I.; Gentry-Maharaj, A.; Burnell, M.; Manchanda, R.; Singh, N.; Sharma, A.; Ryan, A.; Seif, M.W.; Amso, N.N.; Turner, G.; et al. Sensitivity of transvaginal ultrasound screening for endometrial cancer in postmenopausal women: A case-control study within the UKCTOCS cohort. Lancet Oncol. 2010, 12, 38–48. [Google Scholar] [CrossRef]
  32. Bokhman, J.V. Two pathogenetic types of endometrial carcinoma. Gynecol. Oncol. 1983, 15, 10–17. [Google Scholar] [CrossRef]
  33. Doll, A.; Abal, M.; Rigau, M.; Monge, M.; Gonzalez, M.; Demajo, S.; Colás, E.; Llauradó, M.; Alazzouzi, H.; Planagumá, J.; et al. Novel molecular profiles of endometrial cancer-new light through old windows. J. Steroid. Biochem. Mol. Biol. 2008, 108, 221–229. [Google Scholar] [CrossRef]
  34. Kandoth, C.; Schultz, N.; Cherniack, A.; Akbani, R.; Liu, Y.; Shen, H.; Robertson, A.; Pashtan, I.; Shen, R.; Benz, C.; et al. Integrated genomic characterization of endometrial carcinoma. Nature 2013, 497, 67–73. [Google Scholar]
  35. Talhouk, A.; McConechy, M.; Leung, S.; Yang, W.; Lum, A.; Senz, J.; Boyd, N.; Pike, J.; Anglesio, M.; Kwon, J.; et al. Confirmation of ProMisE: A simple, genomics-based clinical classifier for endometrial cancer. Cancer 2017, 123, 802–813. [Google Scholar] [CrossRef] [PubMed]
  36. Le Gallo, M.; Bell, D. The emerging genomic landscape of endometrial cancer. Clin. Chem. 2014, 60, 98–110. [Google Scholar] [CrossRef] [PubMed]
  37. Holbro, T.; Civenni, G.; Hynes, N.E. The ErbB receptors and their role in cancer progression. Exp. Cell Res. 2003, 284, 99–110. [Google Scholar] [CrossRef] [PubMed]
  38. Marmor, M.D.; Skaria, K.B.; Yarden, Y. Signal transduction and oncogenesis by ErbB/HER receptors. Int. J. Radiat. Oncol. 2004, 58, 903–913. [Google Scholar] [CrossRef]
  39. Überall, I.; Kolář, Z.; Trojanec, R.; Berkovcová, J.; Hajdúch, M. The status and role of ErbB receptors in human cancer. Exp. Mol. Pathol. 2008, 84, 79–89. [Google Scholar] [CrossRef] [PubMed]
  40. Lemmon, M.A.; Schlessinger, J. Cell Signaling by Receptor Tyrosine Kinases. Cell 2010, 141, 1117–1134. [Google Scholar] [CrossRef]
  41. McDonell, L.M.; Kernohan, K.D.; Boycott, K.M.; Sawyer, S.L. Receptor tyrosine kinase mutations in developmental syndromes and cancer: Two sides of the same coin. Hum. Mol. Genet. 2015, 24, R60–R66. [Google Scholar] [CrossRef]
  42. Du, Z.; Lovly, C.M. Mechanisms of receptor tyrosine kinase activation in cancer. Mol. Cancer 2018, 17, 58. [Google Scholar] [CrossRef]
  43. Casalini, P.; Iorio, M.V.; Galmozzi, E.; Ménard, S. Role of HER receptors family in development and differentiation. J. Cell. Physiol. 2004, 200, 343–350. [Google Scholar] [CrossRef]
  44. Wieduwilt, M.J.; Moasser, M.M. The epidermal growth factor receptor family: Biology driving targeted therapeutics. Cell. Mol. Life Sci. 2008, 65, 1566–1584. [Google Scholar] [CrossRef]
  45. Linggi, B.; Carpenter, G. ErbB receptors: New insights on mechanisms and biology. Trends Cell Biol. 2006, 16, 649–656. [Google Scholar] [CrossRef] [PubMed]
  46. Roskoski, R., Jr. The ErbB/HER family of protein-tyrosine kinases and cancer. Pharmacol. Res. 2014, 79, 34–74. [Google Scholar] [CrossRef] [PubMed]
  47. Hunter, T. The Croonian Lecture 1997. The phosphorylation of proteins on tyrosine: Its role in cell growth and disease. Philos. Trans. R. Soc. B Biol. Sci. 1998, 353, 583–605. [Google Scholar] [CrossRef] [PubMed]
  48. Mass, R.D. The HER receptor family: A rich target for therapeutic development. Int. J. Radiat. Oncol. 2004, 58, 932–940. [Google Scholar] [CrossRef]
  49. Ogiso, H.; Ishitani, R.; Nureki, O.; Fukai, S.; Yamanaka, M.; Kim, J.H.; Saito, K.; Sakamoto, A.; Inoue, M.; Shirouzu, M.; et al. Crystal structure of the complex of human epidermal growth factor and receptor extracellular domains. Cell 2002, 110, 775–787. [Google Scholar] [CrossRef]
  50. Blobel, C.P. ADAMs: Key components in EGFR signalling and development. Nat. Rev. Mol. Cell Biol. 2005, 6, 32–43. [Google Scholar] [CrossRef]
  51. Ferguson, K.M.; Berger, M.B.; Mendrola, J.M.; Cho, H.-S.; Leahy, D.J.; Lemmon, M.A. EGF Activates Its Receptor by Removing Interactions that Autoinhibit Ectodomain Dimerization. Mol. Cell 2003, 11, 507–517. [Google Scholar] [CrossRef]
  52. Dawson, J.P.; Berger, M.; Lin, C.-C.; Schlessinger, J.; Lemmon, M.A.; Ferguson, K.M. Epidermal Growth Factor Receptor Dimerization and Activation Require Ligand-Induced Conformational Changes in the Dimer Interface. Mol. Cell. Biol. 2005, 25, 7734–7742. [Google Scholar] [CrossRef]
  53. Özcan, F.; Klein, P.; Lemmon, M.A.; Lax, I.; Schlessinger, J. On the nature of low- and high-affinity EGF receptors on living cells. Proc. Natl. Acad. Sci. USA 2006, 103, 5735–5740. [Google Scholar] [CrossRef]
  54. Olayioye, M.; Neve, R.; Lane, H.; Hynes, N. The ErbB signaling network: Receptor heterodimerization in development and cancer. EMBO J. 2000, 19, 3159–3167. [Google Scholar] [CrossRef]
  55. Qian, X.; LeVea, C.; Freeman, J.; Dougall, W.; Greene, M. Heterodimerization of epidermal growth factor receptor and wild-type or kinase-deficient Neu: A mechanism of interreceptor kinase activation and transphosphorylation. Proc. Natl. Acad. Sci. USA 1994, 91, 1500–1504. [Google Scholar] [CrossRef]
  56. Zhang, X.; Gureasko, J.; Shen, K.; Cole, P.A.; Kuriyan, J. An allosteric mechanism for activation of the kinase domain of epidermal growth factor receptor. Cell 2006, 125, 1137–1149. [Google Scholar] [CrossRef]
  57. Graus-Porta, D.; Beerli, R.R.; Daly, J.M.; Hynes, N.E. ErbB-2, the preferred heterodimerization partner of all ErbB receptors, is a mediator of lateral signaling. EMBO J. 1997, 16, 1647–1655. [Google Scholar] [CrossRef]
  58. Garrett, T.P.; McKern, N.M.; Lou, M.; Elleman, T.C.; Adams, T.E.; Lovrecz, G.O.; Kofler, M.; Jorissen, R.N.; Nice, E.C.; Burgess, A.W.; et al. The Crystal Structure of a Truncated ErbB2 Ectodomain Reveals an Active Conformation, Poised to Interact with Other ErbB Receptors. Mol. Cell 2003, 11, 495–505. [Google Scholar] [CrossRef]
  59. Citri, A.; Skaria, K.B.; Yarden, Y. The deaf and the dumb: The biology of ErbB-2 and ErbB-3. Exp. Cell Res. 2003, 284, 54–65. [Google Scholar] [CrossRef]
  60. Yarden, Y.; Sliwkowski, M.X. Untangling the ErbB signalling network. Nat. Rev. Mol. Cell Biol. 2001, 2, 127–137. [Google Scholar]
  61. Zhou, S.; Shoelson, S.E.; Chaudhuri, M.; Gish, G.; Pawson, T.; Haser, W.G.; King, F.; Roberts, T.; Ratnofsky, S.; Lechleider, R.J.; et al. SH2 domains recognize specific phosphopeptide sequences. Cell 1993, 72, 767–778. [Google Scholar] [CrossRef]
  62. Zhang, H.; Berezov, A.; Wang, Q.; Zhang, G.; Drebin, J.; Murali, R.; Greene, M.I. ErbB receptors: From oncogenes to targeted cancer therapies. J. Clin. Investig. 2007, 117, 2051–2058. [Google Scholar] [CrossRef]
  63. Scaltriti, M.; Baselga, J. The Epidermal Growth Factor Receptor Pathway: A Model for Targeted Therapy. Clin. Cancer Res. 2006, 12, 5268–5272. [Google Scholar] [CrossRef]
  64. Molina, J.; Adjei, A. The Ras/Raf/MAPK pathway. J. Thorac. Oncol. 2006, 1, 7–9. [Google Scholar] [CrossRef]
  65. Cary, L.; Han, D.; Guan, J. Integrin-mediated signal transduction pathways. Histol. Histopathol. 1999, 14, 1001–1009. [Google Scholar] [PubMed]
  66. Stacey, D.W. Cyclin D1 serves as a cell cycle regulatory switch in actively proliferating cells. Curr. Opin. Cell Biol. 2003, 15, 158–163. [Google Scholar] [CrossRef] [PubMed]
  67. Kranenburg, O.; Gebbink, M.F.; Voest, E.E. Stimulation of angiogenesis by Ras proteins. Biochim. Biophys. Acta Rev. Cancer 2004, 1654, 23–37. [Google Scholar] [CrossRef] [PubMed]
  68. Lowenstein, E.; Daly, R.; Batzer, A.; Li, W.; Margolis, B.; Lammers, R.; Ullrich, A.; Skolnik, E.; Bar-Sagi, D.; Schlessinger, J. The SH2 and SH3 domain-containing protein GRB2 links receptor tyrosine kinases to ras signaling. Cell 1992, 70, 431–442. [Google Scholar] [CrossRef]
  69. Rozakis-Adcock, M.; McGlade, J.; Mbamalu, G.; Pelicci, G.; Daly, R.; Li, W.; Batzer, A.; Thomas, S.; Brugge, J.; Pelicci, P.G.; et al. Association of the Shc and Grb2/Sem5 SH2-containing proteins is implicated in activation of the Ras pathway by tyrosine kinases. Nature 1992, 360, 689–692. [Google Scholar] [CrossRef]
  70. Batzer, A.G.; Rotin, D.; Urena, J.M.; Skolnik, E.Y.; Schlessinger, J. Hierarchy of Binding Sites for Grb2 and Shc on the Epidermal Growth Factor Receptor. Mol. Cell. Biol. 1994, 14, 5192–5201. [Google Scholar]
  71. Buday, L. Membrane-targeting of signalling molecules by SH2/SH3 domain-containing adaptor proteins. Biochim. Biophys. Acta 1999, 1422, 187–204. [Google Scholar] [CrossRef]
  72. Guo, Y.; Pan, W.; Liu, S.B.; Shen, Z.; Xu, Y.; Hu, L. ERK/MAPK signalling pathway and tumorigenesis. Exp. Ther. Med. 2020, 19, 1997–2007. [Google Scholar] [CrossRef]
  73. Pierre, S.; Bats, A.-S.; Coumoul, X. Understanding SOS (Son of Sevenless). Biochem. Pharmacol. 2011, 82, 1049–1056. [Google Scholar] [CrossRef]
  74. Bandaru, P.; Kondo, Y.; Kuriyan, J. The Interdependent Activation of Son-of-Sevenless and Ras. Cold Spring Harb. Perspect. Med. 2018, 9, a031534. [Google Scholar] [CrossRef]
  75. Freedman, T.; Sondermann, H.; Friedland, G.; Kortemme, T.; Bar-Sagi, D.; Marqusee, S.; Kuriyan, J. A Ras-induced conformational switch in the Ras activator Son of sevenless. Proc. Natl. Acad. Sci. USA 2006, 103, 16692–16697. [Google Scholar] [CrossRef] [PubMed]
  76. Simanshu, D.K.; Nissley, D.V.; McCormick, F. RAS Proteins and Their Regulators in Human Disease. Cell 2017, 170, 17–33. [Google Scholar] [CrossRef] [PubMed]
  77. Marais, R.; Light, Y.; Paterson, H.; Marshall, C. Ras recruits Raf-1 to the plasma membrane for activation by tyrosine phosphorylation. EMBO J. 1995, 14, 3136–3145. [Google Scholar] [CrossRef] [PubMed]
  78. Stokoe, D.; McCormick, F. Activation of c-Raf-1 by Ras and Src through different mechanisms: Activation in vivo and in vitro. EMBO J. 1997, 16, 2384–2396. [Google Scholar] [CrossRef]
  79. Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov. 2009, 8, 627–644. [Google Scholar] [CrossRef]
  80. Vivanco, I.; Sawyers, C.L. The phosphatidylinositol 3-Kinase–AKT pathway in human cancer. Nat. Rev. Cancer 2002, 2, 489–501. [Google Scholar] [CrossRef]
  81. Hemmings, B.; Restuccia, D. PI3K-PKB/Akt pathway. Cold Spring Harb. Perspect. Biol. 2012, 4, a011189. [Google Scholar] [CrossRef]
  82. Porta, C.; Paglino, C.; Mosca, A. Targeting PI3K/Akt/mTOR Signaling in Cancer. Front. Oncol. 2014, 4, 64. [Google Scholar] [CrossRef]
  83. Vanhaesebroeck, B.; Alessi, D. The PI3K-PDK1 connection: More than just a road to PKB. Biochem. J. 2000, 346, 561–576. [Google Scholar] [CrossRef]
  84. Stokoe, D.; Stephens, L.R.; Copeland, T.; Gaffney, P.R.J.; Reese, C.B.; Painter, G.F.; Holmes, A.B.; McCormick, F.; Hawkins, P.T. Dual Role of Phosphatidylinositol-3,4,5-trisphosphate in the Activation of Protein Kinase B. Science 1997, 277, 567–570. [Google Scholar] [CrossRef]
  85. Carpenter, C.; Auger, K.; Chanudhuri, M.; Yoakim, M.; Schaffhausen, B.; Shoelson, S.; Cantley, L. Phosphoinositide 3-kinase is activated by phosphopeptides that bind to the SH2 domains of the 85-kDa subunit. J. Biol. Chem. 1993, 268, 9478–9483. [Google Scholar] [CrossRef] [PubMed]
  86. Mattoon, D.R.; Lamothe, B.; Lax, I.; Schlessinger, J. The docking protein Gab1 is the primary mediator of EGF-stimulated activation of the PI-3K/Akt cell survival pathway. BMC Biol. 2004, 2, 24. [Google Scholar] [CrossRef] [PubMed]
  87. Rawlings, J.; Rosler, K.; Harrison, D. The JAK/STAT signaling pathway. J. Cell Sci. 2004, 117, 1281–1283. [Google Scholar] [CrossRef] [PubMed]
  88. Haura, E.B.; Turkson, J.; Jove, R. Mechanisms of Disease: Insights into the emerging role of signal transducers and activators of transcription in cancer. Nat. Clin. Pract. Oncol. 2005, 2, 315–324. [Google Scholar] [CrossRef] [PubMed]
  89. Harrison, D. The Jak/STAT pathway. Cold Spring Harb. Perspect. Biol. 2012, 4, a011205. [Google Scholar] [CrossRef] [PubMed]
  90. Olayioye, M.A.; Beuvink, I.; Horsch, K.; Daly, J.M.; Hynes, N.E. ErbB Receptor-induced Activation of Stat Transcription Factors Is Mediated by Src Tyrosine Kinases. J. Biol. Chem. 1999, 274, 17209–17218. [Google Scholar] [CrossRef]
  91. Haura, E. SRC and STAT pathways. J. Thorac. Oncol. 2006, 1, 403–405. [Google Scholar] [CrossRef]
  92. Playford, M.P.; Schaller, M.D. The interplay between Src and integrins in normal and tumor biology. Oncogene 2004, 23, 7928–7946. [Google Scholar] [CrossRef]
  93. Bromann, P.; Korkaya, H.; Courtneidge, S. The interplay between Src family kinases and receptor tyrosine kinases. Oncogene 2004, 23, 7957–7968. [Google Scholar] [CrossRef]
  94. Leu, T.; Maa, M. Functional implication of the interaction between EGF receptor and c-Src. Front. Biosci. 2003, 8, s28–s38. [Google Scholar] [CrossRef]
  95. Jorissen, R.N.; Walker, F.; Pouliot, N.; Garrett, T.P.; Ward, C.W.; Burgess, A.W. Epidermal growth factor receptor: Mechanisms of activation and signalling. Exp. Cell Res. 2003, 284, 31–53. [Google Scholar] [CrossRef] [PubMed]
  96. Frame, M. Src in cancer: Deregulation and consequences for cell behaviour. Biochim. Biophys. Acta 2002, 1602, 114–130. [Google Scholar] [CrossRef] [PubMed]
  97. Yeatman, T. A renaissance for SRC. Nat. Rev. Cancer 2004, 4, 470–480. [Google Scholar] [CrossRef] [PubMed]
  98. Kamat, A.; Carpenter, G. Phospholipase C-gamma1: Regulation of enzyme function and role in growth factor-dependent signal transduction. Cytokine Growth Factor Rev. 1997, 8, 109–117. [Google Scholar] [CrossRef] [PubMed]
  99. Rotin, D.; Honegger, A.; Margolis, B.; Ullrich, A.; Schlessinger, J. Presence of SH2 domains of phospholipase C gamma 1 enhances substrate phosphorylation by increasing the affinity toward the epidermal growth factor receptor. J. Biol. Chem. 1992, 267, 9678–9683. [Google Scholar] [CrossRef]
  100. Chattopadhyay, A.; Vecchi, M.; Ji, Q.; Mernaugh, R.; Carpenter, G. The role of individual SH2 domains in mediating association of phospholipase C-gamma1 with the activated EGF receptor. J. Biol. Chem. 1999, 274, 26091–26097. [Google Scholar] [CrossRef]
  101. Kadamur, G.; Ross, E. Mammalian phospholipase C. Annu. Rev. Physiol. 2013, 75, 127–154. [Google Scholar] [CrossRef]
  102. Marais, R.; Light, Y.; Mason, C.; Paterson, H.; Olson, M.F.; Marshall, C.J. Requirement of Ras-GTP-Raf Complexes for Activation of Raf-1 by Protein Kinase C. Science 1998, 280, 109–112. [Google Scholar] [CrossRef]
  103. Schönwasser, D.C.; Marais, R.M.; Marshall, C.J.; Parker, P.J. Activation of the Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase Pathway by Conventional, Novel, and Atypical Protein Kinase C Isotypes. Mol. Cell. Biol. 1998, 18, 790–798. [Google Scholar] [CrossRef]
  104. Spangle, J.M.; Roberts, T.M. Epigenetic regulation of RTK signaling. J. Mol. Med. 2017, 95, 791–798. [Google Scholar] [CrossRef]
  105. Berger, S.L.; Kouzarides, T.; Shiekhattar, R.; Shilatifard, A. An operational definition of epigenetics: Figure 1. Minerva Anestesiol. 2009, 23, 781–783. [Google Scholar] [CrossRef] [PubMed]
  106. Inoue, F.; Sone, K.; Toyohara, Y.; Takahashi, Y.; Kukita, A.; Hara, A.; Taguchi, A.; Tanikawa, M.; Tsuruga, T.; Osuga, Y. Targeting Epigenetic Regulators for Endometrial Cancer Therapy: Its Molecular Biology and Potential Clinical Applications. Int. J. Mol. Sci. 2021, 22, 2305. [Google Scholar] [CrossRef] [PubMed]
  107. Klutstein, M.; Nejman, D.; Greenfield, R.; Cedar, H. DNA Methylation in Cancer and Aging. Cancer Res. 2016, 76, 3446–3450. [Google Scholar] [CrossRef] [PubMed]
  108. Retis-Resendiz, A.M.; González-García, I.N.; León-Juárez, M.; Camacho-Arroyo, I.; Cerbón, M.; Vázquez-Martínez, E.R. The role of epigenetic mechanisms in the regulation of gene expression in the cyclical endometrium. Clin. Epigenetics 2021, 13, 116. [Google Scholar] [CrossRef] [PubMed]
  109. Kang, Y.; Lee, H.; Kim, W. Promoter methylation and silencing of PTEN in gastric carcinoma. Lab. Investig. 2002, 82, 285–291. [Google Scholar] [CrossRef] [PubMed]
  110. Wu, X.; Zhang, Y. TET-mediated active DNA demethylation: Mechanism, function and beyond. Nat. Rev. Genet. 2017, 18, 517–534. [Google Scholar] [CrossRef]
  111. Song, H.; Li, C.; Labaff, A.; Lim, S.; Li, L.; Kan, S.; Chen, Y.; Zhang, K.; Lang, J.; Xie, X.; et al. Acetylation of EGF receptor contributes to tumor cell resistance to histone deacetylase inhibitors. Biochem. Biophys. Res. Commun. 2011, 404, 68–73. [Google Scholar] [CrossRef]
  112. Wei, J.; Huang, K.; Yang, C.; Kang, C. Non-coding RNAs as regulators in epigenetics (Review). Oncol. Rep. 2017, 373–379. [Google Scholar]
  113. Pathania, A.S. Crosstalk between Noncoding RNAs and the Epigenetics Machinery in Pediatric Tumors and Their Microenvironment. Cancers 2023, 15, 2833. [Google Scholar] [CrossRef]
  114. Kumar, S.; Gonzalez, E.A.; Rameshwar, P.; Etchegaray, J.-P. Non-Coding RNAs as Mediators of Epigenetic Changes in Malignancies. Cancers 2020, 12, 3657. [Google Scholar] [CrossRef]
  115. Schickel, R.; Boyerinas, B.; Park, S.; Peter, M. MicroRNAs: Key players in the immune system, differentiation, tumorigenesis and cell death. Oncogene 2008, 27, 5959–5974. [Google Scholar] [CrossRef] [PubMed]
  116. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed]
  117. Kim, Y.J.; Maizel, A.; Chen, X. Traffic into silence: Endomembranes and post-transcriptional RNA silencing. EMBO J. 2014, 33, 968–980. [Google Scholar] [CrossRef]
  118. Ponting, C.P.; Oliver, P.L.; Reik, W. Evolution and Functions of Long Noncoding RNAs. Cell 2009, 136, 629–641. [Google Scholar] [CrossRef]
  119. Clark, M.B.; Mattick, J.S. Long noncoding RNAs in cell biology. Semin. Cell Dev. Biol. 2011, 22, 366–376. [Google Scholar]
  120. Morlando, M.; Fatica, A. Alteration of Epigenetic Regulation by Long Noncoding RNAs in Cancer. Int. J. Mol. Sci. 2018, 19, 570. [Google Scholar] [CrossRef]
  121. Vierbuchen, T.; Agarwal, S.; Johnson, J.L.; Galia, L.; Lei, X.; Stein, K.; Olagnier, D.; Gaede, K.I.; Herzmann, C.; Holm, C.K.; et al. The lncRNA LUCAT1 is elevated in inflammatory disease and restrains inflammation by regulating the splicing and stability of NR4A2. Proc. Natl. Acad. Sci. USA 2023, 120, e2213715120. [Google Scholar] [CrossRef]
  122. Sauvageau, M. Diverging RNPs: Toward Understanding lncRNA-Protein Interactions and Functions. Adv. Exp. Med. Biol. 2019, 1203, 285–312. [Google Scholar]
  123. Ninomiya, K.; Adachi, S.; Natsume, T.; Iwakiri, J.; Terai, G.; Asai, K.; Hirose, T. LncRNA-dependent nuclear stress bodies promote intron retention through SR protein phosphorylation. EMBO J. 2020, 39, e102729. [Google Scholar] [CrossRef]
  124. Taniue, K.; Kurimoto, A.; Sugimasa, H.; Nasu, E.; Takeda, Y.; Iwasaki, K.; Nagashima, T.; Okada-Hatakeyama, M.; Oyama, M.; Kozuka-Hata, H.; et al. Long noncoding RNA UPAT promotes colon tumorigenesis by inhibiting degradation of UHRF1. Proc. Natl. Acad. Sci. USA 2016, 113, 1273–1278. [Google Scholar] [CrossRef]
  125. Li, Y.; Zhang, Y.; Li, X.; Yi, S.; Xu, J. Gain-of-Function Mutations: An emerging advantage for cancer biology. Trends Biochem. Sci. 2019, 44, 659–674. [Google Scholar] [CrossRef] [PubMed]
  126. Stratton, M.; Campbell, P.; Futreal, P. The cancer genome. Nature 2009, 458, 719–724. [Google Scholar] [CrossRef] [PubMed]
  127. Pon, J.; Marra, M. Driver and passenger mutations in cancer. Annu. Rev. Pathol. 2015, 10, 25–50. [Google Scholar] [CrossRef] [PubMed]
  128. Albertson, D.; Collins, C.; McCormick, F.; Gray, J. Chromosome aberrations in solid tumors. Nat. Genet. 2003, 34, 369–376. [Google Scholar] [CrossRef]
  129. Albertson, D. Gene amplification in cancer. Trends Genet. 2006, 22, 447–455. [Google Scholar] [CrossRef]
  130. Jen, K. Gene Amplification. In Brenner’s Encyclopedia of Genetics, 2nd ed.; Maloy, S., Hughes, K., Eds.; Academic Press: San Diego, CA, USA, 2013; pp. 171–172. [Google Scholar]
  131. Worthylake, R.; Opresko, L.K.; Wiley, H.S. ErbB-2 Amplification Inhibits Down-regulation and Induces Constitutive Activation of Both ErbB-2 and Epidermal Growth Factor Receptors. J. Biol. Chem. 1999, 274, 8865–8874. [Google Scholar] [CrossRef]
  132. Carraway, K., 3rd; Sweeney, C. EGF receptor activation by heterologous mechanisms. Cancer Cell 2002, 1, 405–406. [Google Scholar] [CrossRef]
  133. Holland, A.J.; Cleveland, D.W. Chromoanagenesis and cancer: Mechanisms and consequences of localized, complex chromosomal rearrangements. Nat. Med. 2012, 18, 1630–1638. [Google Scholar] [CrossRef]
  134. Hasty, P.; Montagna, C. Chromosomal Rearrangements in Cancer: Detection and potential causal mechanisms. Mol. Cell. Oncol. 2014, 1, e29904. [Google Scholar] [CrossRef]
  135. Stransky, N.; Cerami, E.; Schalm, S.; Kim, J.L.; Lengauer, C. The landscape of kinase fusions in cancer. Nat. Commun. 2014, 5, 4846. [Google Scholar] [CrossRef]
  136. Sporn, M.; Todaro, G. Autocrine secretion and malignant transformation of cells. N. Engl. J. Med. 1980, 303, 878–880. [Google Scholar] [CrossRef] [PubMed]
  137. Walsh, J.; Karnes, W.; Cuttitta, F.; Walker, A. Autocrine growth factors and solid tumor malignancy. West. J. Med. 1991, 155, 152–163. [Google Scholar]
  138. Rizzino, A. Understanding the roles of growth factors in carcinogenesis: Modulation of autocrine growth control by differentiation. Int. J. Dev. Biol. 1993, 37, 61–65. [Google Scholar]
  139. Nicholson, K.; Streuli, C.; Anderson, N. Autocrine Signalling Through erbB Receptors Promotes Constitutive Activation of Protein Kinase B/Akt in Breast Cancer Cell Lines. Breast Cancer Res. Treat. 2003, 81, 117–128. [Google Scholar] [CrossRef]
  140. Singh, A.B.; Harris, R.C. Autocrine, paracrine and juxtacrine signaling by EGFR ligands. Cell. Signal. 2005, 17, 1183–1193. [Google Scholar] [CrossRef]
  141. Srinivasan, R.; Benton, E.; McCormick, F.; Thomas, H.; Gullick, W. Expression of the c-erbB-3/HER-3 and c-erbB-4/HER-4 growth factor receptors and their ligands, neuregulin-1 alpha, neuregulin-1 beta, and betacellulin, in normal endometrium and endometrial cancer. Clin. Cancer Res. 1999, 5, 2877–2883. [Google Scholar]
  142. Ejskjaer, K.; Sørensen, B.; Poulsen, S.; Mogensen, O.; Forman, A.; Nexø, E. Expression of the epidermal growth factor system in human endometrium during the menstrual cycle. Mol. Hum. Reprod. 2005, 11, 543–551. [Google Scholar] [CrossRef]
  143. Ejskjaer, K.; Sorensen, B.S.; Poulsen, S.S.; Forman, A.; Nexo, E.; Mogensen, O. Expression of the epidermal growth factor system in endometrioid endometrial cancer. Gynecol. Oncol. 2007, 104, 158–167. [Google Scholar] [CrossRef]
  144. Brys, M.; Semczuk, A.; Rechberger, T.; Krajewska, W.M. Expression of erbB-1 and erbB-2 genes in normal and pathological human endometrium. Oncol. Rep. 2007, 18, 261–265. [Google Scholar] [CrossRef]
  145. Reinartz, J.; George, E.; Lindgren, B.; Niehans, G. Expression of p53, transforming growth factor alpha, epidermal growth factor receptor, and c-erbB-2 in endometrial carcinoma and correlation with survival and known predictors of survival. Hum. Pathol. 1994, 25, 1075–1083. [Google Scholar] [CrossRef]
  146. Khalifa, M.A.; Mannel, R.S.; Haraway, S.D.; Walker, J.; Min, K.-W. Expression of EGFR, HER-2/neu, P53, and PCNA in Endometrioid, Serous Papillary, and Clear Cell Endometrial Adenocarcinomas. Gynecol. Oncol. 1994, 53, 84–92. [Google Scholar] [CrossRef]
  147. Scambia, G.; Panici, P.B.; Ferrandina, G.; Battaglia, F.; Distefano, M.; D'Andrea, G.; De Vincenzo, R.; Maneschi, F.; Ranelletti, F.O.; Mancuso, S. Significance of epidermal growth factor receptor expression in primary human endometrial cancer. Int. J. Cancer 2007, 56, 26–30. [Google Scholar] [CrossRef] [PubMed]
  148. Niikura, H.; Sasano, H.; Kaga, K.; Sato, S.; Yajima, A. Expression of epidermal growth factor family proteins and epidermal growth factor receptor in human endometrium. Hum. Pathol. 1996, 27, 282–289. [Google Scholar] [CrossRef] [PubMed]
  149. Konecny, G.E.; Santos, L.; Winterhoff, B.; Hatmal, M.; Keeney, G.L.; Mariani, A.; Jones, M.; Neuper, C.; Thomas, B.; Muderspach, L.; et al. HER2 gene amplification and EGFR expression in a large cohort of surgically staged patients with nonendometrioid (type II) endometrial cancer. Br. J. Cancer 2008, 100, 89–95. [Google Scholar] [CrossRef]
  150. Adonakis, G.; Androutsopoulos, G.; Koumoundourou, D.; Liava, A.; Ravazoula, P.; Kourounis, G. Expression of the epidermal growth factor system in endometrial cancer. Eur. J. Gynaecol. Oncol. 2008, 29, 450–454. [Google Scholar]
  151. Adonakis, G.; Androutsopoulos, G. The role of ErbB receptors in endometrial cancer. In Cancer of the Uterine Endometrium—Advances and Controversies; Saldivar, J.S., Ed.; IntechOpen: London, UK, 2012; pp. 23–38. [Google Scholar]
  152. Androutsopoulos, G.; Adonakis, G.; Gkermpesi, M.; Gkogkos, P.; Ravazoula, P.; Kourounis, G. Expression of the epidermal growth factor system in endometrial cancer after adjuvant tamoxifen treatment for breast cancer. Eur. J. Gynaecol. Oncol. 2006, 27, 490–494. [Google Scholar]
  153. Reyes, H.D.; Thiel, K.W.; Carlson, M.J.; Meng, X.; Yang, S.; Stephan, J.-M.; Leslie, K.K. Comprehensive profiling of EGFR/HER receptors for personalized treatment of gynecologic cancers. Mol. Diagn. Ther. 2014, 18, 137–151. [Google Scholar] [CrossRef]
  154. Androutsopoulos, G.; Michail, G.; Adonakis, G.; Decavalas, G. Molecular mechanisms, expression and clinical role of ErbB receptors in endometrial cancer. Int. J. Clin. Ther. Diagn. 2015, S1, 28–32. [Google Scholar]
  155. Michail, G.; Styliara, I.; Panas, P.; Markatos, F.; Koumoundourou, D.; Ravazoula, P.; Adonakis, G.; Androutsopoulos, G. EP472 ErbB receptors profile in non-selected patients with endometrial cancer. Int. J. Gynecol. Cancer 2019, 29, A298–A299. [Google Scholar]
  156. Styliara, I.; Zarogianni, E.; Panas, P.; Michail, G.; Koumoundourou, D.; Ravazoula, P.; Adonakis, G.; Androutsopoulos, G. 299 EGF System receptors profiling in various histologic subgroups of endometrial cancer. Eur. J. Obstet. Gynecol. Reprod. Biol. 2022, 270, e85–e86. [Google Scholar] [CrossRef]
  157. Androutsopoulos, G.; Adonakis, G.; Liava, A.; Ravazoula, P.; Decavalas, G. Expression and potential role of ErbB receptors in type II endometrial cancer. Eur. J. Obstet. Gynecol. Reprod. Biol. 2013, 168, 204–208. [Google Scholar] [CrossRef]
  158. Michail, G.; Panas, P.; Markatos, F.; Styliara, I.; Koumoundourou, D.; Ravazoula, P.; Adonakis, G.; Androutsopoulos, G. ErbB receptors profiling in selected patients with type II endometrial cancer. Int. J. Gynecol. Cancer 2019, 29 (Suppl. S4), A299. [Google Scholar]
  159. Zarogianni, E.; Panas, P.; Styliara, I.; Michail, G.; Koumoundourou, D.; Ravazoula, P.; Adonakis, G.; Androutsopoulos, G. EGF system receptors status in aggressive subtypes of endometrial cancer. Eur. J. Obstet. Gynecol. Reprod. Biol. 2022, 270, e86. [Google Scholar] [CrossRef]
  160. Morrison, C.; Zanagnolo, V.; Ramirez, N.; Cohn, D.; Kelbick, N.; Copeland, L.; Maxwell, G.; Fowler, J. HER-2 is an independent prognostic factor in endometrial cancer: Association with outcome in a large cohort of surgically staged patients. J. Clin. Oncol. 2006, 24, 2376–2385. [Google Scholar] [CrossRef]
  161. Engelsen, I.; Stefansson, I.; Beroukhim, R.; Sellers, W.; Meyerson, M.; Akslen, L.; Salvesen, H. HER-2/neu expression is associated with high tumor cell proliferation and aggressive phenotype in a population based patient series of endometrial carcinomas. Int. J. Oncol. 2008, 32, 307–316. [Google Scholar] [CrossRef]
  162. Coronado, P.; Vidart, J.; Lopez-asenjo, J.; Fasero, M.; Furio-bacete, V.; Magrina, J.; Escudero, M. P53 overexpression predicts endometrial carcinoma recurrence better than HER-2/neu overexpression. Eur. J. Obstet. Gynecol. Reprod. Biol. 2001, 98, 103–108. [Google Scholar] [CrossRef]
  163. Halperin, R.; Zehavi, S.; Habler, L.; Hadas, E.; Bukovsky, I.; Schneider, D. Comparative immunohistochemical study of endometrioid and serous papillary carcinoma of endometrium. Eur. J. Gynaecol. Oncol. 2001, 22, 122–126. [Google Scholar]
  164. Santin, A.; Bellone, S.; Siegel, E.; Palmieri, M.; Thomas, M.; Cannon, M.; Kay, H.; Roman, J.; Burnett, A.; Pecorelli, S. Racial differences in the overexpression of epidermal growth factor type II receptor (HER2/neu): A major prognostic indicator in uterine serous papillary cancer. Am. J. Obstet. Gynecol. 2005, 192, 813–818. [Google Scholar] [CrossRef]
  165. Santin, A.; Bellone, S.; Van Stedum, S.; Bushen, W.; Palmieri, M.; Siegel, E.; De Las Casas, L.; Roman, J.; Burnett, A.; Pecorelli, S. Amplification of c-erbB2 oncogene: A major prognostic indicator in uterine serous papillary carcinoma. Cancer 2005, 104, 1391–1397. [Google Scholar] [CrossRef]
  166. Slomovitz, B.; Broaddus, R.; Burke, T.; Sneige, N.; Soliman, P.; Wu, W.; Sun, C.; Munsell, M.; Gershenson, D.; Lu, K. Her-2/neu overexpression and amplification in uterine papillary serous carcinoma. J. Clin. Oncol. 2004, 22, 3126–3132. [Google Scholar] [CrossRef]
  167. Grushko, T.; Filiaci, V.; Mundt, A.; Ridderstrale, K.; Olopade, O.; Fleming, G. An exploratory analysis of HER-2 amplification and overexpression in advanced endometrial carcinoma: A Gynecologic Oncology Group study. Gynecol. Oncol. 2008, 108, 3–9. [Google Scholar] [CrossRef]
  168. Lukes, A.; Kohler, M.; Pieper, C.; Kerns, B.; Bentley, R.; Rodriguez, G.; Soper, J.; Clarke-Pearson, D.; Bast, R., Jr.; Berchuck, A. Multivariable analysis of DNA ploidy, p53, and HER-2/neu as prognostic factors in endometrial cancer. Cancer 1994, 73, 2380–2385. [Google Scholar] [CrossRef]
  169. Odicino, F.; Bignotti, E.; Rossi, E.; Pasinetti, B.; Tassi, R.; Donzelli, C.; Falchetti, M.; Fontana, P.; Grigolato, P.; Pecorelli, S. HER-2/neu overexpression and amplification in uterine serous papillary carcinoma: Comparative analysis of immunohistochemistry, real-time reverse transcription-polymerase chain reaction, and fluorescence in situ hybridization. Int. J. Gynecol. Cancer 2008, 18, 14–21. [Google Scholar] [CrossRef]
  170. Togami, S.; Sasajima, Y.; Oi, T.; Ishikawa, M.; Onda, T.; Ikeda, S.; Kato, T.; Tsuda, H.; Kasamatsu, T. Clinicopathological and prognostic impact of human epidermal growth factor receptor type 2 (HER2) and hormone receptor expression in uterine papillary serous carcinoma. Cancer Sci. 2012, 103, 926–932. [Google Scholar] [CrossRef]
  171. Díaz-Montes, T.; Ji, H.; Smith Sehdev, A.; Zahurak, M.; Kurman, R.; Armstrong, D.; Bristow, R. Clinical significance of Her-2/neu overexpression in uterine serous carcinoma. Gynecol. Oncol. 2006, 100, 139–144. [Google Scholar] [CrossRef]
  172. Androutsopoulos, G.; Styliara, I.; Zarogianni, E.; Michail, G.; Adonakis, G. Is it time to reconsider the clinical role of ErbB targeted therapy in endometrial cancer? In Endometrial Cancer; Farghaly, S., Ed.; Nova Science Publishers: Hauppauge, NY, USA, 2022; pp. 299–327. [Google Scholar]
  173. Villella, J.; Cohen, S.; Smith, D.; Hibshoosh, H.; Hershman, D. HER-2/neu overexpression in uterine papillary serous cancers and its possible therapeutic implications. Int. J. Gynecol. Cancer 2006, 16, 1897–1902. [Google Scholar] [CrossRef]
  174. Santin, A.; Bellone, S.; Roman, J.; McKenney, J.; Pecorelli, S. Trastuzumab treatment in patients with advanced or recurrent endometrial carcinoma overexpressing HER2/neu. Int. J. Gynaecol. Obstet. 2008, 102, 128–131. [Google Scholar] [CrossRef]
  175. Elsahwi, K.; Santin, A. ErbB2 overexpression in uterine serous cancer: A molecular target for trastuzumab therapy. Obstet. Gynecol. Int. 2011, 2011, 128295. [Google Scholar] [CrossRef]
  176. Androutsopoulos, G.; Michail, G.; Adonakis, G.; Decavalas, G. Molecular biology, expression and clinical significance of ErbB receptors in endometrial cancer. Hell. J. Obst. Gynecol. 2014, 13, 77–83. [Google Scholar]
  177. Fader, A.; Roque, D.; Siegel, E.; Buza, N.; Hui, P.; Abdelghany, O.; Chambers, S.; Secord, A.; Havrilesky, L.; O’Malley, D.; et al. Randomized Phase II Trial of Carboplatin-Paclitaxel Versus Carboplatin-Paclitaxel-Trastuzumab in Uterine Serous Carcinomas That Overexpress Human Epidermal Growth Factor Receptor 2/neu. J. Clin. Oncol. 2018, 36, 2044–2051. [Google Scholar] [CrossRef]
  178. Fader, A.; Roque, D.; Siegel, E.; Buza, N.; Hui, P.; Abdelghany, O.; Chambers, S.; Secord, A.; Havrilesky, L.; O’Malley, D.; et al. Randomized Phase II Trial of Carboplatin-Paclitaxel Compared with Carboplatin-Paclitaxel-Trastuzumab in Advanced (Stage III-IV) or Recurrent Uterine Serous Carcinomas that Overexpress Her2/Neu (NCT01367002): Updated Overall Survival Analysis. Clin. Cancer Res. 2020, 26, 3928–3935. [Google Scholar] [CrossRef]
Figure 1. Schematic structure of ErbB receptors. EC domain: extracellular ligand-binding domain. TM domain: transmembrane domain. JM section: juxtamembrane section. K domain: intracellular bilobed tyrosine kinase domain. CT tail: tyrosine-containing C-terminal tail.
Figure 1. Schematic structure of ErbB receptors. EC domain: extracellular ligand-binding domain. TM domain: transmembrane domain. JM section: juxtamembrane section. K domain: intracellular bilobed tyrosine kinase domain. CT tail: tyrosine-containing C-terminal tail.
Epigenomes 07 00024 g001
Figure 2. ErbB receptors’ signaling pathways.
Figure 2. ErbB receptors’ signaling pathways.
Epigenomes 07 00024 g002
Table 1. ErbB ligands and their affinity for ErbB receptors.
Table 1. ErbB ligands and their affinity for ErbB receptors.
ErbB-1ErbB-2ErbB-3ErbB-4
EGF+---
TGF-a+---
Amphiregulin+---
HB-EGF+--+
Betacellulin+--+
Epigen+--+
Epiregulin+--+
Neuregulin-1--++
Neuregulin-2--++
Neuregulin-3---+
Neuregulin-4---+
Neuroglycan C--+-
Tomoregulin---+
+ means positive, while - means negative.
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

Androutsopoulos, G.; Styliara, I.; Zarogianni, E.; Lazurko, N.; Valasoulis, G.; Michail, G.; Adonakis, G. The ErbB Signaling Network and Its Potential Role in Endometrial Cancer. Epigenomes 2023, 7, 24. https://doi.org/10.3390/epigenomes7040024

AMA Style

Androutsopoulos G, Styliara I, Zarogianni E, Lazurko N, Valasoulis G, Michail G, Adonakis G. The ErbB Signaling Network and Its Potential Role in Endometrial Cancer. Epigenomes. 2023; 7(4):24. https://doi.org/10.3390/epigenomes7040024

Chicago/Turabian Style

Androutsopoulos, Georgios, Ioanna Styliara, Evgenia Zarogianni, Nadia Lazurko, George Valasoulis, Georgios Michail, and Georgios Adonakis. 2023. "The ErbB Signaling Network and Its Potential Role in Endometrial Cancer" Epigenomes 7, no. 4: 24. https://doi.org/10.3390/epigenomes7040024

Article Metrics

Back to TopTop