Next Article in Journal
Deep Convolutional Framelets for Dose Reconstruction in Boron Neutron Capture Therapy with Compton Camera Detector
Next Article in Special Issue
Correlation Between Intravascular Platelet Aggregation in Tumors and Hypoxia-Inducible Factor 1 Alpha Expression in Epithelial Ovarian Cancer: Implications for Prognosis and Staging
Previous Article in Journal
Challenging the Binary Classification of Endometrioid Endometrial Adenocarcinoma: The Evaluation of Grade 2 as an Independent Entity Based on Prognostic Characteristics and Recurrence Patterns
Previous Article in Special Issue
Expression of CK17 and SOX2 in Vulvar Intraepithelial Neoplasia: A Comprehensive Analysis of 150 Vulvar Lesions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Insulin-like Growth Factor 1 (IGF1) and Its Isoforms: Insights into the Mechanisms of Endometrial Cancer

by
Abdul Muzhill Hannaan Abdul Hafizz
1,*,
Norfilza Mohd Mokthar
2,
Reena Rahayu Md Zin
3,
Nigel P. Mongan
4,
Mohd Nazzary Mamat @ Yusof
1,
Nirmala Chandralega Kampan
1,
Kah Teik Chew
1 and
Mohamad Nasir Shafiee
1,*
1
Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
2
Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
3
Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
4
Biodiscovery Institute, Faculty of Medicine and Health Sciences, The University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK
*
Authors to whom correspondence should be addressed.
Cancers 2025, 17(1), 129; https://doi.org/10.3390/cancers17010129
Submission received: 4 December 2024 / Revised: 25 December 2024 / Accepted: 1 January 2025 / Published: 3 January 2025

Simple Summary

Metabolic conditions such as obesity, diabetes, and insulin resistance, combined with hormonal imbalances, can disrupt the body’s natural growth regulation systems, including the insulin-like growth factor-1 (IGF1) pathway. These disruptions significantly increase the risk of endometrial cancer (EC), a cancer that develops in the lining of the uterus. This review delves into the roles of IGF1 and its variants; IGF1-Ea, IGF1-Eb, and IGF1-Ec, and their interactions with hormones like oestrogen in driving tumour growth and spread. By unravelling these connections, we aim to identify innovative strategies for preventing and treating this cancer, bringing hope for improved patient outcomes and advancing research in hormone-driven cancers.

Abstract

Endometrial cancer (EC) is a common gynaecological malignancy associated with metabolic dysfunctions such as obesity, diabetes and insulin resistance, as well as hormonal imbalances, particularly involving oestrogen and progesterone. These factors disrupt normal cellular metabolism, heightening the risk of developing endometrioid EC (EEC), the most prevalent subtype of EC. The insulin-like growth factor-1 (IGF1) pathway, a key regulator of growth, metabolism, and organ function, is implicated in EC progression. Recent research highlights the distinct roles of IGF1 isoforms, including IGF1-Ea, IGF1-Eb, and IGF1-Ec, in promoting tumour growth, metastasis, and hormone signalling interactions, particularly with oestrogen. This review examines the function and clinical significance of IGF-1 isoforms, emphasising their mechanisms in gynaecological physiology and their contributions to EC pathogenesis. Evidence from other cancers further underscores the relevance of IGF1 isoforms in driving tumour behaviours, offering valuable insights into their potential as biomarkers and therapeutic targets. Understanding these mechanisms provides opportunities for novel approaches to the prevention, diagnosis, and treatment of EC, improving patient outcomes and advancing the broader field of hormone-driven cancers.

1. Introduction

Endometrial cancer (EC) arises from the uterine lining and is influenced by multifaceted factors, including metabolic dysfunctions such as abdominal obesity, hyperglycaemia, hypertension, and atherogenic dyslipidaemia [1,2]. Visceral fat deposition disrupts insulinotropic and anti-inflammatory cytokines, leading to insulin resistance and heightened susceptibility to developing EC in postmenopausal women [1]. Hormonal imbalances significantly impact endometrial health, potentially increasing the risk of polycystic ovary syndrome (PCOS), endometriosis, and EC [2,3]. Molecular and hormonal variations contribute to EC aetiology, with unopposed oestrogen and endocrine system dysfunction being notable risk factors [4,5].
Recent studies have explored changes in circulating insulin, insulin-like growth factors (IGFs), insulin-like growth factor binding proteins (IGFBPs), inflammatory cytokines, adipokines, and sex hormones in EC [1,2,6,7]. IGF1 has been identified as a central hub gene, alongside others like CCNA2, CDK1, CCND1, FGF2, BCL2, and VEGFA, which are potential targets for gynaecological cancer prevention and early intervention [8]. These genes regulate critical intracellular signalling pathways, including the phosphatidylinositol 3-kinase (PI3K)/AKT and mitogen-activated protein kinase (MAPK) pathways, specifically the extracellular signal-regulated kinase (ERK) pathway, which trigger cancer development [7,8]. Building on these molecular insights, current therapeutic strategies for EC include surgery, chemotherapy, and radiotherapy [9]. Chemotherapy with agents like paclitaxel and carboplatin remains the standard approach, especially for advanced or recurrent cases [10,11]. Emerging therapies, such as IGF1R and mTOR inhibitors, target these oncogenic pathways, while hormonal therapies and immune checkpoint inhibitors like pembrolizumab are actively being studied [12,13,14,15,16]. These advancements emphasise the critical need to further investigate IGF1 isoforms, and this review explores their roles in EC pathogenesis and their potential as novel therapeutic targets.
Further research is warranted to explore epigenetic pathways involving microRNA and sex steroid profiling in EC [5]. Investigating these pathways is essential for understanding the interplay between metabolic and epigenetic factors in endocrine-related tumour development. These pathways may elucidate how behavioural and microenvironmental stimuli induce changes in cell function, thereby altering the functionality of IGF1 and its associated genes. Metabolic syndromes, hormonal imbalances, and sex steroid milieu have been linked to IGF1 signalling activation in endocrine-related tumour progression [6,17,18]. IGF1, ubiquitously expressed with complex metabolic regulatory roles, has emerged as a promising therapeutic target for cancer [18,19]. The objective of the review is to explore the association between IGF1 and its isoforms in the pathogenesis of EC, providing insights into potential novel targets for its treatment.

2. Insulin-like Growth Factor 1 (IGF1)

The mature IGF1 protein, also known as somatomedin C, consists of a 70-amino-acid single-chain basic polypeptide with a molecular weight of approximately 7.5 kDa. It exerts tissue-specific effects through the IGF1 receptor (IGF1R), playing a crucial role in controlling the IGF hormonal network. Human IGF1 is primarily produced in the liver and released into the bloodstream under the regulation of growth hormone (GH) [17,19]. Upon binding to the α subunit of IGF1R, the IGF1 ligand induces a conformational change, activating receptor tyrosine kinase activity [18]. Phosphorylation of IGF1R further triggers signal transmission across the Ras(Rat Sarcoma)–Raf(Rapidly Accelerated Fibrosarcoma)–MAPK and PI3K-AKT pathways, influencing processes such as cell proliferation, differentiation, apoptosis, and carcinogenesis [20,21].
The GH-IGF1 signalling pathway plays a critical role in female reproductive physiology, impacting processes such as folliculogenesis, ovarian steroidogenesis, and embryo implantation [7]. Figure 1 illustrates how the pulsatile release of GH triggers the liver to produce and release IGF1, which then affects the female reproductive system. Cellular IGF1 expression extends beyond surface receptors, with emerging recognition of nuclear functions for both IGF1 and its receptor [19]. Investigating IGF1 genes and their isoforms across various contexts is essential to better understand their roles in diseases, including EC. IGF1R, a key player, regulates gene expression by modulating chromatin remodelling proteins and participating in DNA damage-tolerance mechanisms [19,22]. Further research is needed to explore the potential of specific antibodies for IGF1R internalisation and degradation.
As IGF1 alternative splicing (AS) has emerged as a potential therapeutic target in oncology, it is important to gain a deeper understanding of the associated signalling components. The IGF1 gene generates numerous isoforms, each potentially serving a distinct physiological function. This review elucidates the significance of IGF1 isoform expression in physiology and pathogenesis, in addition to the mechanisms by which IGF1 spliced variants contribute to tumour biology and their potential as a target in EC research.

3. The IGF1 and Cancer Development

As the central dogma of IGF1 applies to most human malignancies, the IGF1 hormonal network (including ligands, cell-surface receptors, and IGF-binding proteins) can react through different autocrine, paracrine, juxtracrine, or endocrine signalling pathways [18]. Recent findings have revealed that IGF1 is secreted locally by various cell types, depending on cancer cell behaviours rather than solely from the liver [18]. Additionally, alterations in its receptor (IGF1R) have attracted significant attention in malignant cells [19,22], with IGF1R overexpression and AKT pathway activation linked to early stages of endometrial carcinogenesis in hyperplastic endometrium [23]. Interest in understanding the biological control linked with IGF1 has been driven by both theoretical and practical elements of clinical oncology. Dysregulation of the IGF1 pathway is associated with malignancies, including less common types [7]. Epidemiological studies consistently link elevated circulating IGF1 levels to primary malignancies such as lung, breast, colorectal, ovarian, and EC [6,7,18,24,25]. Our comprehension of IGF1 regulation in gynaecological cancer biology is still lacking. Christopoulos et al. (2015) have brought attention to the diverse expression patterns of IGF1 isoforms across various types of cancer in recent studies [26]. The diverse expression patterns of IGF1 isoforms in different types of cancer are significant, as IGF1 is moderately linked with an increased risk of total cancer in both men and women, with divergent associations observed for specific cancer types, as highlighted by Qian and Huo (2020) [24]. These isoforms may impact the IGF1 signalling pathway, systemic circulation, and organ-specific interactions. To deepen our understanding, it is imperative to investigate the mechanisms governing local IGF1 synthesis and its isoforms within the cellular microenvironment. Understanding these complexities will contribute to advancing cancer research and clinical practice.

4. Therapeutic Potential and Challenges of IGF1 Targeting in Cancer

The IGF1 signalling pathway is a vital modulator of cancer progression, including EC, rendering it a compelling therapeutic target. Preclinical studies demonstrate that IGF1R inhibitors decrease tumour development, promote apoptosis, and interfere with critical pathways such as PI3K/AKT and MAPK/ERK, which are essential for tumour survival and proliferation [27,28,29]. Moreover, combination treatments that target IGF1R alongside agents like mTOR inhibitors have demonstrated enhanced efficiency in preclinical models, suggesting potential strategies to improve treatment outcomes [21,28]. However, clinical trials investigating IGF1R inhibitors, such as figitumumab and ganitumab, have produced mixed results, with modest responses and significant metabolic side-effects, including hyperglycaemia, due to IGF’s role in glucose metabolism [30,31,32].
Several challenges hinder the clinical success of IGF1-targetted therapies. The structural similarity between IGF1R and the insulin receptor (INSR) leads to off-target effects, exacerbating metabolic issues [18,32]. Furthermore, cancer cells often activate compensatory pathways, such as HER2 or RAS/MAPK signalling, in response to IGF1R inhibition, diminishing therapeutic efficiency [33]. The heterogeneity of EC, including variations in IGF1 expression across tumour subtypes, further complicates the development of effective therapies and underscores the need for predictive biomarkers to identify patients most likely to benefit from IGF1-targeted treatments [34,35].
The tumour microenvironment (TME), particularly hypoxia, significantly influences IGF1 signalling, posing challenges in cancer therapy [36,37]. Hypoxia-inducible factors (HIFs) activate pathways such as PI3K/AKT and MAPK, driving tumour progression and resistance to treatment [36,38]. IGF1 has been shown to respond to hypoxic conditions, contributing to extracellular matrix remodelling, tumour invasion, and metastasis [37]. Additionally, hypoxia enhances IGF1-mediated VEGF expression, promoting angiogenesis and sustaining tumour growth [39]. While the role of IGF1 isoforms in hypoxic conditions remains unexplored, future research could investigate their potential contributions to tumour progression and therapeutic resistance in the TME, offering new insights into cancer biology.
Future research should focus on developing highly selective IGF1R inhibitors to minimise off-target effects, as well as exploring combination therapies to counteract compensatory mechanisms. Incorporating biomarkers for patient stratification will enhance therapeutic precision, ensuring that treatments are tailored to the molecular characteristics of individual tumours. Furthermore, understanding the distinct roles of IGF1 isoforms in tumour biology may provide insight into optimising IGF1-targeted therapies overcoming resistance mechanisms.

5. Circulating IGF1 Levels and EC Development

The relationship between circulating IGF1 and EC risk remains contentious, with studies yielding contradictory results. Ayabe’s research team initially suggested that elevated IGF1 and reduced IGFBP1 levels are associated with an increased risk of postmenopausal EC, highlighting IGF1′s potential role in carcinogenesis [40]. Conversely, Petridou et al. (2003) linked EC to elevated IGF2 and decreased IGF1 levels, adding complexity to the narrative [41]. Gunter et al. (2008) further nuanced this understanding by identifying an inverse association between IGF1 and EEC, particularly in obese women [42]. Despite these findings, several studies have reported no significant differences in serum IGF1 concentrations between EC patients and controls [43,44,45]. McGrath et al. (2011) did not find any impact of IGF1 genetic variability on EC occurrence but noted an inverse association of IGF2 with EC risk, contradicting the earlier findings of Petridou et al. (2003) [41,46]. In addition, free IGF1 plasma levels were significantly lower in EC patients with myometrial invasion compared to those without myometrial invasion [47]. This array of conflicting results underscores the complexity of the IGF system in EC pathogenesis and indicates the need for further research to elucidate these relationships. While conflicting results exist regarding the role of IGF1 in EC development, this review delves into its potential mechanisms and contexts.
Impaired glucose tolerance and hyperinsulinaemia are also implicated in increasing EC risk among women [1,2,48]. Notably, women with type II diabetes exhibit significantly higher serum IGF1 levels compared to the general population [49]. Ethnic differences, hormonal factors, and the effects of hormone replacement therapy (HRT) further complicate this relationship. Some studies have related the elevated IGF1 serum with different ethnic in their studies [50,51]. Nevertheless, some of the cited above-mentioned studies failed to account for hormone replacement therapy (HRT) or hormonal contraceptive use. For instance, oral HRT has been shown to significantly reduce serum IGF1 levels, likely due to the direct inhibition of IGF1 production in the liver by orally administered oestrogen [17,52,53]. In addition, elevated serum IGF1 was also reported in PCOS, which significantly increased the EC risk [6]. Despite these insights, the precise mechanisms through which IGF1 contributes to endometrial carcinogenesis remain unclear, necessitating further research to elucidate these pathways.

6. Alternative Splicing (AS)—A Phenomenon in Physiology and Pathophysiology in EC

Alternative splicing (AS) has broad implications across disciplines. In medicine, understanding AS regulation sheds light on physiological and pathophysiological mechanisms, aiding diagnostic marker identification and therapeutic target discovery [54,55]. Aberrant AS events are linked to cancer development, but the impact of genetic variants on EC risk remains uncertain. AS generates distinct protein isoforms, modulating protein function in various physiological conditions [55]. Complex interactions between spliceosomal machinery and regulatory components, including splicing enhancers and silencers, contribute to proteome diversity [54,55]. They play a vital role in enhancing the functional diversity of the proteome by enabling cells to generate proteins with unique structural and functional characteristics.
Pre-mRNA splicing, a critical process orchestrated by the spliceosome, is fundamental in maintaining cellular function and plays a pivotal role in disease and cancer progression. Dysregulation of splicing factors, including Splicing Factor 3B Subunit 1 (SF3B1), contributes to abnormal mature transcripts associated with tumorigenesis [56]. This highlights the importance of understanding splicing mechanisms at a tissue-specific level, as researchers are now delving into these mechanisms to uncover novel biomarkers for disease [57]. Recent studies on SP3B1 inhibitors and an AS signature model for EC prognosis provide valuable insights [56]. Aberrant AS of IGF1 isoforms may alter cellular functions, disrupting keys pathways like PI3K/AKT and MAPK/ERK, driving cell proliferation, survival, and migration in EC. These splicing events highlight the critical role of IGF1 isoforms in shaping the tumour microenvironment, as discussed in detail in the next section.

7. IGF1 Gene and Its Isoforms

The human IGF1 gene, located on chromosome 12 (12q22–12q24), comprises six exons and five long introns, with two transcriptional promoters (P1 and P2) [58]. As depicted in Figure 2, precursor IGF1 polypeptides include the signal peptide, mature IGF1, and the E peptide. Two classes of mRNA variation arise: class 1 transcripts initiate from exon 1 (P1), while class 2 transcripts use exon 2 (P2). Post-translational modifications, including proteolytic processing and glycosylation, yield diverse pre-pro-peptide transcript variants [58]. Exons 3 and 4 encode the conserved mature IGF1 peptide. Exon 3 contributes to the signal peptide, while exon 4 includes part of the mature peptide and the E-amino-terminal region shared by all IGF1 mRNAs. Alternative splicing of exons 5 and 6 generates three carboxyl(C)-terminal E peptides and distinct 3′UTR: Ea (exon 6, the most abundant transcript), Eb (exon 5), and Ec (fragments of exons 5 and 6, also known as mechano-growth factor (MCF)).
The human IGF1 gene undergoes alternative splicing, resulting in three protein isoforms (pro-IGF1-A, pro-IGF1-B, and pro-IGF1-C). Post-translational cleavage eliminates the signal and E peptide, yielding mature IGF1 and E-peptides. These isoforms, secreted from cells, act as obligate ligands for IGF1R phosphorylation, driving mitogenic signalling in targeted cells. Their distinct stabilities, binding partners, and activities suggest tissue-specific roles [58]. Understanding the functional effects of IGF1 gene alternative splicing may reveal new molecular mechanisms of carcinogenesis and guide anti-cancer therapeutic strategies.

8. IGF1 Isoforms in Physiology

IGF1, a network of cellular and secreted proteins, acts as a potent mitogen with proliferative and anti-apoptotic effects. While liver hepatocytes are the primary source of IGF1 secretion, other organs also contribute to its biosynthesis. Notably, healthy mature hepatocytes express low levels of IGF1R compared to other tissues [18]. Physiologically, IGF1 isoforms are predominantly found in the liver, skeletal muscles, and adipose tissue [17]. However, the precise roles of local versus circulating IGF1 fractions remain incompletely understood.
Growing evidence highlights distinct roles for IGF1 isoforms (IGF1-Ea, IGF1-Eb, and IGF1-Ec) in skeletal muscle development, regeneration, and maintenance [58]. Following muscle damage, IGF1-Eb mRNA is initially upregulated, promoting myoblast proliferation, while subsequent IGF1-Ea upregulation is associated with myofiber differentiation [58,59]. These locally acting isoforms also exhibit neuroprotective properties and shield cardiomyocytes from stress [60]. Notably, IGF1-Ea mRNA prevails in multiple organs, suggesting an organ-specific regulatory mechanism [58]. Furthermore, the E-peptides may influence the cellular entry of mature IGF1, adding another layer of complexity to their function [61]. The regulation of IGF1 isoforms in systemic protection mechanisms, including tissue repair, may provide insights into their role in EC development.
The relative abundance of IGF1 isoforms may stem from variations in their maturation. The pro-receptor form of IGF1 undergoes proteolytic cleavage in the Golgi compartment, controlled by the convertase furin. Human muscle and liver protein lysates confirm that pro-IGF-1Ec can be cleaved to yield the free Ec peptide [62]. Notably, IGF1-Ec also plays a significant role in bone development and repair [63]. Post-translational modifications of the IGF1-Ec pro-peptide likely occur and are influenced by the surrounding cell microenvironment.
The existence of different IGF1 isoforms raises questions about their distinct biological activities and tissue-specific control of IGF1 production. Recent research suggests that IGF1-Ea and IGF1-Eb hold promise as therapeutic agents for preventing muscular weakness [64]. These isoforms enhance the autophagy/lysosome system, counteracting age-related changes, and increase Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-Aplha (PGC1-α) expression, regulating mitochondrial function and inflammation associated with ageing. Furthermore, IGF1-Ea’s modulation of endothelial-like tubular structures may have clinical implications for regenerative therapies [65].
The IGF1 system, which exhibits expression variations influenced by menstrual cycle phases, was initially described in endometrial function in 1998 [66]. Both IGF1 signalling and ERα-mediated responses are essential for a complete uterine response to oestrogen and growth [67]. While Milingos et al. (2011) reported IGF1 isoform expression in endometrial tissues, the current literature lacks data on isoform variability in normal endometrial physiology [68]. Future research on IGF1 isoforms, including their role during menstrual cycles, holds promise for improving clinical outcomes in gynaecological fields.

9. IGF1 Isoforms in Pathophysiology and Cancer Biology

The involvement of IGF1 signalling pathways in endocrinology, paediatrics, ageing, and oncology has captured researchers’ attention. Alterations in IGF1 isoforms may persistently impact the cellular milieu, potentially leading to mutations in target genes that initiate cancer development. The growth of abnormal cells may indeed be related to specific IGF1 isoforms. In cardiac remodelling, the delivery of IGF1-Ea pro-peptide has been shown to improve outcomes after myocardial infarction by enhancing cardiomyocyte survival, promoting regeneration, and modulating immune cell recruitment and cytokine expression [60].
The IGF1 isoforms exhibit diverse roles in various physiological contexts. IGF1-Eb is crucial for mitochondrial protein synthesis in skeletal muscle, and its decreased mRNA levels may contribute to impaired protein synthesis in obese individuals [69]. Meanwhile, IGF1-Ec emerges as a potential therapeutic target in osteosarcoma due to its distinct physiological role compared to IGF1 [63]. Notably, IGF1-Ec is independently associated with reduced skeletal muscle mass, BMI, and gender [69]. In endometriosis, all IGF1 isoforms are expressed in eutopic endometrium and ovarian endometrioma, but their levels significantly decrease in endometriotic cysts [70]. This reduction correlates with disease status and fibrotic tissue presence in late-stage endometriosis. Additionally, IGF1-Ec (MGF) expression is preferentially observed in glandular epithelial cells of ectopic endometrium, suggesting its role in tissue remodelling, fibrosis, and cellular proliferation [68]. These functions may contribute to the maintenance and progression of ectopic endometrial lesions, highlighting IGF1-Ec as a potential therapeutic target or biomarker for late-stage endometriosis.
Research spanning nearly two decades has linked IGF1 to the onset of various malignancies. Investigations into IGF1 mRNAs and pro-peptide variants aim to unravel their biological significance in carcinogenesis. Most examined cancers exhibit a prevalence of class I transcript expression over class II, although some studies report minor differences in colorectal cancer (CRC) [71,72]. Indeed, EC may aberrantly express each component of IGF1 isoforms. As a growing number of studies have sought to decipher the molecular mechanisms underlying the association of IGF1 systems with EC, the AS of IGF1 has emerged as the current area of cancer research interest. IGF1-Ea predominates in CRC and cervical cancer (CC), while cultured cell lines also show the presence of IGF1-Ea in EC, breast cancer (BC), and melanoma [26,71,72,73,74]. The other mRNA isoforms, IGF1-Eb and IGF1-Ec, and their corresponding pro-peptides may be overexpressed in different human cancers [26,58,75,76].
IGF1-Ec is believed to play a mitogenic role in tumours, supported by research on human cancer cell lines [26,58,72,75,76,77,78]. In contrast, the role of IGF1-Eb remains controversial, with conflicting evidence suggesting both anti-cancer and cancer-promoting effects [75,79]. Some studies report increased affinity of certain IGF1 isoforms to IGF1R, implicating them in cancer development [63,79]. However, contradictory findings indicate that IGF1 variants can act independently of IGF1R and INSR [26,58].
In BC, the hormonal status (Estrogen Receptor (ER)-positive and ER-negative) affects how IGF1-Ea and IGF1-Ec influence cell proliferation, progression, and resistance to therapy through interaction with ER, particularly in ER-positive cases [75,76] The absence of IGF1-Eb transcripts may enhance cancer cell aggressiveness, while studies debate its role as an anti-cancer factor by inhibiting BC cell growth, invasion, and angiogenesis [75], while in CC, human papillomaviruses (HPVs) interact with IGF axis components, potentially influencing spliceosome elements [74]. IGF1-Eb exhibits pro-proliferative activity in CC, with complex nucleolar/nuclear localisation [79,80]. Additionally, hepatocellular carcinoma involvement depends on factors like inflammation and hepatotropic virus infection, necessitating further research [81]. IGF1 isoforms also impact lung cancer (LC), osteosarcoma (OS), prostate cancer (PC), thyroid cancer (TC) and other neuroendocrine neoplasms where IGF1-Ec plays an oncogenic role by stimulating proliferation, progression through epithelial to mesenchymal transition (EMT), and metastasis via ERK1/ERK2 activation [26,58,74,75,77,82]. All the related findings of IGF1 isoforms in various cancer research are summarised in Table 1.

10. Potential Role of IGF1 Isoforms in the Biology of EC

Notably, the IGF1 level is not the only factor that activates the IGF1/IGF1R pathway. Although research on the effects of abnormal post-translational modifications in IGF1 precursors in EC is limited, these modifications have been reported to contribute to the activity of IGF1 [26,58,70,86]; the findings are summarised in Table 2. Stavropoulos et al. (2020) had retrospectively reported that IGF-1Ec immunohistochemical staining was significantly higher in 99 EC archival specimens compared to normal endometrium and endometrium hyperplasia (EH) (p < 0.05) [86]. IGF1-Ec has been observed to antagonise PTEN’s tumour-suppressive functions by activating downstream pathways such as PI3K/AKT and MAPK, which promote cell proliferation and inhibit apoptosis [86,87]. Similarly, IGF1 signalling can attenuate p53-medated cell cycle arrest and apoptosis, enhancing tumour growth [88]. Survivin, an apoptosis inhibitor, is upregulated by IGF1 signalling, contributing to therapy resistance and sustained tumour survival [89].
Nevertheless, this in vivo study exclusively identified IGF1-Ec isoforms and did not compare them to other IGF1 variants, while several studies only concerned the KLE cell line (non-endometrioid EC subtype), which did not represent the major subtype of EC [26,68]. Intriguingly, Christopoulos et al. (2015) demonstrated that the IGF1-Ea and IGF1-Ec isoforms are expressed at the highest levels in KLE cell lines compared to other cancer cell lines studied [26]. The distinct potential molecular functions of IGF1 and E peptides, particularly those of its various classes in EC, should be investigated, and their discovery may yield new insight into this hormone-dependent cancer pathogenesis, especially endometrioid subtype.

11. Interaction Between the IGF1 Isoforms and Oestrogen in EC

Recent research has focused on understanding the role of specific IGF1 isoforms in cancer biology, yet their involvement in the development of EC remains inconclusive. Since endometrial tissues express both the ER and progesterone receptor (PR), the uterine lining is extremely sensitive to hormone action, which plays a major role under various physiological and pathological conditions [18,90,91]. Unopposed oestrogen exposure is a well-established risk factor for EC, whereas growth factors and related peptides are believed to mediate and modulate the autocrine/paracrine actions of hormones on their target tissues [18,92]. Imbalances in the endocrine system, such as excess oestrogen, may accelerate oncogenesis, particularly in EEC [26]. The interaction between IGF1 and oestrogen has been extensively studied in reproductive tissues, revealing complex regulatory mechanisms that govern cellular functions [7].
Interestingly, administration of Estradiol-17β (E2) has been shown to elevate IGF1-Ea and IGF1-Eb mRNA levels in ovariectomised mice, with a more pronounced increase observed for IGF1-Eb [93]. This upregulation is mediated by ERα through Estrogen Response Element (ERE)-dependent gene regulation, which is crucial for the full uterine response to E2 [67]. In PC3 cell lines, a human prostate cancer model commonly used to study hormonal signalling pathways, E2 modulates the expression of IGF1-Ea and IGF1-Eb while concurrently downregulating IGF1-Ec [26]. Although originating from prostate cancer, this model offers valuable insights into oestrogen-regulated IGF1 isoform expression, potentially relevant to oestrogen-sensitive tissues like the endometrium. These findings suggest that oestrogen-induced changes in IGF1 isoform expression could significantly affect uterine function and aid in developing targeted therapies for EC, particularly the EEC subtype. Stavropoulos et al. (2020) emphasised that IGF1-Ec expression may vary depending on hormonal regulation and tissue type, further supporting its potential role in oestrogen-sensitive tissues like the endometrium [86]. Additionally, another study focusing on conditions like endometriosis underscored the dynamic interplay between IGF1 isoforms and oestrogen-sensitive tissues, shedding light on hormone-regulated pathways that may also influence EC progression and therapeutic approaches [94].

12. Future Perspectives

Given the increasing prevalence of EC, further investigations are warranted to elucidate the endocrine interplay between oestrogen and IGF1 isoforms. A comprehensive exploration of the GH/IGF1/IGF1 isoforms axis in oestrogen-dependent EC would shed light on the distinct contributions of various IGF1 isoforms to both normal endometrial tissue development and cancer progression. These insights could inform the development of targeted IGF1R therapies, considering the well-established antagonistic effects of IGF1 isoforms on IGF1 signalling. Moreover, unravelling the molecular mechanisms underlying EC via IGF1 isoforms may pave the way for innovative diagnostic and therapeutic strategies, particularly tailored to specific subgroups of the population.

13. Conclusions

Although numerous studies implicate IGF1 in EC development, the evidence is complex, with conflicting findings requiring further investigation. This multifactorial and complex disease contributed to contradictory findings, including methodological inconsistencies, variances in the molecular subtypes investigated, population genetic diversity, and cancer heterogeneity. Given the expanding evidence of the significance of IGF1 isoforms in different physiology and pathophysiology, discovering these tendencies in EC seems dubious and warrants further investigation. Understanding the mechanisms governing IGF1 isoform expression in EC is essential for developing a cohesive and coherent strategy that addresses preventing, diagnosing, and treating this disease. Emerging RNA-based strategies for the treatment of EC with abnormally alternative splicing isoforms merit investigation. Additional studies are still required to address the association between alternative splicing and EC in greater depth.

Author Contributions

Conceptualization, A.M.H.A.H., N.M.M., R.R.M.Z., N.P.M. and M.N.S.; methodology, A.M.H.A.H. and M.N.S.; resources, A.M.H.A.H., N.M.M., R.R.M.Z., N.P.M., N.C.K., M.N.M.@.Y. and M.N.S.; data curation, A.M.H.A.H. and M.N.M.@.Y.; writing—original draft preparation, A.M.H.A.H.; writing—review and editing, N.M.M., R.R.M.Z., N.P.M., M.N.M.@.Y., N.C.K., K.T.C. and M.N.S.; visualization, A.M.H.A.H.; supervision, N.M.M., R.R.M.Z., N.P.M. and M.N.S.; project administration, A.M.H.A.H. and M.N.S.; funding acquisition, M.N.S. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Ministry of Higher Education Malaysia through the Fundamental Research Grant Scheme (FRGS) under project code FRGS/1/2021/SKK01/UKM/01/1.

Institutional Review Board Statement

Not applicable. This study did not involve humans or animals and is a review of existing literature.

Informed Consent Statement

Not applicable. This study did not involve humans and is a review of existing literature.

Data Availability Statement

Not applicable. This study did not generate or analyse any new data and is based solely on a review of existing literature.

Acknowledgments

The authors would like to acknowledge the Ministry of Higher Education Malaysia for funding this study through the Fundamental Research Grant Scheme (FRGS) under project code FRGS/1/2021/SKK01/UKM/01/1.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Amin, M.N.; Hussain, M.S.; Sarwar, M.S.; Rahman Moghal, M.M.; Das, A.; Hossain, M.Z.; Chowdhury, J.A.; Millat, M.S.; Islam, M.S. How the association between obesity and inflammation may lead to insulin resistance and cancer. Diabetes Metab. Syndr. Clin. Res. Rev. 2019, 13, 1213–1224. [Google Scholar] [CrossRef] [PubMed]
  2. Shafiee, M.N.; Ortori, C.A.; Barrett, D.A.; Mongan, N.P.; Abu, J.; Atiomo, W. Lipidomic Biomarkers in Polycystic Ovary Syndrome and Endometrial Cancer. Int. J. Mol. Sci. 2020, 21, 4753. [Google Scholar] [CrossRef] [PubMed]
  3. Abdul Karim, A.K.; Shafiee, M.N.; Abd Aziz, N.H.; Omar, M.H.; Abdul Ghani, N.A.; Lim, P.S.; Md Zin, R.R.; Mokhtar, N. Reviewing the role of progesterone therapy in endometriosis. Gynecol. Endocrinol. 2019, 35, 10–16. [Google Scholar] [CrossRef] [PubMed]
  4. Mokhtar, N.M.; Ramzi, N.H.; Yin-Ling, W.; Rose, I.M.; Hatta Mohd Dali, A.Z.; Jamal, R. Laser capture microdissection with genome-wide expression profiling displayed gene expression signatures in endometrioid endometrial cancer. Cancer Investig. 2012, 30, 156–164. [Google Scholar] [CrossRef] [PubMed]
  5. Sidorkiewicz, I.; Jóźwik, M.; Niemira, M.; Krętowski, A. Insulin resistance and endometrial cancer: Emerging role for microRNA. Cancers 2020, 12, 2559. [Google Scholar] [CrossRef]
  6. Shafiee, M.N.; Seedhouse, C.; Mongan, N.; Chapman, C.; Deen, S.; Abu, J.; Atiomo, W. Up-regulation of genes involved in the insulin signalling pathway (IGF1, PTEN and IGFBP1) in the endometrium may link polycystic ovarian syndrome and endometrial cancer. Mol. Cell. Endocrinol. 2016, 424, 94–101. [Google Scholar] [CrossRef]
  7. Ipsa, E.; Cruzat, V.F.; Kagize, J.N.; Yovich, J.L.; Keane, K.N. Growth Hormone and Insulin-Like Growth Factor Action in Reproductive Tissues. Front. Endocrinol. 2019, 10, 777. [Google Scholar] [CrossRef]
  8. Liu, Y.; Yi, Y.; Wu, W.; Wu, K.; Zhang, W. Bioinformatics prediction and analysis of hub genes and pathways of three types of gynecological cancer. Oncol. Lett. 2019, 18, 617–628. [Google Scholar] [CrossRef] [PubMed]
  9. Kuhn, T.M.; Dhanani, S.; Ahmad, S. An Overview of Endometrial Cancer with Novel Therapeutic Strategies. Curr. Oncol. 2023, 30, 7904–7919. [Google Scholar] [CrossRef]
  10. Rubinstein, M.; Shen, S.; Monk, B.J.; Tan, D.S.P.; Nogueira-Rodrigues, A.; Aoki, D.; Sehouli, J.; Makker, V. Looking beyond carboplatin and paclitaxel for the treatment of advanced/recurrent endometrial cancer. Gynecol. Oncol. 2022, 167, 540–546. [Google Scholar] [CrossRef]
  11. Pignata, S.; Scambia, G.; Schettino, C.; Arenare, L.; Pisano, C.; Lombardi, D.; De Giorgi, U.; Andreetta, C.; Cinieri, S.; De Angelis, C.; et al. Carboplatin and paclitaxel plus avelumab compared with carboplatin and paclitaxel in advanced or recurrent endometrial cancer (MITO END-3): A multicentre, open-label, randomised, controlled, phase 2 trial. Lancet Oncol. 2023, 24, 286–296. [Google Scholar] [CrossRef]
  12. Oda, K.; Ikeda, Y.; Kawana, K.; Osuga, Y.; Fujii, T. mTOR Signaling in Endometrial Cancer: From a Molecular and Therapeutic Point of View. Curr. Obstet. Gynecol. Rep. 2015, 4, 1–10. [Google Scholar] [CrossRef]
  13. Werner, H.; Sarfstein, R.; Bruchim, I. Investigational IGF1R inhibitors in early stage clinical trials for cancer therapy. Expert. Opin. Investig. Drugs 2019, 28, 1101–1112. [Google Scholar] [CrossRef] [PubMed]
  14. Mamat @ Yusof, M.N.; Chew, K.T.; Hafizz, A.M.H.A.; Abd Azman, S.H.; Ab Razak, W.S.; Hamizan, M.R.u.; Kampan, N.C.; Shafiee, M.N. Efficacy and Safety of PD-1/PD-L1 Inhibitor as Single-Agent Immunotherapy in Endometrial Cancer: A Systematic Review and Meta-Analysis. Cancers 2023, 15, 4032. [Google Scholar] [CrossRef] [PubMed]
  15. Mamat @ Yusof, M.N.; Chew, K.T.; Kampan, N.C.; Shafiee, M.N. Expression of PD-1 and PD-L1 in Endometrial Cancer: Molecular and Clinical Significance. Int. J. Mol. Sci. 2023, 24, 15233. [Google Scholar] [CrossRef]
  16. Zhang, Y.; Gao, C.; Cao, F.; Wu, Y.; Chen, S.; Han, X.; Mo, J.; Qiu, Z.; Fan, W.; Zhou, P.; et al. Pan-Cancer Analysis of IGF-1 and IGF-1R as Potential Prognostic Biomarkers and Immunotherapy Targets. Front. Oncol. 2021, 11, 755341. [Google Scholar] [CrossRef] [PubMed]
  17. Kineman, R.D.; Del Rio-Moreno, M.; Sarmento-Cabral, A. 40 YEARS of IGF1: Understanding the tissue-specific roles of IGF1/IGF1R in regulating metabolism using the Cre/loxP system. J. Mol. Endocrinol. 2018, 61, T187–T198. [Google Scholar] [CrossRef] [PubMed]
  18. Werner, H. The IGF1 Signaling Pathway: From Basic Concepts to Therapeutic Opportunities. Int. J. Mol. Sci. 2023, 24, 14882. [Google Scholar] [CrossRef]
  19. Poreba, E.; Durzynska, J. Nuclear localization and actions of the insulin-like growth factor 1 (IGF-1) system components: Transcriptional regulation and DNA damage response. Mutat. Res. Rev. Mutat. Res. 2020, 784, 108307. [Google Scholar] [CrossRef] [PubMed]
  20. Forbes, B.E.; Blyth, A.J.; Wit, J.M. Disorders of IGFs and IGF-1R signaling pathways. Mol. Cell. Endocrinol. 2020, 518, 111035. [Google Scholar] [CrossRef] [PubMed]
  21. Stauffer, S.; Roth, J.S.; Hernandez, E.R.; Kowalczyk, J.T.; Sealover, N.E.; Hebron, K.E.; James, A.; Isanogle, K.A.; Riffle, L.A.; Ileva, L.; et al. Preclinical Therapeutic Efficacy of RAF/MEK/ERK and IGF1R/AKT/mTOR Inhibition in Neuroblastoma. Cancers 2024, 16, 2320. [Google Scholar] [CrossRef] [PubMed]
  22. Rieger, L.; O’Connor, R. Controlled Signaling—Insulin-Like Growth Factor Receptor Endocytosis and Presence at Intracellular Compartments. Front. Endocrinol. 2021, 11, 620013. [Google Scholar] [CrossRef]
  23. McCampbell, A.S.; Broaddus, R.R.; Loose, D.S.; Davies, P.J. Overexpression of the insulin-like growth factor I receptor and activation of the AKT pathway in hyperplastic endometrium. Clin. Cancer Res. 2006, 12, 6373–6378. [Google Scholar] [CrossRef]
  24. Qian, F.; Huo, D. Circulating Insulin-Like Growth Factor-1 and Risk of Total and 19 Site-Specific Cancers: Cohort Study Analyses from the UK Biobank. Cancer Epidemiol. Biomark. Prev. 2020, 29, 2332–2342. [Google Scholar] [CrossRef]
  25. Werner, H.; Laron, Z. Role of the GH-IGF1 system in progression of cancer. Mol. Cell. Endocrinol. 2020, 518, 111003. [Google Scholar] [CrossRef] [PubMed]
  26. Christopoulos, P.F.; Philippou, A.; Koutsilieris, M. Pattern of IGF-1 variants’ expression in human cancer cell lines using a novel q-RT-PCR approach. Anticancer Res. 2015, 35, 107–115. [Google Scholar] [PubMed]
  27. Zorea, J.; Prasad, M.; Cohen, L.; Li, N.; Schefzik, R.; Ghosh, S.; Rotblat, B.; Brors, B.; Elkabets, M. IGF1R upregulation confers resistance to isoform-specific inhibitors of PI3K in PIK3CA-driven ovarian cancer. Cell Death Dis. 2018, 9, 944. [Google Scholar] [CrossRef] [PubMed]
  28. Ayub, A.; Yip, W.K.; Seow, H.F. Dual treatments targeting IGF-1R, PI3K, mTORC or MEK synergize to inhibit cell growth, induce apoptosis, and arrest cell cycle at G1 phase in MDA-MB-231 cell line. Biomed. Pharmacother. 2015, 75, 40–50. [Google Scholar] [CrossRef] [PubMed]
  29. Wang, Y.; Yin, L.; Sun, X. CircRNA hsa_circ_0002577 accelerates endometrial cancer progression through activating IGF1R/PI3K/Akt pathway. J. Exp. Clin. Cancer Res. 2020, 39, 169. [Google Scholar] [CrossRef] [PubMed]
  30. Wang, P.; Mak, V.C.Y.; Cheung, L.W.T. Drugging IGF-1R in cancer: New insights and emerging opportunities. Genes Dis. 2023, 10, 199–211. [Google Scholar] [CrossRef] [PubMed]
  31. Galifi, C.A.; Wood, T.L. Insulin-like growth factor-1 receptor crosstalk with integrins, cadherins, and the tumor microenvironment: Sticking points in understanding IGF1R function in cancer. Endocr. Relat. Cancer 2023, 30, e230031. [Google Scholar] [CrossRef]
  32. Soni, U.K.; Jenny, L.; Hegde, R.S. IGF-1R targeting in cancer—does sub-cellular localization matter? J. Exp. Clin. Cancer Res. 2023, 42, 273. [Google Scholar] [CrossRef] [PubMed]
  33. Ianza, A.; Sirico, M.; Bernocchi, O.; Generali, D. Role of the IGF-1 Axis in Overcoming Resistance in Breast Cancer. Front. Cell Dev. Biol. 2021, 9, 641449. [Google Scholar] [CrossRef] [PubMed]
  34. Joehlin-Price, A.S.; Stephens, J.A.; Zhang, J.; Backes, F.J.; Cohn, D.E.; Suarez, A.A. Endometrial Cancer Insulin-like Growth Factor 1 Receptor (IGF1R) Expression Increases with Body Mass Index and Is Associated with Pathologic Extent and Prognosis. Cancer Epidemiol. Biomark. Prev. 2016, 25, 438–445. [Google Scholar] [CrossRef]
  35. Merritt, M.A.; Strickler, H.D.; Hutson, A.D.; Einstein, M.H.; Rohan, T.E.; Xue, X.; Sherman, M.E.; Brinton, L.A.; Yu, H.; Miller, D.S.; et al. Sex Hormones, Insulin, and Insulin-like Growth Factors in Recurrence of High-Stage Endometrial Cancer. Cancer Epidemiol. Biomark. Prev. 2021, 30, 719–726. [Google Scholar] [CrossRef] [PubMed]
  36. Roy, S.; Kumaravel, S.; Sharma, A.; Duran, C.L.; Bayless, K.J.; Chakraborty, S. Hypoxic tumor microenvironment: Implications for cancer therapy. Exp. Biol. Med. 2020, 245, 1073–1086. [Google Scholar] [CrossRef]
  37. Nwabo Kamdje, A.H.; Seke Etet, P.F.; Kipanyula, M.J.; Vecchio, L.; Tagne Simo, R.; Njamnshi, A.K.; Lukong, K.E.; Mimche, P.N. Insulin-like growth factor-1 signaling in the tumor microenvironment: Carcinogenesis, cancer drug resistance, and therapeutic potential. Front. Endocrinol. 2022, 13, 927390. [Google Scholar] [CrossRef] [PubMed]
  38. Wicks, E.E.; Semenza, G.L. Hypoxia-inducible factors: Cancer progression and clinical translation. J. Clin. Investig. 2022, 132, e159839. [Google Scholar] [CrossRef] [PubMed]
  39. Liu, Q.; Xu, Z.; Mao, S.; Chen, W.; Zeng, R.; Zhou, S.; Liu, J. Effect of hypoxia on hypoxia inducible factor-1α, insulin-like growth factor I and vascular endothelial growth factor expression in hepatocellular carcinoma HepG2 cells. Oncol. Lett. 2015, 9, 1142–1148. [Google Scholar] [CrossRef]
  40. Ayabe, T.; Tsutsumi, O.; Sakai, H.; Yoshikawa, H.; Yano, T.; Kurimoto, F.; Taketani, Y. Increased circulating levels of insulin-like growth factor-I and decreased circulating levels of insulin-like growth factor binding protein-1 in postmenopausal women with endometrial cancer. Endocr. J. 1997, 44, 419–424. [Google Scholar] [CrossRef] [PubMed]
  41. Petridou, E.; Koukoulomatis, P.; Alexe, D.M.; Voulgaris, Z.; Spanos, E.; Trichopoulos, D. Endometrial cancer and the IGF system: A case-control study in Greece. Oncology 2003, 64, 341–345. [Google Scholar] [CrossRef] [PubMed]
  42. Gunter, M.J.; Hoover, D.R.; Yu, H.; Wassertheil-Smoller, S.; Manson, J.E.; Li, J.; Harris, T.G.; Rohan, T.E.; Xue, X.; Ho, G.Y. A prospective evaluation of insulin and insulin-like growth factor-I as risk factors for endometrial cancer. Cancer Epidemiol. Biomark. Prev. 2008, 17, 921–929. [Google Scholar] [CrossRef] [PubMed]
  43. Lacey, J.V., Jr.; Potischman, N.; Madigan, M.P.; Berman, M.L.; Mortel, R.; Twiggs, L.B.; Barrett, R.J.; Wilbanks, G.D.; Lurain, J.R.; Fillmore, C.-M. Insulin-like growth factors, insulin-like growth factor-binding proteins, and endometrial cancer in postmenopausal women: Results from a US case-control study. Cancer Epidemiol. Biomark. Prev. 2004, 13, 607–612. [Google Scholar] [CrossRef]
  44. Lukanova, A.; Zeleniuch-Jacquotte, A.; Lundin, E.; Micheli, A.; Arslan, A.A.; Rinaldi, S.; Muti, P.; Lenner, P.; Koenig, K.L.; Biessy, C.; et al. Prediagnostic levels of C-peptide, IGF-I, IGFBP -1, -2 and -3 and risk of endometrial cancer. Int. J. Cancer 2004, 108, 262–268. [Google Scholar] [CrossRef]
  45. Weiderpass, E.; Brismar, K.; Bellocco, R.; Vainio, H.; Kaaks, R. Serum levels of insulin-like growth factor-I, IGF-binding protein 1 and 3, and insulin and endometrial cancer risk. Br. J. Cancer 2003, 89, 1697–1704. [Google Scholar] [CrossRef]
  46. McGrath, M.; Lee, I.M.; Buring, J.; De Vivo, I. Common genetic variation within IGFI, IGFII, IGFBP-1, and IGFBP-3 and endometrial cancer risk. Gynecol. Oncol. 2011, 120, 174–178. [Google Scholar] [CrossRef] [PubMed]
  47. Baloglu, A.; Bezircioglu, I.; Cetinkaya, B.; Hicyilmaz, L. Prospective clinical study of the association between plasma level of free IGF-1 and myometrial invasion min patients with endometrial adenocarcinoma. Ginekol. Pol. 2010, 81, 501–505. [Google Scholar] [PubMed]
  48. Rutanen, E.M.; Stenman, S.; Blum, W.; Kärkkäinen, T.; Lehtovirta, P.; Stenman, U.H. Relationship between carbohydrate metabolism and serum insulin-like growth factor system in postmenopausal women: Comparison of endometrial cancer patients with healthy controls. J. Clin. Endocrinol. Metab. 1993, 77, 199–204. [Google Scholar]
  49. Rajpathak, S.N.; Gunter, M.J.; Wylie-Rosett, J.; Ho, G.Y.; Kaplan, R.C.; Muzumdar, R.; Rohan, T.E.; Strickler, H.D. The role of insulin-like growth factor-I and its binding proteins in glucose homeostasis and type 2 diabetes. Diabetes Metab. Res. Rev. 2009, 25, 3–12. [Google Scholar] [CrossRef] [PubMed]
  50. Juul, A.; Scheike, T.; Pedersen, A.T.; Main, K.M.; Andersson, A.M.; Pedersen, L.M.; Skakkebaek, N.E. Changes in serum concentrations of growth hormone, insulin, insulin-like growth factor and insulin-like growth factor-binding proteins 1 and 3 and urinary growth hormone excretion during the menstrual cycle. Hum. Reprod. 1997, 12, 2123–2128. [Google Scholar] [CrossRef]
  51. Waksmański, B.; Dudkiewicz, J.; Dabrowski, S. Function of insulin-like growth factor (IGF-I) and its binding protein (IGFBP-1) in pathological proliferation of endometrium. Wiad. Lek. 2001, 54, 656–661. [Google Scholar] [PubMed]
  52. Trobec, K.; von Haehling, S.; Anker, S.D.; Lainscak, M. Growth hormone, insulin-like growth factor 1, and insulin signaling-a pharmacological target in body wasting and cachexia. J. Cachexia Sarcopenia Muscle 2011, 2, 191–200. [Google Scholar] [CrossRef] [PubMed]
  53. Al-Samerria, S.; Radovick, S. Exploring the Therapeutic Potential of Targeting GH and IGF-1 in the Management of Obesity: Insights from the Interplay between These Hormones and Metabolism. Int. J. Mol. Sci. 2023, 24, 9556. [Google Scholar] [CrossRef]
  54. Liu, Q.; Fang, L.; Wu, C. Alternative Splicing and Isoforms: From Mechanisms to Diseases. Genes 2022, 13, 401. [Google Scholar] [CrossRef]
  55. Climente-González, H.; Porta-Pardo, E.; Godzik, A.; Eyras, E. The functional impact of alternative splicing in cancer. Cell Rep. 2017, 20, 2215–2226. [Google Scholar] [CrossRef]
  56. Popli, P.; Richters, M.M.; Chadchan, S.B.; Kim, T.H.; Tycksen, E.; Griffith, O.; Thaker, P.H.; Griffith, M.; Kommagani, R. Splicing factor SF3B1 promotes endometrial cancer progression via regulating KSR2 RNA maturation. Cell Death Dis. 2020, 11, 842. [Google Scholar] [CrossRef] [PubMed]
  57. Liu, Z.; Yoshimi, A.; Wang, J.; Cho, H.; Chun-Wei Lee, S.; Ki, M.; Bitner, L.; Chu, T.; Shah, H.; Liu, B. Mutations in the RNA splicing factor SF3B1 promote tumorigenesis through MYC stabilization. Cancer Discov. 2020, 10, 806–821. [Google Scholar] [CrossRef]
  58. Philippou, A.; Maridaki, M.; Pneumaticos, S.; Koutsilieris, M. The complexity of the IGF1 gene splicing, posttranslational modification and bioactivity. Mol. Med. 2014, 20, 202–214. [Google Scholar] [CrossRef] [PubMed]
  59. Philippou, A.; Papageorgiou, E.; Bogdanis, G.; Halapas, A.; Sourla, A.; Maridaki, M.; Pissimissis, N.; Koutsilieris, M. Expression of IGF-1 isoforms after exercise-induced muscle damage in humans: Characterization of the MGF E peptide actions in vitro. Vivo 2009, 23, 567–575. [Google Scholar]
  60. Gallego-Colon, E.; Villalba, M.; Tonkin, J.; Cruz, F.; Bernal, J.A.; Jimenez-Borregureo, L.J.; Schneider, M.D.; Lara-Pezzi, E.; Rosenthal, N. Intravenous delivery of adeno-associated virus 9-encoded IGF-1Ea propeptide improves post-infarct cardiac remodelling. NPJ Regen. Med. 2016, 1, 16001. [Google Scholar] [CrossRef] [PubMed]
  61. Pfeffer, L.A.; Brisson, B.K.; Lei, H.; Barton, E.R. The insulin-like growth factor (IGF)-I E-peptides modulate cell entry of the mature IGF-I protein. Mol. Biol. Cell 2009, 20, 3810–3817. [Google Scholar] [CrossRef]
  62. Vassilakos, G.; Philippou, A.; Koutsilieris, M. Identification of the IGF-1 processing product human Ec/rodent Eb peptide in various tissues: Evidence for its differential regulation after exercise-induced muscle damage in humans. Growth Horm. IGF Res. 2017, 32, 22–28. [Google Scholar] [CrossRef]
  63. Armakolas, N.; Armakolas, A.; Antonopoulos, A.; Dimakakos, A.; Stathaki, M.; Koutsilieris, M. The role of the IGF-1 Ec in myoskeletal system and osteosarcoma pathophysiology. Crit. Rev. Oncol./Hematol. 2016, 108, 137–145. [Google Scholar] [CrossRef] [PubMed]
  64. Ascenzi, F.; Barberi, L.; Dobrowolny, G.; Villa Nova Bacurau, A.; Nicoletti, C.; Rizzuto, E.; Rosenthal, N.; Scicchitano, B.M.; Musarò, A. Effects of IGF-1 isoforms on muscle growth and sarcopenia. Aging Cell 2019, 18, e12954. [Google Scholar] [CrossRef] [PubMed]
  65. Santini, M.P.; Lexow, J.; Borsellino, G.; Slonimski, E.; Zarrinpashneh, E.; Poggioli, T.; Rosenthal, N. IGF-1Ea induces vessel formation after injury and mediates bone marrow and heart cross-talk through the expression of specific cytokines. Biochem. Biophys. Res. Commun. 2011, 410, 201–207. [Google Scholar] [CrossRef]
  66. Rutanen, E.M. Insulin-like growth factors in endometrial function. Gynecol. Endocrinol. 1998, 12, 399–406. [Google Scholar] [CrossRef] [PubMed]
  67. Hewitt, S.C.; Li, Y.; Li, L.; Korach, K.S. Estrogen-mediated Regulation of Igf1 Transcription and Uterine Growth Involves Direct Binding of Estrogen Receptor α to Estrogen-responsive Elements. J. Biol. Chem. 2010, 285, 2676–2685. [Google Scholar] [CrossRef] [PubMed]
  68. Milingos, D.S.; Philippou, A.; Armakolas, A.; Papageorgiou, E.; Sourla, A.; Protopapas, A.; Liapi, A.; Antsaklis, A.; Mastrominas, M.; Koutsilieris, M. Insulinlike Growth Factor-1Ec (MGF) Expression in Eutopic and Ectopic Endometrium: Characterization of the MGF E-Peptide Actions In Vitro. Mol. Med. 2011, 17, 21–28. [Google Scholar] [CrossRef] [PubMed]
  69. Bian, A.; Ma, Y.; Zhou, X.; Guo, Y.; Wang, W.; Zhang, Y.; Wang, X. Association between sarcopenia and levels of growth hormone and insulin-like growth factor-1 in the elderly. BMC Musculoskelet. Disord. 2020, 21, 214. [Google Scholar] [CrossRef] [PubMed]
  70. Milingos, D.; Katopodis, H.; Milingos, S.; Protopapas, A.; Creatsas, G.; Michalas, S.; Antsaklis, A.; Koutsilieris, M. Insulin-Like Growth Factor–1 Isoform mRNA Expression in Women with Endometriosis. Ann. NY. Acad. Sci. 2006, 1092, 434–439. [Google Scholar] [CrossRef]
  71. Kasprzak, A.; Szaflarski, W.; Szmeja, J.; Andrzejewska, M.; Przybyszewska, W.; Kaczmarek, E.; Koczorowska, M.; Kościński, T.; Zabel, M.; Drews, M. Differential expression of IGF-1 mRNA isoforms in colorectal carcinoma and normal colon tissue. Int. J. Oncol. 2013, 42, 305–316. [Google Scholar] [CrossRef] [PubMed]
  72. Kasprzak, A.; Szaflarski, W.; Szmeja, J.; Andrzejewska, M.; Przybyszewska, W.; Koczorowska, M.; Drews, M.; Kaczmarek, E. Expression of various insulin-like growth factor-1 mRNA isoforms in colorectal cancer. Contemp. Oncol. Współczesna Onkol. 2012, 16, 147–153. [Google Scholar] [CrossRef]
  73. Koczorowska, M.M.; Kwasniewska, A.; Gozdzicka-Jozefiak, A. IGF1 mRNA isoform expression in the cervix of HPV-positive women with pre-cancerous and cancer lesions. Exp. Ther. Med. 2011, 2, 149–156. [Google Scholar] [CrossRef] [PubMed]
  74. Durzyńska, J.; Barton, E. IGF expression in HPV-related and HPV-unrelated human cancer cells. Oncol. Rep. 2014, 32, 893–900. [Google Scholar] [CrossRef] [PubMed]
  75. De Santi, M.; Annibalini, G.; Barbieri, E.; Villarini, A.; Vallorani, L.; Contarelli, S.; Berrino, F.; Stocchi, V.; Brandi, G. Human IGF1 pro-forms induce breast cancer cell proliferation via the IGF1 receptor. Cell. Oncol. 2016, 39, 149–159. [Google Scholar] [CrossRef]
  76. Christopoulos, F.P.; Papageorgiou, E.; Petraki, C.; Koutsilieris, M. The COOH-terminus of the IGF-1Ec Isoform Enhances the Proliferation and Migration of Human MCF-7 Breast Cancer Cells. Anticancer Res. 2017, 37, 2899–2912. [Google Scholar] [PubMed]
  77. Alexandraki, K.I.; Philippou, A.; Boutzios, G.; Theohari, I.; Koutsilieris, M.; Delladetsima, I.K.; Kaltsas, G.A. IGF-IEc expression is increased in secondary compared to primary foci in neuroendocrine neoplasms. Oncotarget 2017, 8, 79003–79011. [Google Scholar] [CrossRef] [PubMed]
  78. Armakolas, A.; Kaparelou, M.; Dimakakos, A.; Papageorgiou, E.; Armakolas, N.; Antonopoulos, A.; Petraki, C.; Lekarakou, M.; Lelovas, P.; Stathaki, M.; et al. Oncogenic Role of the Ec Peptide of the IGF-1Ec Isoform in Prostate Cancer. Mol. Med. 2015, 21, 167–179. [Google Scholar] [CrossRef] [PubMed]
  79. Lo, J.H.; Chen, T.T. Production of bioactive recombinant human Eb-peptide of pro-IGF-I and identification of binding components from the plasma membrane of human breast cancer cells (MDA-MB-231). Exp. Cell Res. 2018, 362, 235–243. [Google Scholar] [CrossRef] [PubMed]
  80. Durzyńska, J. IGF axis and other factors in HPV-related and HPV-unrelated carcinogenesis (Review). Oncol. Rep. 2014, 32, 2295–2306. [Google Scholar] [CrossRef]
  81. Kasprzak, A.; Adamek, A.; Przybyszewska, W.; Pyda, P.; Szmeja, J.; Seraszek-Jaros, A.; Lanzafame, A.; Surdacka, A.; Mozer-Lisewska, I.; Koczorowska, M. Insulin-like growth factor-1 mRNA isoforms and insulin-like growth factor-1 receptor mRNA expression in chronic hepatitis C. World J. Gastroenterol. 2015, 21, 3867–3875. [Google Scholar] [CrossRef]
  82. Karagiannis, A.K.; Philippou, A.; Sofia, T.-B.; Zevolis, E.; Maria, T.N.; Tsopanomichalou-Glotsou, M.; Psarras, V.; Koutsilieris, M. IGF-IEc Expression Is Associated With Advanced Differentiated Thyroid Cancer. Anticancer Res. 2019, 39, 2811–2819. [Google Scholar] [CrossRef]
  83. Durzyńska, J.; Wardziński, A.; Koczorowska, M.; Goździcka-Józefiak, A.; Barton, E. Human Eb peptide: Not just a by-product of pre-pro-IGF1b processing? Horm. Metab. Res. 2013, 45, 415–422. [Google Scholar] [CrossRef] [PubMed]
  84. Argyro, S.; Constantina, P.; Pavlos, M.; Evangelia, D.; Ioannis, X.; Michael, K. IGF-IEc Expression Is Associated with Advanced Clinical and Pathological Stage of Prostate Cancer. Anticancer Res. 2013, 33, 2441–2445. [Google Scholar]
  85. Papageorgiou, E.; Philippou, A.; Armakolas, A.; Christopoulos, P.F.; Dimakakos, A.; Koutsilieris, M. The human Ec peptide: The active core of a progression growth factor with species-specific mode of action. Hormones 2016, 15, 423–434. [Google Scholar] [CrossRef]
  86. Stavropoulos, A.; Varras, M.; Philippou, A.; Vasilakaki, T.; Varra, V.-K.; Varra, F.-N.; Tsavari, A.; Lazaris, A.C.; Koutsilieris, M. Immunohistochemical expression of insulin-like growth factor-1Ec in primary endometrial carcinoma: Association with PTEN, p53 and survivin expression. Oncol. Lett. 2020, 20, 395. [Google Scholar] [CrossRef]
  87. Tao, Y.; Liang, B. PTEN mutation: A potential prognostic factor associated with immune infiltration in endometrial carcinoma. Pathol. Res. Pract. 2020, 216, 152943. [Google Scholar] [CrossRef] [PubMed]
  88. Duan, L.; Maki, C.G. The IGF-1R/AKT pathway determines cell fate in response to p53. Transl. Cancer. Res. 2016, 5, 664–675. [Google Scholar] [CrossRef]
  89. Liu, F.; Sun, Y.; Liu, B.; Lu, J.; Li, H.; Zhu, H.; Gao, H.; Zhou, X.; Chang, H. Insulin-like growth factor-1 induces epithelial-mesenchymal transition in hepatocellular carcinoma by activating survivin. Oncol. Rep. 2018, 40, 952–958. [Google Scholar] [CrossRef] [PubMed]
  90. Yang, S.; Thiel, K.W.; Leslie, K.K. Progesterone: The ultimate endometrial tumor suppressor. Trends Endocrinol. Metab. 2011, 22, 145–152. [Google Scholar] [CrossRef] [PubMed]
  91. Yang, X.; Wang, J. The Role of Metabolic Syndrome in Endometrial Cancer: A Review. Front. Oncol. 2019, 9, 744. [Google Scholar] [CrossRef]
  92. Ito, K.; Utsunomiya, H.; Yaegashi, N.; Sasano, H. Biological roles of estrogen and progesterone in human endometrial carcinoma—New developments in potential endocrine therapy for endometrial cancer. Endocr. J. 2007, 54, 667–679. [Google Scholar] [CrossRef] [PubMed]
  93. Ohtsuki, T.; Otsuki, M.; Murakami, Y.; Maekawa, T.; Yamamoto, T.; Akasaka, K.; Takeuchi, S.; Takahashi, S. Organ-specific and age-dependent expression of insulin-like growth factor-I (IGF-I) mRNA variants: IGF-IA and IB mRNAs in the mouse. Zool. Sci. 2005, 22, 1011–1021. [Google Scholar] [CrossRef]
  94. Blontzos, N.; Mavrogianni, D.; Ntzeros, K.; Kathopoulis, N.; Moustogiannis, A.; Philippou, A.; Koutsilieris, M.; Protopapas, A. Differential Expression of Insulin Growth Factor 1 (IGF-1) Isoforms in Different Types of Endometriosis: Preliminary Results of a Single-Center Study. Biomolecules 2023, 14, 7. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effect of GH and IGF1 secretion in relation to women’s reproductive system physiology. The pulsatile GH secretion stimulates the production and secretion of IGF1 from the liver, which is the primary source of circulating IGF1 and the GH/IGF1 effect.
Figure 1. Effect of GH and IGF1 secretion in relation to women’s reproductive system physiology. The pulsatile GH secretion stimulates the production and secretion of IGF1 from the liver, which is the primary source of circulating IGF1 and the GH/IGF1 effect.
Cancers 17 00129 g001
Figure 2. Schematic representation of the human IGF1 gene. The IGF1 gene consists of six exons (coloured boxes) separated by five introns. Exons 1 and 2 contain multiple transcription initiation sites. Translation initiation codons (AUG) are located at exons 1, 2, and 3. Exons 1, 2, and 3 encode the signal peptide of precursor IGF1 in the cytoplasm. Exons 5 and 6 generate three distinct isoforms: IGF1-Ea, IGF1-Eb, and IGF1-Ec.
Figure 2. Schematic representation of the human IGF1 gene. The IGF1 gene consists of six exons (coloured boxes) separated by five introns. Exons 1 and 2 contain multiple transcription initiation sites. Translation initiation codons (AUG) are located at exons 1, 2, and 3. Exons 1, 2, and 3 encode the signal peptide of precursor IGF1 in the cytoplasm. Exons 5 and 6 generate three distinct isoforms: IGF1-Ea, IGF1-Eb, and IGF1-Ec.
Cancers 17 00129 g002
Table 1. IGF1 isoform expression and significant findings in a variety of cancer types.
Table 1. IGF1 isoform expression and significant findings in a variety of cancer types.
Cancer TypeStudy TypeSummary of FindingsSource
Expression of IGF1 IsoformsSignificant Findings
IGF1-EaIGF1-EbIGF1-Ec
BCIn VitroCancers 17 00129 i001Cancers 17 00129 i002Cancers 17 00129 i001Anti-IGF1R fully inhibits IGF pro-forms, while anti-IGF1 partially inhibits their biological activity.[75]
In VitroCancers 17 00129 i001Cancers 17 00129 i002Cancers 17 00129 i001 *IGF1-Ec (hEc) enhances intracellular ERK1/2 pathway activity, promoting proliferation and migration in MCF7 cells.[76]
In VitroN/ACancers 17 00129 i001 *N/AIGF1-Eb (hEb) acts independently of IGF-IR and enhances its biological activity against cancer cells.[79]
CRCIn VivoCancers 17 00129 i001Cancers 17 00129 i002Cancers 17 00129 i001IGF1-Ea is quantitatively dominant, followed by IGF1-Eb and IGF1-Ec in CRC tissues. IGF1-Eb mRNA is significantly higher in the control large intestine compared to CRC.[72]
In VivoCancers 17 00129 i002Cancers 17 00129 i002Cancers 17 00129 i002All IGF1 transcripts exhibit lower expression in CRC samples compared to controls.[71]
In VitroCancers 17 00129 i001Cancers 17 00129 i001Cancers 17 00129 i002IGF1-Ea and IGF1-Eb isoforms exhibit similar high expression, while IGF1-Ec isoform expression is relatively low in DLD1 cell line.[26]
In VivoN/AN/ACancers 17 00129 i001IGF1-Ec expression is higher in specimens from metastatic sites compared to primary tumours (p = 0.024).[77]
CCIn VivoCancers 17 00129 i002Cancers 17 00129 i001=IGF1-Ea mRNA downregulated in both HPV-positive and HPV-negative cases compared to precancerous lesions (LSIL and HSIL with HPV-positive). IGF1-Eb is increased in CC.[73]
In VitroN/ACancers 17 00129 i001 *N/ASynthetic IGF1-Eb (hEb) promotes HeLa cell proliferation.[83]
In VitroDOCDOCDOCViral proteins impact IGF1 gene splicing and stabilise proteins in crucial cellular processes. IGF1 isoforms exhibit differential expression across various CC cell lines.[74]
In VitroCancers 17 00129 i001Cancers 17 00129 i001Cancers 17 00129 i001In HeLa cells, IGF1-Ea and IGF1-Eb expression levels were similar, while IGF1-Ec exhibited higher expression.[26]
In VivoN/AN/ACancers 17 00129 i001Positive cytoplasmic expression of IGF1-Ec in neuroendocrine uterine CC.[77]
HCCIn VitroCancers 17 00129 i002Cancers 17 00129 i002Cancers 17 00129 i002Overall IGF1 isoforms expression level lower in HepG2 cells compared to K562 cells, which might be influenced by HPV subtypes.[74]
In VivoCancers 17 00129 i002Cancers 17 00129 i002Cancers 17 00129 i001Hepatitis C virus (HCV) can alter the IGF1 splicing profile in HCC. IGF1-Ea and IGF1-Eb decreased in advance grade of HCC. [81]
In VitroCancers 17 00129 i002Cancers 17 00129 i002Cancers 17 00129 i002Overall IGF1 isoforms expression level lower in HuH7 cell lines. [26]
LCIn VitroCancers 17 00129 i001Cancers 17 00129 i001Cancers 17 00129 i001Cancers 17 00129 i001A549 cell lines exhibit elevated IGF1 isoform expression, with IGF1-Ea showing the highest levels, followed by IGF1-Ec and IGF1-Eb.[26]
In VivoN/AN/ACancers 17 00129 i001Positive cytoplasmic expression of IGF1-Ec in half of neuroendocrine LC cases.[77]
OSIn VitroN/ACancers 17 00129 i001 *N/ASynthetic hEb enhances cell growth and motility in stable U2OS cells.[83]
In VitroCancers 17 00129 i002Cancers 17 00129 i001Cancers 17 00129 i001IGF1-Eb is predominantly expressed in U2OS cells compared to other cell lines (HepG2, HeLa, K562)[74]
In VitroCancers 17 00129 i002Cancers 17 00129 i001Cancers 17 00129 i001Higher expression of IGF1-Eb than IGF1-Ea in MG63 cell lines.[26]
PCIn VivoN/AN/ACancers 17 00129 i001Cancers 17 00129 i001IGF1-Ec expression is higher in locally advanced tumours (stage > III).[84]
In Vivo and In VitroN/AN/ACancers 17 00129 i001IGF1-Ec overexpressed in advanced PC; leads to EMT through ERK1/2 pathway activation and ZEB1 expression.[78]
In VitroCancers 17 00129 i001Cancers 17 00129 i001Cancers 17 00129 i001Cancers 17 00129 i001IGF-1Ea and Eb are highly expressed in lnCaP cell lines; Ec peptide reduced under hormonal conditions (oestradiol, dexamethasone and GH- treatment).[26]
In Vivo and In VitroN/AN/ACancers 17 00129 i001Cancers 17 00129 i001 *Synthetic human Ec peptide (hEc) stimulates the human PC3 cell growth through activating ERK1/2 pathway, without affecting Akt phosphorylation. [85]
TCIn VivoN/AN/ACancers 17 00129 i001Cancers 17 00129 i001IGF1-Ec overexpressed in TC, and associated with TNM staging (prominently seen in more aggressive papillary TC) and capsule invasion of TC[82]
GCIn VivoN/AN/ACancers 17 00129 i001Cancers 17 00129 i001IGF1Ec peptide (MGF) was restricted to the tumour cell cytoplasm in gastric mucosa of GC cases. [77]
PPCIn VivoN/AN/ACancers 17 00129 i001Cancers 17 00129 i001IGF1Ec peptide (MGF) was restricted to the tumour cell cytoplasm in pancreatic tissue of PPC cases.[77]
SBCIn VivoN/AN/ACancers 17 00129 i001Cancers 17 00129 i001IGF1Ec peptide (MGF) was restricted to the tumour cell cytoplasm in columnar epithelium in SBC cases.[77]
MELIn VitroCancers 17 00129 i001Cancers 17 00129 i001Cancers 17 00129 i001Cancers 17 00129 i001IGF1-Ea is highly expressed in human melanoma SK-MEL28 cell lines, followed by IGF1Ec and IGF1-Eb. [26]
BC, Breast Cancer; CRC, Colorectal Cancer; CC, Epithelial Cervical Cancer; HCC, Hepatocellular Carcinoma; LC, Lung Cancer; OS, Osteosarcoma; PC, Prostate Cancer; TC, Thyroid Cancer; GC, Gastric Cancer; PCC, Pancreatic Cancer; SBC; Small Bowel Cancer; MEL, Melanoma; Cancers 17 00129 i001, Increase expression; Cancers 17 00129 i001Cancers 17 00129 i001, Overexpression; Cancers 17 00129 i002, Decrease expression; P, Data reported as present; N/A, No available data; *, Action by synthetic peptide; =, Expression level is similar trend (no difference with normal/control group); DOC, Depend on cell types (cell lines).
Table 2. IGF1 isoforms in EC research.
Table 2. IGF1 isoforms in EC research.
Study TypeSummary of FindingsSource
In VivoIGF1-Ec is overexpressed in non-endometrioid carcinoma (serous papillary or clear cell carcinoma) compared to EEC and is highly expressed in areas of tumoral necrosis. It may function oppositely to PTEN, a tumour suppressor gene, while promoting tumour growth via pathways similar to survivin in EC.[86]
In VitroIGF1-Ea and IGF1-Eb levels were found to be elevated in KLE cell line [26]
In VitroIGF1 isoforms are expressed in KLE cells, with IGF1-Ea being most abundant. IGF1-Ec peptides stimulate KLE cell growth independently of IGF1R and INSR.[68]
PTEN, Phosphatase and Tensin Homolog; EC, Endometrial Cancer; IGF1R, Insulin-like Growth Factor-1 Receptor; INSR, Insulin 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

Abdul Hafizz, A.M.H.; Mohd Mokthar, N.; Md Zin, R.R.; P. Mongan, N.; Mamat @ Yusof, M.N.; Kampan, N.C.; Chew, K.T.; Shafiee, M.N. Insulin-like Growth Factor 1 (IGF1) and Its Isoforms: Insights into the Mechanisms of Endometrial Cancer. Cancers 2025, 17, 129. https://doi.org/10.3390/cancers17010129

AMA Style

Abdul Hafizz AMH, Mohd Mokthar N, Md Zin RR, P. Mongan N, Mamat @ Yusof MN, Kampan NC, Chew KT, Shafiee MN. Insulin-like Growth Factor 1 (IGF1) and Its Isoforms: Insights into the Mechanisms of Endometrial Cancer. Cancers. 2025; 17(1):129. https://doi.org/10.3390/cancers17010129

Chicago/Turabian Style

Abdul Hafizz, Abdul Muzhill Hannaan, Norfilza Mohd Mokthar, Reena Rahayu Md Zin, Nigel P. Mongan, Mohd Nazzary Mamat @ Yusof, Nirmala Chandralega Kampan, Kah Teik Chew, and Mohamad Nasir Shafiee. 2025. "Insulin-like Growth Factor 1 (IGF1) and Its Isoforms: Insights into the Mechanisms of Endometrial Cancer" Cancers 17, no. 1: 129. https://doi.org/10.3390/cancers17010129

APA Style

Abdul Hafizz, A. M. H., Mohd Mokthar, N., Md Zin, R. R., P. Mongan, N., Mamat @ Yusof, M. N., Kampan, N. C., Chew, K. T., & Shafiee, M. N. (2025). Insulin-like Growth Factor 1 (IGF1) and Its Isoforms: Insights into the Mechanisms of Endometrial Cancer. Cancers, 17(1), 129. https://doi.org/10.3390/cancers17010129

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