Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Oct 1.
Published in final edited form as: Curr Opin Obstet Gynecol. 2020 Oct;32(5):361–370. doi: 10.1097/GCO.0000000000000650

Non-hormonal mediators of uterine fibroid growth.

Esra Cetin 1, Ayman Al-Hendy 1,2, Michał Ciebiera 1,3
PMCID: PMC8314923  NIHMSID: NIHMS1651302  PMID: 32739973

Abstract

Purpose of the review

Uterine fibroids (UFs) are the most common benign neoplasms of the female reproductive tract and one of the major public health concerns. Although most women with UFs are asymptomatic, over 30% of them will present with varying symptoms.

This review focuses on the role of non-hormonal mediators and pathways in UF biology. Furthermore, it provides data regarding the most recent findings in the field of compounds which use those non-hormonal pathways in the medical therapy of UFs.

Recent findings

Complex signaling pathway alterations are crucial for UF development. The topic of the pathophysiology of UFs focuses mostly on steroids and other hormones. However, other very important pathways exist which are independent of hormones. Some of the most important pathways which are non-hormonal, but in some cases still hormone-depended, include growth factors, cytokines and inflammation, Smad proteins, wingless type/β-catenin and others.

Summary

Much more is known about hormonal than about non-hormonal signaling in UFs. Growth factors, early life exposure and inflammation are key factors in UF biology. Numerous agents depend on those pathways and may find their place in the current and future therapy of UFs.

Keywords: uterine fibroid, leiomyoma, pathophysiology, biology, non-hormonal, mediator, therapy, pharmacology

Introduction

Uterine fibroids (UFs) are the most common benign neoplasms of the female reproductive tract and a major public health concern in reproductive age women. The overall incidence of those lesions is hard to determine since there are only few well performed studies. However, in some populations it may even reach 70% of women [1]. Although most women with UFs are asymptomatic, over 30% of them will present with various symptoms. UFs cause significant morbidity and may compromise daily activities, relationships or workplace performance [2, 3]. The overall quality of life among women with UFs is seriously impacted and it decreases with an increase in the number and severity of symptoms. Lesions that cause these kind of symptoms may often require a medical intervention [4, 5].

Despite their prevalence, not much is known about the reasons they occur and there are still more questions than answers [2, 6]. According to a recent systematic review by Stewart et al. (2017) the most important risk factors for UF are race (e.g. African American), older age, premenopausal state, hypertension, family history of UFs, nulliparity, time since last birth, and different food additives as well as soy [1]. Obesity and vitamin D deficiency are also key factors [7].

UFs are made of abnormal smooth uterine muscle cells and fibroblasts surrounded by extensive amount of extracellular matrix (ECM) including collagens, fibronectin, laminins, and proteoglycans [8, 9]. One of the most important things about UFs is their tendency to grow which, depending on the location, may cause clinical symptoms. Some authors indicated that large UFs grow slowly, while very small ones grow rapidly but with a higher chance of disappearance [10]. The UF life cycle may be divided into two steps – transformation and tumor formation [8, 11]. The transformation of normal uterine smooth muscle cells into abnormal ones occurs mostly through mutations in the mediator complex subunit 12 (MED12) gene [12] and the high mobility group AT-hook 2 (HMGA2) gene [8, 13]. Stem cells transform and grow into UFs mostly under the influence of hormones and the growth of the tumors occurs via massive cell expansion and ECM accumulation [6, 8, 9].

UF cells are dependent on stimulation, especially by estrogen and progesterone. Available data suggest that progesterone plays a more important role in their development [8, 14]. However, despite the data concerning steroids, we cannot disregard other important pathways involved in this process, as the hormones are not the only factor responsible for UF development [15, 16]. Data about those pathways and mediators are still incomplete. However, recent research has provided answers to several difficult questions. Signals forwarded by various growth factors, Smad protein, phosphatidylinositol-3-kinase and the mammalian target of rapamycin – PI3K/AKT/mTOR pathway, mitogen-activated protein kinases/extracellular signal-regulated kinases – MAPK/ERK pathway, wingless-type (Wnt)/β-catenin and retinoic acid pathways are involved in UF growth and proliferation indicating that they could serve as targets for possible therapies [9, 17, 18].

This review focuses on the role of non-hormonal mediators and pathways in UF biology. Furthermore, it provides data regarding the most recent findings in the field of compounds which use those non-hormonal pathways in the medical therapy of UFs.

Review of pathways

Non-hormonal pathways in uterine fibroids

Complex signaling pathway alterations are crucial for UF development, but the exact underlying biology of UFs is unclear. The topic of the pathophysiology of UFs focuses mostly on steroids and other hormones [8, 14, 19]. However, other very important pathways exist which are not only hormone-dependent. This review will add information about hormones only when necessary and we will focus on other pathways. These include:

  • growth factors,

  • cytokines and inflammation,

  • Smad proteins,

  • Wnt/β-catenin,

  • peroxisome proliferator-activated receptor (PPAR),

  • retinoic acid (RA).

The important finding for better understanding is that many of those pathways converge, link or overlap and incorporate input from several different directions [17].

Growth factors and related signaling

One of the main mechanisms of UF growth consists in the overexpression of cytokine-related genes and the increase of growth factor levels directly in the tumor [18, 20]. Their influence results in the activation of intracellular signal transduction pathways that modulate cell life. Growth factors are proteins that regulate numerous aspects of cellular function, including survival, proliferation, migration and differentiation. The term ‘growth factor’ is sometimes used by scientists interchangeably with the term ‘cytokine’ [20, 21]. Some growth factors that play a role in UF biology have been known for years. These are (in alphabetical order): epidermal growth factor (EGF) [22], fibroblast growth factor (FGF) [23], insulin-like growth factor (IGF) [24], platelet-derived growth factor (PDGF) [25], neuronal growth factor (NGF) [26], transforming growth factor (TGF) [18], and vascular endothelial growth factor (VEGF) [27]. Data concerning TGF-β in the pathophysiology of UFs are the most abundant, as it is believed to be one of the major links in both hormonal and non-hormonal pathways [18]. TGF-β3 was shown to be a key player in UFs with its role in cell proliferation and the deposition of the ECM [9, 18], which may serve as a reservoir of pro-fibrotic growth factors and as a stabilizer of their duration of signaling [9]. TGF-β-derived pathways are mostly connected with progesterone. However, many stages do not depend on steroids and they will be described in the present review.

Growth factors (e.g. TGF-β) bind to receptor tyrosine kinases (RTKs) causing the dimerization and phosphorylation of the receptor and activation of several pathways [17, 28]. RTKs are a big family of cell surface receptors that act with growth factors, hormones, neurotrophic factors and other extracellular signaling molecules [29]. According to Yu et al. ‘cross talk’ occurs between estrogens and RTK signaling pathways and estrogen upregulates RTKs in UFs [28]. Abnormalities that affect RTK signaling might lead to cell transformation and even to the onset of a malignant process [29]. RTK activation influences the activation of PI3K kinase and this in turn regulates mTOR, which was found to be upregulated in UFs both in the human and animal model [30, 31]. It also has an effect on forkhead box O transcription factors (FoxO) or Bcl-2 family proteins and different kinases, transcription factors and many other molecules to facilitate cell survival and cell cycle entry [17, 29].

The Ras/Raf/MEK/ERK signaling pathway is the second one that may be influenced by growth factors due to RTK interactions [32]. This pathway plays a major role in different physiological and pathological processes. Its activation triggers a cascade of which leads to the phosphorylation of target proteins in the nucleus and cytoplasm [17]. Briefly, activated Ras activates Raf kinases, then it phosphorylates MEK protein that activates ERK and several other transcription factors [17, 33]. It is critical that the bidirectional interaction between steroid signaling and the Ras/Raf/MEK/ERK pathway occurs, but growth factors may modulate the response to steroids through the effect of ERK on the transcriptional activity of steroid receptors [17]. As this pathway is both hormonal and non-hormonal, the growth factors produced without the influence of hormones might have their effect on its modulation. Since RTK-derived pathways are so important in the context of tumor biology current data suggest that they might be a target of several drugs [29]. Therefore, the extensive research in the area of RTKs, the MAP kinase and PI3K/AKT pathways is an unmet need in new medical therapies in UFs.

Cytokines, inflammation and their role in signaling in UFs

Inflammation is the response of tissues to harmful stimuli and various cytokines play an important role in its regulation [34]. Inflammation and its derived microenvironment have a pivotal influence on the risk of various courses of disease. In case of UFs, inflammation switches mostly to immunosuppression due to tumor evasion from anti-tumor immune response. However, in many cases this pathway is still unknown and hard to describe in detail [35]. Early life and prenatal exposure to different stimuli increase the risk of diseases in later life, due to the inflammation-derived reprogramming of the epigenome. It is frequently one the main causes of UF occurrence [35]. Paracrine signaling plays an important role in the cellular transformation of the myometrium. Cytokines may be responsible for UF-associated symptoms, such as pain or infertility, but nowadays more authors are focusing on their role in UF growth [16, 36].

Inflammation influences UF proliferation and growth via various mechanisms that include the alteration of the ECM. Andaloussi et al. (2017) emphasized that some cross talk occurred between hormones and inflammation in the pathogenesis of UFs. It is mostly related to their possible connection with UF-associated genetic defects including the most important MED12 gene mutation [37]. Moreover, chronic inflammation seems to be even more harmful to the uterus. As found by Orciani et al. (2018) the progenitor cells of UFs secreted significantly higher levels of cytokines with chronic inflammation rather than acute inflammation [15]. It may support the data that obesity [7] or hypertension [1] could also strongly influence the risk factors for UF occurrence. Immune response and DNA damage regulation with different compounds is also an important chapter in UF biology. The lack of different compounds, e.g. vitamin D in diet, is one of the causes of inflammation, genomic instability and cell proliferation induction [38]. Conversely, if some substances like phytoestrogens [39, 40] are in excess, the compounds become involved in DNA damage, immune response, and tumor occurrence and growth [39].

Steroids play a key role in these processes. Estrogen also plays a powerful role in immune response [38]. According to Murphy et al. (2010) those hormones were able to regulate nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activity and the production of pro-inflammatory cytokines through various mechanisms [41]. Progesterone also exerts an important anti-inflammatory effect, as it inhibits the activation of NF-κB and slows down the upregulation of cyclooxygenase 2 (COX-2) [42]. With all those mechanisms UFs have a special ability to avoid immune response. It may be connected with the fact that the uterus has a specific immunological role which is necessary in the maintenance of pregnancy to term [43].

NF-κB, which is a transcription factor involved in cell survival, proliferation and inflammatory response, is one of the pathways in UF-related inflammation. The second highly important pathway is connected with tumor necrosis factor α (TNF-α). It causes the activation of MAPKs and Tank-binding kinase 1 [37]. Numerous cytokines are involved in UF biology, but TNF-α appears to be one of the most important myometrium-associated cytokines, especially in relation to the clinical symptoms [16, 44]. Kurachi et al. (2001) found that TNF-α obtained in the proliferative phase was higher than in the secretory phase in UFs and that progesterone influenced the decrease of TNF-α expression [45]. TNF-α concentration was also found to be elevated in women with clinically symptomatic UFs, which suggests the presence of an inflammatory state in women with UFs [36]. Current data support the hypothesis that activin A exerts a potent fibrotic effect in UFs, and TNF-α was found to increase activin A messenger RNA (mRNA) expression in UFs [46]. It completes a complex cycle where TNF-α plays a role as activin A upregulator, activin A affects progesterone, which, in turn, inhibits TNF-α [44, 47]. Finally, TNF-α has a potent influence on ERKs, which play a role in integrating external signals from different mitogens and are a part of the Ras/Raf/MEK/ERK signal transduction cascade [44, 48]. There is a great deal of research into the impact of inflammation on UFs and their development and growth with an eye to finding clinical interventions.

Smad protein signaling

Smads are a family of structurally similar proteins that are the signal transducers for the TGF-β superfamily. They transmit signals from several cell membrane receptors to the nucleus [49]. Smads are believed to be extremely important for regulating cell biology. They play a role in proteasome-mediated degradation as they interact with ubiquitin ligases [50]. Smad ubiquitination-related factors have been implicated in their degradation. As suggested by available data, the degradation is implicated in the turnover of Smad mutants that may be connected with different lesions and may contribute to the growth and progression [51, 52]. The Smad pathways are complicated. Upon ligand binding to Smad-coupled receptors they heterotrimerize in several steps and the resulting complex moves to the nucleus to act as a transcription factor [53]. The TGF-β pathway includes Smads that play a role in the regulation of cellular proliferation and ECM formation at the transcriptional level [17, 54]. TGF-β modulates the expression of anti-fibrotic microRNA (miRNA) to maintain the pro-fibrotic functions of its signaling pathway. Smads have major connections with miRNAs that are of great importance in UFs. According to available data miR-21 promotes excessive ECM formation by stopping the Smad7 protein [55]. It was also found by Liang et al. (2014) also found that overexpression of that the overexpression of miR-26a inhibited the nuclear translocation of active Smads which might cause collagen deposition [56]. Smad3 and Smad4 were overexpressed in UFs in comparison with healthy myometrium. This overexpression changed when gonadotropin-releasing hormone (GnRH) agonist treatment was implemented [57]. The findings clearly indicate that this pathway is of high importance and may be the target of various anti-UF agents, including hormones.

Wingless type/β-catenin signaling

The Wnt signaling is a group of signal transduction pathways which are fundamental mechanisms that direct cell proliferation, polarity and fate determination. A critical and the most studied pathway in this signaling is the canonical Wnt, which plays a major role in the control of key developmental gene expression programs [58]. The Wnt pathways include various glycolipoproteins that pass signals into cells through their surface receptors [58]. The surface receptors, called Frizzled receptors, after connecting with their ligands, cause receptor activation and the phosphorylation of the cytoplasmic proteins, which leads to decreased β-catenin degradation and its accumulation in the cytoplasm. Its several steps trigger the activation of various transcription factors [17]. Accumulated cytoplasmic β-catenin reacts with chromatin and the family of T-cell transcription factor proteins. It changes the expression of a large number of genes and alters key cellular functions which might lead to tumorigenesis [8].

More evidence is available on Wnt/β-catenin and UF occurrence and growth. Research started around 2005, when it was found that the Wnt-derived gene is overexpressed in UFs [59]. Later, Tanwar et al. (2009) discovered that the constitutive expression of activated β-catenin in the animal model led to mesenchymal UF-like tumor development [60]. Mäkinen et al. (2011) found one of the major genetic bases of UFs. The authors examined several UFs via exome sequencing and identified tumor-specific mutations in the mediator complex subunit 12 (MED12) gene in about 70% of patients [12]. Stem cells derived from UF tissue, but not from healthy myometrium, were the MED12 gene mutations carriers. It suggests that a genetic hit transforms a myometrial stem cell into an abnormal one which will constitute the beginning of a UF tumor [8]. Mutations in the MED12 gene may lead to alterations in the interactions between this gene and β-catenin, leading to the inhibition of β-catenin transactivation in response to Wnt signaling [61].

According to other authors MED12 mutations overexpress Wnt4, a Wnt protein family member. Along with estrogen activation, it may lead to the occurrence of UF-like lesions in murine models [62]. A recent study by Ali et al. (2020) showed that estradiol induced β-catenin nuclear translocation and, consequently, its responsive genes in both the myometrium and UFs [63]. In 2013 Ono et al. demonstrated that the paracrine activation of the Wnt/β-catenin pathway in UF stem cells promoted tumor growth [64]. It was then suggested that the canonical Wnt pathway may be a potential therapeutic target for the treatment of UFs [65] or it may function as a biomarker [13]. Further research revealed that MED12 silencing reduced the proliferation of UF cells and it was mediated by this canonical pathway [66]. El Andaloussi et al. (2020) demonstrated that MED12 mutation presented a potential to transform cells by dysregulating Wnt4/β-catenin which affected mTOR signaling and might cause autophagy abrogation, cell proliferation, and tumorigenesis [67]. Furthermore, very recent interesting data by Harada et al. (2018) suggested that the inhibition of the canonical pathway signaling under hypoxia and starvation may initiate adipocyte differentiation and metaplasia in UFs, which might trigger its change into lipoleiomyoma [68].

Additionally, the Wnt/β-catenin pathway leads to the increased levels of TGF-β3 which is considered to be one of the key factors in the pathophysiology of UFs [18]. Some of those findings were supported with studies that used anti-UF agents which caused the attenuation of this pathway by reducing TGF-β3 signal and protein expression, resulting in a reduction in TGF-β canonical signaling [69]. Corachán et al. (2019) has recently found that the pleiotropic effect of vitamin D might also influence this pathway, as it causes cell growth arrest and Wnt/β-catenin pathway inhibition, but not via apoptosis regulation [70]. Further research in this field is of great importance as current data indicate that this pathway might be one of the best targets for UF therapy.

Peroxisome proliferator-activated receptor signaling

PPARs are a group of nuclear receptor proteins that function as transcription factors [71]. PPARs play various roles, most of them connected with the regulation of cell development, differentiation, metabolism and potential tumor formation [71, 72]. There are three types of those receptors: alpha, delta/beta and gamma [71]. They act as different heterodimers in association with an activator complex that binds to DNA sequence, which leads to the transactivation and transrepression of various genes [71].

There is growing evidence for the role of PPARγ signaling in UF development and growth, especially concerning the gamma family. PPARγ levels were higher in UFs in comparison with healthy myometrium [73]. PPARγ signaling may lead to the inhibition of UF growth through modulating steroid signaling as the stimulation of PPARγ causes the inhibition of estrogen-derived gene expression [74]. Finally, it was found that some agents might influence UF cells through those receptors [75]. Therefore, it was concluded that this pathway might be a potential target to be modulated by new therapeutic compounds [17]. Curcumin, a natural agent, was found to act as a PPARγ ligand and inhibited UF cell proliferation [76]. Further research also showed that it extensively regulated other key pathways, such as MAPK and TGF-β/Smad, which may suggest that the pathways do not work on their own, but are a complicated network of molecular action [77]. However, data about PPARs and UFs are still scarce and there is not much research in the field. We hope that the forthcoming years will bring more high quality studies in this area.

Retinoic acid signaling

All-trans-retinoic acid, commonly known as retinoic acid (RA), is an active metabolite of vitamin A that mediates the functions of this vitamin required for growth and development [78]. RA may be produced in humans in two sequential oxidation steps [79]. RA receptors act as heterodimers with retinoid X receptors (RXRs) [80]. Moreover, RXRs also act as heterodimers for several different nuclear receptors, including the vitamin D receptor (VDR), PPARs and the thyroid hormone receptors [81]. These connections should be treated as a kind of evidence that aberrations in the RA pathways may play a role in UF development and growth, especially considering their relations with PPARs or the VDR.

Available data showed that UF cells responded to RA [82]. Various researchers found that the gene expression of some proteins, enzymes and receptors varied in the RA molecular pathway in UFs in comparison with healthy myometrium [8385]. Further research revealed that different levels of RXRs might be the cause of abnormal transcriptional activity in UFs [17].

Modulating RA signaling may be a potential target in therapy. Pharmacological studies support the findings about the influence of RA signaling in UFs. RXR ligands were found to reduce the size of UFs in the animal model [86]. Gilden et al. (2012) found that an RA metabolic blocking agent, liarozole, inhibited ECM protein production in immortalized UF cells, whereas normal myometrial cells demonstrated no significant alteration in ECM regulation [87].

Pharmacological treatment of uterine fibroids – hormonal vs non-hormonal agents

Increasing knowledge of the role of hormonal and non-hormonal signaling links in the pathophysiology of UFs may present an opportunity to understand this difficult topic and constitute a step towards the use of specific signal transduction inhibitors for the treatment and prevention of UFs.

Treatment for UFs depends on the patient’s symptoms and her personal goals [88, 89]. Definitive treatment such as surgery may be important in patients wishing to conceive quickly whereas medical interventions may be indicated in patients whose primary concerns are bleeding and/or pain [90, 91]. And undeniable problem associated with UFs is that pharmacological treatment options are limited [4, 92].

Poor understanding and the lack of data about the precise molecular mechanism in UFs is the reason for the limitation of medical treatment. However, the benefits of medical treatment, such as lower costs or better acceptance in patients, are tempered with average efficacy [93]. Recent publications have shed light on new interesting hormonal compounds, such as selective progesterone receptor modulators (SPRMs): ulipristal acetate (UPA) [92], vilaprisan (VPR) [94] and new oral gonadotropin-releasing hormone (GnRH) analogs, like elagolix [95, 96], relugolix [97] or linzagolix [98]. However, UPA might cause negative effect on the liver [99] and VPR studies were discontinued due to the negative findings in its long-term use in animals [94]. Therefore, the field of pharmacological treatment in UFs is still open.

Non-hormonal interventions, both natural and synthetic, would be appealing to patients if effective [100, 101]. Current data concerning various compounds that use non-hormonal pathways in UFs are presented in Table 1.

Table 1.

Examples of compounds using non-hormonal pathways in medical treatment of uterine fibroids.

Natural compound Author Example of non-hormonal molecular effect in uterine fibroids
Berberine Wu et al. 2015 [102]
Chuang et al. 2017 [103]
Inflammation inhibition – reduced COX-2 expression.
Curcumin Malik et al. 2009 [104]
Tsuiji et al. 2011 [76]
Yu et al. 2019 [77]
ERK 1, ERK 2, and nuclear factor kappa B (NF-κB) pathway inhibition.
Cell proliferation inhibition, PPARγ ligand.
MAPK, PPAR and TGF-β/Smad regulation.
Epigallocatechin gallate (EGCG) Zhang et al. 2010 [105] The upregulation of the TGF-β and stress pathways, the inhibition of the NF-κB-dependent inflammatory pathway.
Decrease in the expression of proliferating cell nuclear antigen (PCNA), cyclin-dependent kinase 4 and B-cell lymphoma 2 (Bcl-2).
Apoptosis increase with the expression of the Bcl-2-associated X (Bax).
Fucoidan Chen et al. 2018 [106] The downregulation of fibronectin, vimentin, α-smooth muscle actin (α-SMA) and collagen 1A1 protein levels in TGFβ3-induced UF cells. The abrogation of TGFβ3-induced levels of Smad2 and ERK1/2, as well as β-catenin translocation into the nucleus.
Halofuginone Grudzien et al. 2010 [107]
Koohestani et al. 2016 [108]
The reduction of collagen type I and collagen type III mRNA levels, as well as of the profibrotic factor TGFbeta1 mRNA levels.
Tumor volume reduction through an increase in apoptosis and a reduction in cell proliferation, but no significant changes at the transcript level of the expression of TGFβ1, TGFβ3 and transforming growth factor β receptors I and II (TGFβ-R1, TGFβ-R2) genes.
Indole-3-carbinol Greco et al. 2020 [109] mRNA levels of collagen 1A1 decrease in UF cells.
Pathway connected with signaling in the ECM.
Isoliquiritigenin Lin et al. 2019 [110] Co-treatment with estradiol and isoliquiritigenin inhibited ERK1/2 activation.
p38 and c-Jun N-terminal kinase (JNK) activation enhancement.
The downregulation of the expression of ECM-associated proteins and matrix metalloproteinases (MMPs).
Locostatin Janjusevic et al. 2016 [111] The activation of the MAPK signaling pathway and ERK phosphorylation.
Reduced glycogen synthase kinase 3 β (GSK3β) expression, which even with the above-mentioned activation led to the reduction of the proliferation and migration of UF cells.
Pifenidone Lee et al. 1998 [112] The inhibition of the proliferation of UF cells and suppression of the mRNA levels of collagen type I and collagen type III.
Resveratrol Wu et al. 2016 [113]
Chen et al. 2019 [114]
Cell cycle arrest and apoptosis regulation. ECM-related protein expression reduction. MMP-2 and MMP-9 expression inhibition.
PCNA, fibronectin protein expression decrease, Bax and Bcl-2 in vivo.
Fibronectin, collagen type 1 and α-SMA protein expression.
β-catenin protein level reduction.
Retinoic acid Friedman et al. 2016 [115] RA significantly affects growth, signaling pattern and interactions among PI3K/Bcl2/retinol proteins.
β-catenin level depends on the interaction between response to treatment and cell type.
Romina strawberry (anthocyanin fraction) Giamperi et al. 2019 [116] Mechanism still unclear.
Collagen 1A1, fibronectin, versican, and activin A messenger RNA expression decrease. Pathway connected with signaling in the ECM.
Simvastatin Borahay et al. 2014 [117]
Borahay et al. 2015 [118]
Malik et al. 2018 [104]
The decrease of ERK activation and decrease of Ras isoprenylation.
The inhibition of the Akt signaling pathway in UF cells.
The inhibition of the expression of major ECM proteins – collagen I, collagen III, fibronectin, versican, and brevican.
Quercetin Greco et al. 2020 [109] mRNA levels of collagen 1A1 and fibronectin decrease in UF cells.
Pathway connected with signaling in the ECM.
Verteporfin Islam et al. 2019 [119] The reduction of mRNA levels of versican and MMP-14.
Inflammation inhibition, decrease in interleukin 8 (IL-8) levels.
TGF-β1 and TGF-β3 mRNA expression levels decreased.

The majority of substances presented in Table 1 are of natural origin, which suggests that various substances make up our environment and it is advisable to take a closer look to find new solutions that might be useful in UF treatment [120, 121]. In case of UFs substances using non-hormonal pathways are a new issue which seems to be an attractive option for future research. Therefore, our review appears to contain even more necessary information for researchers interested in the topic. Obviously, it seems rather unlikely that preparations using non-hormonal pathways might completely replace hormonal treatment. It is mostly due to the fact that hormonal determinants are unquestionably of key importance in UFs, while non-hormonal ones are rather additional pathways which interlock with them. Another issue is related to the early stages of knowledge concerning those associations and the paucity of studies. Currently, the proposal of combined therapies, e.g. hormonal and non-hormonal, is an attractive option for further research. Such a treatment might aim at obtaining synergism with one compound assisting the other, making the clinical effect considerably more beneficial [122]. The use of several substances for a specific indication is not as common in UF treatment as in, for example, cardiology [123, 124], but in our viewpoint it seems to be highly rational. Preliminary studies are available is this area, e.g. in case of vitamin D and UPA [125] or vitamin D and EGCG [126], However, the data are scarce for the time being. Seemingly, it is important to suggest that other researchers, both clinicians and laboratory ones, should seek new effective combinations of substances to obtain a more beneficial clinical effect and the optimal symptom reduction in patients. We hope that research will provide simple, non-invasive, and effective treatments.

Conclusion

UF growth involves a complex network of various signaling pathways. Increasing data about the role of hormonal signaling in UFs stand in disproportion to the level of knowledge concerning non-hormonal signaling. However, as UFs are still a great mystery, research concerning non-hormonal pathways in those tumors may present an opportunity to understand this difficult issue and be a step towards the use of specific signal transduction inhibitors for treatment and prevention.

Numerous non-hormonal agents, both natural and synthetic ones, may find their place in the therapy of UFs. Since all the pathways are linked in some manner, we face important therapeutic implications. Finding non-hormonal therapeutic targets will help to serve a group of patients who would prefer an alternative to surgical intervention

Key points.

  • Growth factors are important hormonal and non-hormonal mediators in uterine fibroid biology.

  • Early life exposure and inflammation are key factors in uterine fibroid development and growth.

  • The Wnt/β-catenin pathway is of considerable importance in uterine fibroids being a subject of extensive research these years.

  • Modulating retinoic acid signaling may be a potential target in uterine fibroid therapy.

  • Oral gonadotropin-releasing hormone analogs and non-hormonal compounds are the future in uterine fibroid therapy.

Acknowledgements

Financial support and sponsorship

This study was supported in part by the US National Institutes of Health grants: R01 HD094378–04, R01 ES 028615–02, R01 HD100367–01, U54 MD007602 and R01 HD094380–02.

Michał Ciebiera scholarship at University of Illinois at Chicago was founded by The Kosciuszko Foundation, The American Center of Polish Culture.

Footnotes

Conflicts of interest

Ayman Al-Hendy is a consultant for Abbvie, Allergan, Bayer, Myovant Sciences and Repros. No conflicts were declared for the remaining authors.

References

  • 1.Stewart EA, Cookson CL, Gandolfo RA, Schulze-Rath R. Epidemiology of uterine fibroids: a systematic review. BJOG. 2017;124(10):1501–12.** One of the most important systematic reviews on UF incidence, prevalence and associated risk factors.
  • 2.Al-Hendy A, Myers ER, Stewart E. Uterine Fibroids: Burden and Unmet Medical Need. Semin Reprod Med. 2017;35(6):473–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Gracia M, Carmona F. Uterine myomas: Clinical impact and pathophysiological bases. Eur J Obstet Gynecol Reprod Biol. 2020. Epub. Jan 30;S0301–2115(20)30052-X. [DOI] [PubMed] [Google Scholar]
  • 4.Ulin M, Ali M, Chaudhry ZT, Al-Hendy A, Yang Q. Uterine fibroids in menopause and perimenopause. Menopause. 2020;27(2):238–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Murji A, Bedaiwy M, Singh SS, Bougie O, CAPTURE Registry Steering Committee. Influence of Ethnicity on Clinical Presentation and Quality of Life in Women With Uterine Fibroids: Results From a Prospective Observational Registry. J Obstet Gynaecol Can. 2019. Epub. Dec 25 [DOI] [PubMed] [Google Scholar]
  • 6.Stewart EA, Laughlin-Tommaso SK, Catherino WH, Lalitkumar S, Gupta D, Vollenhoven B. Uterine fibroids. Nat Rev Dis Primers. 2016;2:16043. [DOI] [PubMed] [Google Scholar]
  • 7.Ciebiera M, Wlodarczyk M, Slabuszewska-Jozwiak A, Nowicka G, Jakiel G. Influence of vitamin D and transforming growth factor beta3 serum concentrations, obesity, and family history on the risk for uterine fibroids. Fertil Steril. 2016;106(7):1787–92. [DOI] [PubMed] [Google Scholar]
  • 8.Bulun SE. Uterine fibroids. N Engl J Med. 2013;369(14):1344–55. [DOI] [PubMed] [Google Scholar]
  • 9.Islam MS, Ciavattini A, Petraglia F, Castellucci M, Ciarmela P. Extracellular matrix in uterine leiomyoma pathogenesis: a potential target for future therapeutics. Hum Reprod Update. 2018;24(1):59–85.** Comprehensive review about the biology the role of ECM in UFs. Authors discuss also potential targets for therapy.
  • 10.Baird DD, Patchel SA, Saldana TM, Umbach DM, Cooper T, Wegienka G, et al. Uterine fibroid incidence and growth in an ultrasound-based, prospective study of young African-Americans. Am J Obstet Gynecol. 2020. Epub. Feb 24;S0002–9378(20)30196–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.McWilliams MM, Chennathukuzhi VM. Recent Advances in Uterine Fibroid Etiology. Semin Reprod Med. 2017;35(2):181–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Makinen N, Mehine M, Tolvanen J, Kaasinen E, Li Y, Lehtonen HJ, et al. MED12, the mediator complex subunit 12 gene, is mutated at high frequency in uterine leiomyomas. Science. 2011;334(6053):252–5. [DOI] [PubMed] [Google Scholar]
  • 13.Mehine M, Kaasinen E, Heinonen HR, Makinen N, Kampjarvi K, Sarvilinna N, et al. Integrated data analysis reveals uterine leiomyoma subtypes with distinct driver pathways and biomarkers. Proc Natl Acad Sci U S A. 2016;113(5):1315–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ishikawa H, Ishi K, Serna VA, Kakazu R, Bulun SE, Kurita T. Progesterone is essential for maintenance and growth of uterine leiomyoma. Endocrinology. 2010;151(6):2433–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Orciani M, Caffarini M, Biagini A, Lucarini G, Delli Carpini G, Berretta A, et al. Chronic Inflammation May Enhance Leiomyoma Development by the Involvement of Progenitor Cells. Stem Cells Int. 2018;2018:1716246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Protic O, Toti P, Islam MS, Occhini R, Giannubilo SR, Catherino WH, et al. Possible involvement of inflammatory/reparative processes in the development of uterine fibroids. Cell Tissue Res. 2016;364(2):415–27. [DOI] [PubMed] [Google Scholar]
  • 17.Borahay MA, Al-Hendy A, Kilic GS, Boehning D. Signaling Pathways in Leiomyoma: Understanding Pathobiology and Implications for Therapy. Mol Med. 2015;21:242–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ciebiera M, Wlodarczyk M, Wrzosek M, Meczekalski B, Nowicka G, Lukaszuk K, et al. Role of Transforming Growth Factor beta in Uterine Fibroid Biology. Int J Mol Sci. 2017;18(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Borahay MA, Asoglu MR, Mas A, Adam S, Kilic GS, Al-Hendy A. Estrogen Receptors and Signaling in Fibroids: Role in Pathobiology and Therapeutic Implications. Reprod Sci. 2017;24(9):1235–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ciarmela P, Islam MS, Reis FM, Gray PC, Bloise E, Petraglia F, et al. Growth factors and myometrium: biological effects in uterine fibroid and possible clinical implications. Hum Reprod Update. 2011;17(6):772–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Burgess A. Growth factors and cytokines. Rev Cell Biol Mol Medicine. 2015;1(2). [Google Scholar]
  • 22.Vollenhoven BJ, Herington AC, Healy DL. Epidermal growth factor and transforming growth factor-beta in uterine fibroids and myometrium. Gynecol Obstet Invest. 1995;40(2):120–4. [DOI] [PubMed] [Google Scholar]
  • 23.Helmke BM, Markowski DN, Muller MH, Sommer A, Muller J, Moller C, et al. HMGA proteins regulate the expression of FGF2 in uterine fibroids. Mol Hum Reprod. 2011;17(2):135–42. [DOI] [PubMed] [Google Scholar]
  • 24.Peng L, Wen Y, Han Y, Wei A, Shi G, Mizuguchi M, et al. Expression of insulin-like growth factors (IGFs) and IGF signaling: molecular complexity in uterine leiomyomas. Fertil Steril. 2009;91(6):2664–75. [DOI] [PubMed] [Google Scholar]
  • 25.Liang M, Wang H, Zhang Y, Lu S, Wang Z. Expression and functional analysis of platelet-derived growth factor in uterine leiomyomata. Cancer Biol Ther. 2006;5(1):28–33. [DOI] [PubMed] [Google Scholar]
  • 26.Luddi A, Marrocco C, Governini L, Semplici B, Pavone V, Capaldo A, et al. Increased expression of neurogenic factors in uterine fibroids. Hum Reprod. 2019;34(11):2153–62. [DOI] [PubMed] [Google Scholar]
  • 27.Tal R, Segars JH. The role of angiogenic factors in fibroid pathogenesis: potential implications for future therapy. Hum Reprod Update. 2014;20(2):194–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Yu L, Moore AB, Dixon D. Receptor tyrosine kinases and their hormonal regulation in uterine leiomyoma. Semin Reprod Med. 2010;28(3):250–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Regad T. Targeting RTK Signaling Pathways in Cancer. Cancers (Basel). 2015;7(3):1758–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Crabtree JS, Jelinsky SA, Harris HA, Choe SE, Cotreau MM, Kimberland ML, et al. Comparison of human and rat uterine leiomyomata: identification of a dysregulated mammalian target of rapamycin pathway. Cancer Res. 2009;69(15):6171–8. [DOI] [PubMed] [Google Scholar]
  • 31.Al-Hendy A, Diamond MP, Boyer TG, Halder SK. Vitamin D3 Inhibits Wnt/beta-Catenin and mTOR Signaling Pathways in Human Uterine Fibroid Cells. J Clin Endocrinol Metab. 2016;101(4):1542–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell. 2010;141(7):1117–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Li L, Zhao GD, Shi Z, Qi LL, Zhou LY, Fu ZX. The Ras/Raf/MEK/ERK signaling pathway and its role in the occurrence and development of HCC. Oncol Lett. 2016;12(5):3045–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, et al. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget. 2018;9(6):7204–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Yang Q, Ali M, El Andaloussi A, Al-Hendy A. The emerging spectrum of early life exposure-related inflammation and epigenetic therapy. Cancer Stud Mol Med. 2018;4(1):13–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ciebiera M, Wlodarczyk M, Wrzosek M, Wojtyla C, Meczekalski B, Nowicka G, et al. TNF-alpha serum levels are elevated in women with clinically symptomatic uterine fibroids. Int J Immunopathol Pharmacol. 2018;32:2058738418779461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.El Andaloussi A, Chaudhry Z, Al-Hendy A, Ismail N. Uterine Fibroids: Bridging Genomic Defects and Chronic Inflammation. Semin Reprod Med. 2017;35(6):494–8. [DOI] [PubMed] [Google Scholar]
  • 38.Elhusseini H, Elkafas H, Abdelaziz M, Halder S, Atabiekov I, Eziba N, et al. Diet-induced vitamin D deficiency triggers inflammation and DNA damage profile in murine myometrium. Int J Womens Health. 2018;10:503–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yang Q, Diamond MP, Al-Hendy A. Early Life Adverse Environmental Exposures Increase the Risk of Uterine Fibroid Development: Role of Epigenetic Regulation. Front Pharmacol. 2016;7:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ciebiera M, Szymanska-Majchrzak J, Sentkowska A, Kilian K, Rogulski Z, Nowicka G, et al. Alpha-Tocopherol Serum Levels Are Increased in Caucasian Women with Uterine Fibroids: A Pilot Study. Biomed Res Int. 2018;2018:6793726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Murphy AJ, Guyre PM, Pioli PA. Estradiol suppresses NF-kappa B activation through coordinated regulation of let-7a and miR-125b in primary human macrophages. J Immunol. 2010;184(9):5029–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hardy DB, Janowski BA, Corey DR, Mendelson CR. Progesterone receptor plays a major antiinflammatory role in human myometrial cells by antagonism of nuclear factor-kappaB activation of cyclooxygenase 2 expression. Mol Endocrinol. 2006;20(11):2724–33. [DOI] [PubMed] [Google Scholar]
  • 43.Lee SK, Kim CJ, Kim DJ, Kang JH. Immune cells in the female reproductive tract. Immune Netw. 2015;15(1):16–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ciebiera M, Wlodarczyk M, Zgliczynska M, Lukaszuk K, Meczekalski B, Kobierzycki C, et al. The Role of Tumor Necrosis Factor alpha in the Biology of Uterine Fibroids and the Related Symptoms. Int J Mol Sci. 2018;19(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kurachi O, Matsuo H, Samoto T, Maruo T. Tumor necrosis factor-alpha expression in human uterine leiomyoma and its down-regulation by progesterone. J Clin Endocrinol Metab. 2001;86(5):2275–80. [DOI] [PubMed] [Google Scholar]
  • 46.Islam MS, Catherino WH, Protic O, Janjusevic M, Gray PC, Giannubilo SR, et al. Role of activin-A and myostatin and their signaling pathway in human myometrial and leiomyoma cell function. J Clin Endocrinol Metab. 2014;99(5):E775–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Protic O, Islam MS, Greco S, Giannubilo SR, Lamanna P, Petraglia F, et al. Activin A in Inflammation, Tissue Repair, and Fibrosis: Possible Role as Inflammatory and Fibrotic Mediator of Uterine Fibroid Development and Growth. Semin Reprod Med. 2017;35(6):499–509. [DOI] [PubMed] [Google Scholar]
  • 48.Wang Y, Feng G, Wang J, Zhou Y, Liu Y, Shi Y, et al. Differential effects of tumor necrosis factor-alpha on matrix metalloproteinase-2 expression in human myometrial and uterine leiomyoma smooth muscle cells. Hum Reprod. 2015;30(1):61–70. [DOI] [PubMed] [Google Scholar]
  • 49.Macias MJ, Martin-Malpartida P, Massague J. Structural determinants of Smad function in TGF-beta signaling. Trends Biochem Sci. 2015;40(6):296–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Alarcon C, Zaromytidou AI, Xi Q, Gao S, Yu J, Fujisawa S, et al. Nuclear CDKs drive Smad transcriptional activation and turnover in BMP and TGF-beta pathways. Cell. 2009;139(4):757–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Izzi L, Attisano L. Regulation of the TGFbeta signalling pathway by ubiquitin-mediated degradation. Oncogene. 2004;23(11):2071–8. [DOI] [PubMed] [Google Scholar]
  • 52.Imamura T, Oshima Y, Hikita A. Regulation of TGF-beta family signalling by ubiquitination and deubiquitination. J Biochem. 2013;154(6):481–9. [DOI] [PubMed] [Google Scholar]
  • 53.Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature. 2003;425(6958):577–84. [DOI] [PubMed] [Google Scholar]
  • 54.Miyazawa K, Miyazono K. Regulation of TGF-beta Family Signaling by Inhibitory Smads. Cold Spring Harb Perspect Biol. 2017;9(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Noetel A, Kwiecinski M, Elfimova N, Huang J, Odenthal M. microRNA are Central Players in Anti- and Profibrotic Gene Regulation during Liver Fibrosis. Front Physiol. 2012;3:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Liang H, Xu C, Pan Z, Zhang Y, Xu Z, Chen Y, et al. The antifibrotic effects and mechanisms of microRNA-26a action in idiopathic pulmonary fibrosis. Mol Ther. 2014;22(6):1122–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Chegini N, Luo X, Ding L, Ripley D. The expression of Smads and transforming growth factor beta receptors in leiomyoma and myometrium and the effect of gonadotropin releasing hormone analogue therapy. Mol Cell Endocrinol. 2003;209(1–2):9–16. [DOI] [PubMed] [Google Scholar]
  • 58.MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell. 2009;17(1):9–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Mangioni S, Vigano P, Lattuada D, Abbiati A, Vignali M, Di Blasio AM. Overexpression of the Wnt5b gene in leiomyoma cells: implications for a role of the Wnt signaling pathway in the uterine benign tumor. J Clin Endocrinol Metab. 2005;90(9):5349–55. [DOI] [PubMed] [Google Scholar]
  • 60.Tanwar PS, Lee HJ, Zhang L, Zukerberg LR, Taketo MM, Rueda BR, et al. Constitutive activation of Beta-catenin in uterine stroma and smooth muscle leads to the development of mesenchymal tumors in mice. Biol Reprod. 2009;81(3):545–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kim S, Xu X, Hecht A, Boyer TG. Mediator is a transducer of Wnt/beta-catenin signaling. J Biol Chem. 2006;281(20):14066–75. [DOI] [PubMed] [Google Scholar]
  • 62.Markowski DN, Bartnitzke S, Loning T, Drieschner N, Helmke BM, Bullerdiek J. MED12 mutations in uterine fibroids--their relationship to cytogenetic subgroups. Int J Cancer. 2012;131(7):1528–36. [DOI] [PubMed] [Google Scholar]
  • 63.Ali M, Shahin SM, Sabri NA, Al-Hendy A, Yang Q. Activation of beta-Catenin Signaling and its Crosstalk With Estrogen and Histone Deacetylases in Human Uterine Fibroids. J Clin Endocrinol Metab. 2020;105(4).* New data about the β-catenin nuclear translocation contribution to UF phenotype and its modulation by estradiol and histone deacetylases activity.
  • 64.Ono M, Yin P, Navarro A, Moravek MB, Coon JSt, Druschitz SA, et al. Paracrine activation of WNT/beta-catenin pathway in uterine leiomyoma stem cells promotes tumor growth. Proc Natl Acad Sci U S A. 2013;110(42):17053–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ono M, Yin P, Navarro A, Moravek MB, Coon VJ, Druschitz SA, et al. Inhibition of canonical WNT signaling attenuates human leiomyoma cell growth. Fertil Steril. 2014;101(5):1441–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Al-Hendy A, Laknaur A, Diamond MP, Ismail N, Boyer TG, Halder SK. Silencing Med12 Gene Reduces Proliferation of Human Leiomyoma Cells Mediated via Wnt/beta-Catenin Signaling Pathway. Endocrinology. 2017;158(3):592–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.El Andaloussi A, Al-Hendy A, Ismail N, Boyer TG, Halder SK. Introduction of Somatic Mutation in MED12 Induces Wnt4/beta-Catenin and Disrupts Autophagy in Human Uterine Myometrial Cell. Reprod Sci. 2020;27(3):823–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Harada H, Tsuda Y, Yabuki K, Shiba E, Uchihashi K, Matsuyama A, et al. Inhibition of WNT/beta-catenin signaling under serum starvation and hypoxia induces adipocytic transdifferentiation in human leiomyoma cells. Lab Invest. 2018;98(4):439–48. [DOI] [PubMed] [Google Scholar]
  • 69.Lewis TD, Malik M, Britten J, Parikh T, Cox J, Catherino WH. Ulipristal acetate decreases active TGF-beta3 and its canonical signaling in uterine leiomyoma via two novel mechanisms. Fertil Steril. 2019;111(4):806–15 e1. [DOI] [PubMed] [Google Scholar]
  • 70.Corachan A, Ferrero H, Aguilar A, Garcia N, Monleon J, Faus A, et al. Inhibition of tumor cell proliferation in human uterine leiomyomas by vitamin D via Wnt/beta-catenin pathway. Fertil Steril. 2019;111(2):397–407.*Very recent data about connections between vitamin D and Wnt/beta-catenin pathway in UFs.
  • 71.Tyagi S, Gupta P, Saini AS, Kaushal C, Sharma S. The peroxisome proliferator-activated receptor: A family of nuclear receptors role in various diseases. J Adv Pharm Technol Res. 2011;2(4):236–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Berger J, Moller DE. The mechanisms of action of PPARs. Annu Rev Med. 2002;53:409–35. [DOI] [PubMed] [Google Scholar]
  • 73.Jeong YJ, Noh EM, Lee YR, Yu HN, Jang KY, Lee SJ, et al. 17beta-estradiol induces up-regulation of PTEN and PPARgamma in leiomyoma cells, but not in normal cells. Int J Oncol. 2010;36(4):921–7. [DOI] [PubMed] [Google Scholar]
  • 74.Houston KD, Copland JA, Broaddus RR, Gottardis MM, Fischer SM, Walker CL. Inhibition of proliferation and estrogen receptor signaling by peroxisome proliferator-activated receptor gamma ligands in uterine leiomyoma. Cancer Res. 2003;63(6):1221–7. [PubMed] [Google Scholar]
  • 75.Nam DH, Ramachandran S, Song DK, Kwon KY, Jeon DS, Shin SJ, et al. Growth inhibition and apoptosis induced in human leiomyoma cells by treatment with the PPAR gamma ligand ciglitizone. Mol Hum Reprod. 2007;13(11):829–36. [DOI] [PubMed] [Google Scholar]
  • 76.Tsuiji K, Takeda T, Li B, Wakabayashi A, Kondo A, Kimura T, et al. Inhibitory effect of curcumin on uterine leiomyoma cell proliferation. Gynecol Endocrinol. 2011;27(7):512–7. [DOI] [PubMed] [Google Scholar]
  • 77.Yu CH, Zhao JS, Zhao H, Peng T, Shen DC, Xu QX, et al. Transcriptional profiling of uterine leiomyoma rats treated by a traditional herb pair, Curcumae rhizoma and Sparganii rhizoma. Braz J Med Biol Res. 2019;52(6):e8132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Rhinn M, Dolle P. Retinoic acid signalling during development. Development. 2012;139(5):843–58. [DOI] [PubMed] [Google Scholar]
  • 79.Kedishvili NY. Retinoic Acid Synthesis and Degradation. Subcell Biochem. 2016;81:127–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Allenby G, Bocquel MT, Saunders M, Kazmer S, Speck J, Rosenberger M, et al. Retinoic acid receptors and retinoid X receptors: interactions with endogenous retinoic acids. Proc Natl Acad Sci U S A. 1993;90(1):30–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Niederreither K, Dolle P. Retinoic acid in development: towards an integrated view. Nat Rev Genet. 2008;9(7):541–53. [DOI] [PubMed] [Google Scholar]
  • 82.Boettger-Tong H, Shipley G, Hsu CJ, Stancel GM. Cultured human uterine smooth muscle cells are retinoid responsive. Proc Soc Exp Biol Med. 1997;215(1):59–65. [DOI] [PubMed] [Google Scholar]
  • 83.Zaitseva M, Vollenhoven BJ, Rogers PA. Retinoic acid pathway genes show significantly altered expression in uterine fibroids when compared with normal myometrium. Mol Hum Reprod. 2007;13(8):577–85. [DOI] [PubMed] [Google Scholar]
  • 84.Zaitseva M, Vollenhoven BJ, Rogers PA. Retinoids regulate genes involved in retinoic acid synthesis and transport in human myometrial and fibroid smooth muscle cells. Hum Reprod. 2008;23(5):1076–86. [DOI] [PubMed] [Google Scholar]
  • 85.Catherino WH, Malik M. Uterine leiomyomas express a molecular pattern that lowers retinoic acid exposure. Fertil Steril. 2007;87(6):1388–98. [DOI] [PubMed] [Google Scholar]
  • 86.Gamage SD, Bischoff ED, Burroughs KD, Lamph WW, Gottardis MM, Walker CL, et al. Efficacy of LGD1069 (Targretin), a retinoid X receptor-selective ligand, for treatment of uterine leiomyoma. J Pharmacol Exp Ther. 2000;295(2):677–81. [PubMed] [Google Scholar]
  • 87.Gilden M, Malik M, Britten J, Delgado T, Levy G, Catherino WH. Leiomyoma fibrosis inhibited by liarozole, a retinoic acid metabolic blocking agent. Fertil Steril. 2012;98(6):1557–62. [DOI] [PubMed] [Google Scholar]
  • 88.Bonafede MM, Pohlman SK, Miller JD, Thiel E, Troeger KA, Miller CE. Women with Newly Diagnosed Uterine Fibroids: Treatment Patterns and Cost Comparison for Select Treatment Options. Popul Health Manag. 2018;21(S1):S13–S20. [DOI] [PubMed] [Google Scholar]
  • 89.Giuliani E, As-Sanie S, Marsh EE. Epidemiology and management of uterine fibroids. Int J Gynaecol Obstet. 2020;149(1):3–9. [DOI] [PubMed] [Google Scholar]
  • 90.Donnez J, Dolmans MM. Uterine fibroid management: from the present to the future. Hum Reprod Update. 2016;22(6):665–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Dolmans MM, Donnez J, Fellah L. Uterine fibroid management: Today and tomorrow. J Obstet Gynaecol Res. 2019;45(7):1222–9. [DOI] [PubMed] [Google Scholar]
  • 92.Ali M, Chaudhry ZT, Al-Hendy A. Successes and failures of uterine leiomyoma drug discovery. Expert Opin Drug Discov. 2018;13(2):169–77. [DOI] [PubMed] [Google Scholar]
  • 93.Gurusamy KS, Vaughan J, Fraser IS, Best LM, Richards T. Medical Therapies for Uterine Fibroids - A Systematic Review and Network Meta-Analysis of Randomised Controlled Trials. PLoS One. 2016;11(2):e0149631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Ciebiera M, Vitale SG, Ferrero S, Vilos GA, Barra F, Caruso S, et al. Vilaprisan, a New Selective Progesterone Receptor Modulator in Uterine Fibroid Pharmacotherapy - Will It Really Be a Breakthrough? Curr Pharm Des. 2020;26(3):300–309. [DOI] [PubMed] [Google Scholar]
  • 95.Schlaff WD, Ackerman RT, Al-Hendy A, Archer DF, Barnhart KT, Bradley LD, et al. Elagolix for Heavy Menstrual Bleeding in Women with Uterine Fibroids. N Engl J Med. 2020;382(4):328–40.* Very recent data about the new effective compound for UF medical terapy - elagolix.
  • 96.Neri M, Melis GB, Giancane E, Vallerino V, Pilloni M, Piras B, et al. Clinical Utility Of Elagolix As An Oral Treatment For Women With Uterine Fibroids: A Short Report On The Emerging Efficacy Data. Int J Womens Health. 2019;11:535–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Barra F, Seca M, Della Corte L, Giampaolino P, Ferrero S. Relugolix for the treatment of uterine fibroids. Drugs Today (Barc). 2019;55(8):503–12. [DOI] [PubMed] [Google Scholar]
  • 98.Pohl O, Marchand L, Bell D, Gotteland JP. Effects of combined GnRH receptor antagonist linzagolix and hormonal add-back therapy on vaginal bleeding-delayed add-back onset does not improve bleeding pattern. Reprod Sci. 2020;27(4):988–95. [DOI] [PubMed] [Google Scholar]
  • 99.Mahase E. Uterine fibroid drug is recalled after case of liver failure requiring transplant prompts EU review. BMJ. 2020;368:m1112. [DOI] [PubMed] [Google Scholar]
  • 100.Ciebiera M, Lukaszuk K, Meczekalski B, Ciebiera M, Wojtyla C, Slabuszewska-Jozwiak A, et al. Alternative Oral Agents in Prophylaxis and Therapy of Uterine Fibroids-An Up-to-Date Review. Int J Mol Sci. 2017;18(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Islam MS, Segars JH, Castellucci M, Ciarmela P. Dietary phytochemicals for possible preventive and therapeutic option of uterine fibroids: Signaling pathways as target. Pharmacol Rep. 2017;69(1):57–70. [DOI] [PubMed] [Google Scholar]
  • 102.Wu HL, Chuang TY, Al-Hendy A, Diamond MP, Azziz R, Chen YH. Berberine inhibits the proliferation of human uterine leiomyoma cells. Fertil Steril. 2015;103(4):1098–106. [DOI] [PubMed] [Google Scholar]
  • 103.Chuang TY, Min J, Wu HL, McCrary C, Layman LC, Diamond MP, et al. Berberine Inhibits Uterine Leiomyoma Cell Proliferation via Downregulation of Cyclooxygenase 2 and Pituitary Tumor-Transforming Gene 1. Reprod Sci. 2017;24(7):1005–13. [DOI] [PubMed] [Google Scholar]
  • 104.Malik M, Britten J, Borahay M, Segars J, Catherino WH. Simvastatin, at clinically relevant concentrations, affects human uterine leiomyoma growth and extracellular matrix production. Fertil Steril. 2018;110(7):1398–407 e1. [DOI] [PubMed] [Google Scholar]
  • 105.Zhang D, Al-Hendy M, Richard-Davis G, Montgomery-Rice V, Rajaratnam V, Al-Hendy A. Antiproliferative and proapoptotic effects of epigallocatechin gallate on human leiomyoma cells. Fertil Steril. 2010;94(5):1887–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Chen HY, Huang TC, Lin LC, Shieh TM, Wu CH, Wang KL, et al. Fucoidan Inhibits the Proliferation of Leiomyoma Cells and Decreases Extracellular Matrix-Associated Protein Expression. Cell Physiol Biochem. 2018;49(5):1970–86. [DOI] [PubMed] [Google Scholar]
  • 107.Grudzien MM, Low PS, Manning PC, Arredondo M, Belton RJ Jr., Nowak RA. The antifibrotic drug halofuginone inhibits proliferation and collagen production by human leiomyoma and myometrial smooth muscle cells. Fertil Steril. 2010;93(4):1290–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Koohestani F, Qiang W, MacNeill AL, Druschitz SA, Serna VA, Adur M, et al. Halofuginone suppresses growth of human uterine leiomyoma cells in a mouse xenograft model. Hum Reprod. 2016;31(7):1540–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Greco S, Islam MS, Zannotti A, Delli Carpini G, Giannubilo SR, Ciavattini A, Petraglia F, Ciarmela P. Quercetin and indole-3-carbinol inhibit extracellular matrix expression in human primary uterine leiomyoma cells. Reprod Biomed Online. 2020;40(4):593–602. [DOI] [PubMed] [Google Scholar]
  • 110.Lin PH, Kung HL, Chen HY, Huang KC, Hsia SM. Isoliquiritigenin Suppresses E2-Induced Uterine Leiomyoma Growth through the Modulation of Cell Death Program and the Repression of ECM Accumulation. Cancers (Basel). 2019;11(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Janjusevic M, Greco S, Islam MS, Castellucci C, Ciavattini A, Toti P, et al. Locostatin, a disrupter of Raf kinase inhibitor protein, inhibits extracellular matrix production, proliferation, and migration in human uterine leiomyoma and myometrial cells. Fertil Steril. 2016;106(6):1530–8 e1. [DOI] [PubMed] [Google Scholar]
  • 112.Lee BS, Margolin SB, Nowak RA. Pirfenidone: a novel pharmacological agent that inhibits leiomyoma cell proliferation and collagen production. J Clin Endocrinol Metab. 1998;83(1):219–23. [DOI] [PubMed] [Google Scholar]
  • 113.Wu CH, Shieh TM, Wei LH, Cheng TF, Chen HY, Huang TC, Wang KL, Hsia SM. Resveratrol inhibits proliferation of myometrial and leiomyoma cells and decreases extracellular matrix-associated protein expression. J Funct Foods. 2016;23(May):241–52. [Google Scholar]
  • 114.Chen HY, Lin PH, Shih YH, Wang KL, Hong YH, Shieh TM, et al. Natural Antioxidant Resveratrol Suppresses Uterine Fibroid Cell Growth and Extracellular Matrix Formation In Vitro and In Vivo. Antioxidants (Basel). 2019;8(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Friedman N, Shushan A, Rojansky N, Shveiky D, Levitzki R, Chaouat M, et al. Targeting leiomyomas with all-trans-retinoic acid at phosphoinositide 3-kinase pathway suppression: Effective roles of beta-catenin and of signaling interactions. J Obstet Gynaecol Res. 2016;42(10):1343–53. [DOI] [PubMed] [Google Scholar]
  • 116.Giampieri F, Islam MS, Greco S, Gasparrini M, Forbes Hernandez TY, Delli Carpini G, et al. Romina: A powerful strawberry with in vitro efficacy against uterine leiomyoma cells. J Cell Physiol. 2019;234(5):7622–33. [DOI] [PubMed] [Google Scholar]
  • 117.Borahay MA, Kilic GS, Yallampalli C, Snyder RR, Hankins GD, Al-Hendy A, et al. Simvastatin potently induces calcium-dependent apoptosis of human leiomyoma cells. J Biol Chem. 2014;289(51):35075–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Borahay MA, Vincent K, Motamedi M, Sbrana E, Kilic GS, Al-Hendy A, et al. Novel effects of simvastatin on uterine fibroid tumors: in vitro and patient-derived xenograft mouse model study. Am J Obstet Gynecol. 2015;213(2):196 e1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Islam MS, Maher JY, Arfin S, Su S, Segars J. Verteporfin inhibits fibrosis, inflammation and angiogenesis related genes in uterine fibroid cells. Fertil Steril. 2019;112(3):e349. [Google Scholar]
  • 120.Ciebiera M, Ali M, Prince L, Jackson-Bey T, Atabiekov I, Zgliczynski S, et al. The Evolving Role of Natural Compounds in the Medical Treatment of Uterine Fibroids. J Clin Med. 2020;9(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Ali M, Prince L, Al-Hendy A. Vitamin D and uterine fibroids: preclinical evidence is in; time for an overdue clinical study! Fertil Steril. 2020;113(1):89–90. [DOI] [PubMed] [Google Scholar]
  • 122.Roell KR, Reif DM, Motsinger-Reif AA. An Introduction to Terminology and Methodology of Chemical Synergy-Perspectives from Across Disciplines. Front Pharmacol. 2017;8:158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Canbakan B. Rational approaches to the treatment of hypertension: drug therapy-monotherapy, combination, or fixed-dose combination? Kidney Int Suppl (2011). 2013;3(4):349–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Welsh L, Ferro A. Drug treatment of essential hypertension: the case for initial combination therapy. Int J Clin Pract. 2004;58(10):956–63. [DOI] [PubMed] [Google Scholar]
  • 125.Ali M, Shahin SM, Sabri NA, Al-Hendy A, Yang Q. 1,25 Dihydroxyvitamin D3 Enhances the Antifibroid Effects of Ulipristal Acetate in Human Uterine Fibroids. Reprod Sci. 2019;26(6):812–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Porcaro GS A; Giordano D; Angelozzi P;. Vitamin D plus epigallocatechin gallate: a novel promising approach for uterine myomas. Eur Rev Med Pharmacol Sci. 2020;24(6):3344–51. [DOI] [PubMed] [Google Scholar]

RESOURCES