Next Article in Journal
The Non-Canonical Aspects of MicroRNAs: Many Roads to Gene Regulation
Previous Article in Journal
Sex Differences in High Fat Diet-Induced Metabolic Alterations Correlate with Changes in the Modulation of GRK2 Levels
Previous Article in Special Issue
Late Onset of Estrogen Therapy Impairs Carotid Function of Senescent Females in Association with Altered Prostanoid Balance and Upregulation of the Variant ERα36
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Estrogen Receptors and Melanoma: A Review

1
Dermatology Section, Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, 40138 Bologna, Italy
2
Pathology Unit, Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, 40138 Bologna, Italy
3
Laboratory of Oncologic Molecular Pathology, S.Orsola-Malpighi Hospital, 40138 Bologna, Italy
4
Histology, Embryology and Applied Biology Unit Department of Experimental, Diagnostic and Specialty Medicine—DIMES University of Bologna, 40138 Bologna, Italy
5
Division of Dermatology, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
*
Author to whom correspondence should be addressed.
Cells 2019, 8(11), 1463; https://doi.org/10.3390/cells8111463
Submission received: 12 October 2019 / Revised: 14 November 2019 / Accepted: 16 November 2019 / Published: 19 November 2019
(This article belongs to the Special Issue Estrogen Receptor Expression and Function in Health and Disease)

Abstract

:
In the last three decades cutaneous melanoma has been widely investigated as a steroid hormone-sensitive cancer. Following this hypothesis, many epidemiological studies have investigated the relationship between estrogens and melanoma. No evidence to date has supported this association due to the great complexity of genetic, external and environmental factors underlying the development of this cancer. Molecular mechanisms through which estrogen and their receptor exert a role in melanoma genesis are still under investigation with new studies increasingly focusing on the discovery of new molecular targets for therapeutic treatments.

1. Introduction

Melanoma incidence continues to increase globally, giving rise to many questions about its exact pathogenesis [1]. Intriguingly, to date, most of the epidemiological data has noted a significant gender divergence in melanoma incidence as well as in other cancers. Indeed, in the last decades, melanoma incidence has been increasing more rapidly among males compared to females in all age categories, except for young women (≤39 years) who appear to be at higher risk. In line with these observations, slightly higher rates of melanoma have been reported for young women (20–45 years) which subsequently decreases after 45 years of age. On the other hand, in males, melanoma incidence progressively increases after 50 years of age [2,3,4,5].
The gender difference in melanoma survival appears to be limited to the early stages of the disease, since no difference in survival has been detected in advanced metastatic melanoma by the Surveillance, Epidemiology and End Results (SEER) database. This analysis was based on 106,511 cases of melanoma collected over 20 years in the United States, but not confined to pre- or perimenopausal age groups [5]. Confirming that for melanoma, women have a significant survival advantage over men [6]. Nowadays, this data is particularly relevant as women continue to delay childbearing into their 30′s and 40′s, increasing the likelihood of melanoma diagnosis during pregnancy [7]. Indeed, a meta-analysis of epidemiological studies highlighted that the risk of cutaneous melanoma is positively associated with the age of first pregnancy and inversely associated with parity [8]. Recent studies have suggested an important area of research to be female endogenous hormone exposure from menarche to menopause and its correlation with ultraviolet radiation [9].
The reasons why women seem to have a better prognosis are not completely understood. A large body of epidemiological studies have verified that gender is an independent prognostic factor after adjusting for other known melanoma risk factors such as: Breslow thickness, ulceration, histologic subtype, location, sentinel lymph node positivity and age [10,11,12]. Gender disparity in melanoma outcomes are consistently observed, leading to the suggestion that gender status should be incorporated in staging algorithms [13]. The gender disparity may be partially due to the differences in behavior and biology between the two genders. For example, women tend to have more ultraviolet protection and more frequent medical visits compared to men, who are more likely to have thicker, ulcerated tumors, as a result of diagnostic delays [14,15]. However, these factors don’t fully explain why gender is still a key prognostic factor after considering the gap between different lifestyle and healthcare delivery [16,17]. Regarding biology, many studies have pointed to differences in pharmacokinetics, immune response/inflammation and hormones between genders [18,19]. As is well known, there are gender differences in all the main pharmacokinetic parameters such as the absorption, distribution, metabolism, and elimination of drugs, all of which could impact the outcome of the disease [20].
Although melanoma is classically considered a non-hormone-related cancer, increasing evidence support a direct correlation between sex hormones (estrogens, in particular) and melanoma. Estrogen was found to play a role in the female survival advantage that seems to be abrogated in the postmenopausal period when estrogen levels decrease [20,21]. Additional evidence report that gender has an impact only in local tumor invasion [22], and that the gender effect is limited to lymphatic or hematogenous metastasis [23,24]. There are limited studies that consider metastatic patients, although, the results remain controversial [5,6,25,26]. The role of the immune system is crucial, particularly in the era of anticancer immunotherapy. There is evidence of a clear immunological difference between genders. Women have a better response in terms of effectiveness in both cellular and humoral immunity and exhibit a complex plasticity during the pregnancy, enabling them to “tolerate” a genetically different organism inside their bodies [19].

2. The Source of Data

The present review was conducted and reported using validated search strategies from the following databases:
  • PUBMED
  • Ovid MEDLINE
  • ISI Web of Science
The keywords and/or MESH terms used were: estrogen/oestrogen, estrogen/oestrogen receptors, and melanoma. Additional studies were found through cross-referencing of reference lists of the retrieved articles and previous reviews on the topic. Our search was not limited to human studies as it also considered molecular studies.

3. Estrogens and Melanoma: Epidemiological Studies

The distinct gender differences in relation to survival, local tumor invasion and more controversially, metastatic spread, warrant further investigation of the assumption that melanoma is a non-hormone related cancer [22,23,24,25,26]. Furthermore, the reduction of the survival advantage in postmenopausal women with melanoma provides an additional indication of the important immunological role of estrogen in relation to melanoma genesis and treatment [21,22].
More recent clinical, epidemiological, and laboratory studies have shed some light on the relationship among hormones, nevi, and melanoma in pregnancy. Similarities between the pathologic mechanisms of cancer and the physiologic process of placentation (e.g., proliferation, invasion, and local/systemic tolerance) have been researched for many years. Sex hormones such as human chorionic gonadotropin, estrogens, progesterone, and others have been shown to play contributory roles in the growth of sex hormone sensitive cancers such as breast, prostrate, endometrial and ovarian tumors [27]. Despite this, the involvement of sex hormones as putative mediators of the immunologic escape of cancer are still not known.
The sex hormones may be involved in a mechanism responsible for the immunotolerant environment during pregnancy, which blocks maternal T-cell responses and prevents fetal rejection. Indeed, progesterone and estrogen are important regulators of the immune system and angiogenesis in several types of cancers and in pregnancy [27,28]. Additionally, androgen receptors have been detected in melanoma cells, raising the hypothesis of its role in melanoma among men [29]. Questions regarding the relationship between the role of sex steroid hormones and melanoma have prompted studies examining epidemiological data about the impact of estrogen use for therapeutic purposes in patients with melanoma. A US study of 167,503 women, found a strong association between ambient UV exposure and melanoma in women who experienced menarche at an early age and menopause at a late age [9], further suggesting an important hormone related role in melanoma pathogenesis.
The possible link between exogenous sex steroid consumption and melanoma development has also been extensively investigated. Exogenous female hormones, either as oral contraceptives or as hormonal replacement among patients with a personal history of melanoma, have been investigated in order to find an association with the risk of developing melanoma. There is no convincing evidence that both the hormone replacement therapy and the oral contraceptive pill, effect the risk of developing melanoma. Moreover, hormone replacement therapy and oral contraceptives are not contraindicated in women who have had melanoma [30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45]. Recently a cohort study of 684,696 Norwegian women, found an increased risk of melanoma among menopausal women using estrogen-only hormone therapy (HT) (tablet and vaginal forms). Whereas, combined estrogen–progestin HT was not associated with an increased risk. This finding seems to suggest exogenous estrogen as a risk factor for melanoma. More studies are required to confirm these findings [46].
Several studies with conflicting results have focused on fertility drugs as drivers of melanoma. During in vitro fertilization (IVF) women are exposed to levels of estrogen and progesterone that can be 10 times greater than those present physiologically [47]. Previous studies have demonstrated an increased incidence of melanoma in women undergoing controlled ovarian stimulation (COS) [48,49] especially in relation to clomiphene citrate, a drug used in COS. Conversely, a number of studies have also found no association between malignant melanoma risk and the use of fertility drugs [50,51,52,53,54,55].

4. Estrogens and Melanoma: Molecular Studies

Steroid hormones such as estrogen act through their cognate receptors: Estrogen Receptor alfa (ERα) and Estrogen Receptor beta (ERβ). ERs belong to the nuclear receptor superfamily, which act as transcription factors upon homo- or heterodimers. Estrogen binding to the nuclear receptors are responsible for a nuclear translocation, with consequent activation of genomic pathways and transcription of multiple target genes. More rapid effects of steroid hormones are elicited by a “non-genomic pathway” activation. In this latter case, membrane-bound estrogen receptors are responsible for change in the cytosolic signaling, leading to increased activity of the RAS/BRAF/MEK axis [56] (Figure 1).
Notably, targeting the MAPK pathway by the BRAF inhibitor, vemurafenib, or the MEK inhibitor, dabrafenib, have been approved by the FDA for V600E mutant BRAF-harboring melanomas. Mutant BRAF is an important driver in melanoma development, occurring in about 50% of cutaneous melanomas. Thus, the cross-talk of steroid hormones with the route responsible for the melanoma transformation should be thoroughly addressed in order to determine the exact involvement of steroid hormones in melanoma development. The physiological relevance of the cross-talk of the steroid driven non-genomic pathway and MAPK activation was clearly demonstrated in an ERα KO mice model. Where the uterus displayed a growth response upon MAPK activation by epithelial growth factor. This action was not present with the absence of ERα, suggesting a key interaction between these two pathways. This is not surprising given a broad body of literature has suggested a coupling interaction of steroid hormones and growth factor receptor, with a fine-tuning of the MAPK/ERK axis, strongly impacting on cellular output in physiological or pathological conditions [57,58].
The estrogen receptors are encoded by two different genes that are located on chromosomes 6 and 14. Synthetic or natural ligands bind to ERα or ERβ with different affinities according to their chemical structure [56,57]. Increasing evidence supports a relationship between the perturbation of estrogen signaling and cancer initiation, promotion, and progression. Overall, it is now well accepted that ERα plays a role in tumorigenesis by stimulating cell proliferation, while ERβ seem to have a significant antitumor activity [57]
ERα is the main estrogen receptor in the human epidermis and its expression is decreased in benign or dysplastic nevi, in metastatic or primary melanoma, as well as in pregnancy-associated melanoma. Thus, the overall abundance of the receptor does not appear to be associated with the pathophysiology of melanoma precursor lesions or melanomas [59]. However, a recent immunohistochemical study reported a cytosolic localization of ERα in melanoma of women who underwent more than one cycle of in vitro fertilization [60]. In all individuals the probability of inherited susceptibility to melanoma could be partially due to different genomic ERα single nucleotide polymorphisms. These polymorphisims have been found to impact the course of the disease as well as the response to chemotherapy [61].
On the other hand, ERβ has been reported to be the predominant ER subtype in all melanocytic lesions such as moles, dysplastic nevi and melanoma [62]. Furthermore, ERβ expression is diminished after in vitro cellular exposure to UV and is associated with thinner lesions with a prominent epidermal component, such as severely dysplastic nevi or melanoma in situ. It has been suggested that ERβ could represent a marker for metastatic potential and prognosis in malignant melanoma [63]. ERβ receptor appears inversely associated with an increased Breslow depth [64] and displays a protective role in the metastatic process. Similar data has been reported in breast, prostate and ovarian tumors and colon cancers [65,66,67,68,69,70,71,72]. Furthermore, ERβ has been found to be more highly expressed in melanomas of pregnant women than in men, and a trend indicating a higher expression among women in comparison to men has been shown. This result was obtained by evaluating hormone receptor expression in the melanomas of stage and age-matched patients of pregnant women, non-pregnant women, and men [73].
An equally important type of estrogen receptor is the G protein-coupled estrogen receptor (GPER). GPER belongs to G protein-coupled receptor family of cellular membrane molecules that are found to be involved in development and progression of different cancer types [74]. GPER has specific agonists, G1 and 17β-estradiol. It is responsible for many cellular functions. In skin, it regulates melanin production and is expressed in melanoma cells [75]. It promotes melanogenesis via an intracellular pathway resulting in increasing cAMP levels. There is a subsequent activation of intracellular cAMP-protein kinase (PK) to an elevation of cAMP-response element-binding protein (CREB) phosphorylation as well as activation of microphthalmia-associated transcription factor (MITF). This latter regulates melanocyte growth, differentiation and function (Figure 1). GPER signaling pathway acts independently from ERs function [76,77]. In vitro GPER agonists inhibit melanoma cell proliferation. [75] GPER co-expressed with ERβ in melanoma have been found to have better outcomes, with lower Breslow thickness, lower mitotic rate and higher presence of peritumoral lymphocyte infiltration [78]

5. Present and Future Perspectives

Based on the above evidence, it is possible to postulate that ERβ might have a role in explaining the differences in melanoma prognosis between women and men. As stated previously, studies of estrogen-sensitive cancers showed that loss or decreasing levels of ERβ or an increased ERα to ERβ ratio may be involved in carcinogenesis, thereby suggesting that ERβ has a tumor-suppressive function.
In vitro, the ER antagonist tamoxifen (TAM) has been shown to induce cell death in malignant melanoma cells and to reduce metastatic behavior. Despite these promising results, the treatment of melanoma with TAM has not been shown to be effective in the treatment of advanced melanoma [79]. The effects of chemotherapy with and without TAM for the treatment of aggressive melanoma were compared in different clinical trials. These studies conclude that combination treatment with TAM and chemotherapy may result in improvements in response rates, although it is often accompanied by increased toxicity and no consistent survival benefit [79,80]. A recent meta-analysis, that included nine randomized controlled trials, showed that patients treated with TAM are more likely to respond to chemotherapy. However, no mortality improvement was demonstrated. The authors currently only suggest the use of TAM in clinical trials, given its high hematologic toxicity [80]
It has been hypothesized that tamoxifen decreases cell proliferation when it binds to ERα, while it may increase cell proliferation when it binds to ERβ. Thus, the antitumor vs. pro-survival effects of tamoxifen could depend on the different ERα/ERβ ratios in the given tissue [79,80]. In line with this observation, low levels of expression of ERβ were shown to correlate with tamoxifen resistance in breast cancer cells. There is also epidemiological evidence that women with breast cancer have an increased risk of melanoma and this risk is greater for patients who do not receive anti-estrogen therapy [81,82]. On the contrary, others have found no correlation between receptor beta and Breslow depth or disease stage at diagnosis, and no difference in expression between pregnant and non-pregnant women [83]. TAM likely has more than an ER antagonist action. Indeed, it behaves as an agonist of GPER in vitro and the role of this non-classical estrogen signaling pathway could be crucial to our understanding of its anticancer activity [74].
Endoxifen, an active metabolite of tamoxifen, has been shown to be a safe and effective anti-melanogenic agent in animal models. It exerts its activity by blocking ER transcription. It has been found to be up to 100 times more potent than TAM but with a more favorable toxicity and safety profile. For all these reasons endoxifen could be a promising new therapeutic agent for advanced melanoma [84]. Selective pharmacological activation of ERβ through its agonist LY500307 has been shown to have a potent suppressive action against lung metastasis from melanoma in vitro and in vivo. It acts by recruiting antitumor neutrophils to the metastatic niche though IL-1β, a potent chemotactic factor released from cancer cells upon ERβ stimulation. The activation of ERβ could augment the innate immunity’s ability to suppress cancer metastasis [85]. These pharmacological agents highlight the importance of estrogen signaling in melanoma management and may serve as an important future area of research.
Regarding estrogen signaling and the regulation of the immune system, Natale A. et al. demonstrated through a human-engineered melanoma xenografted into an animal model, that GPER signaling promotes differentiation in melanoma cells, inhibits cancer proliferation, and critically, promotes a cellular phenotype that makes the tumor more susceptible to immune checkpoint blockade [86].
The linkage between melanoma risk and hormonal/reproductive factors requires further investigation. In vitro, sex hormones and gonadotropins stimulate melanogenesis with direct action on the melanocytes. However, in vivo, their antagonists and agonists seem to be effective anticancer drugs. Although these mechanisms alone cannot completely explain all the clinical heterogeneity seen in melanoma cases.
Another issue that requires further investigation is the emerging interplay between microRNAs and estrogen receptor pathways. As is well known, microRNA (miRNA) expression is altered in cancer cells, where its dysregulation can play an oncogenic, metastatic or tumor suppressive role [87]. Several studies have shown the existence of a network involving ERs and miRNAs, either demonstrating a direct effect of estrogen administration in miRNA expression [88] or revealing a direct regulatory effect of miRNAs on ER expression, by directly targeting the 3′-UTR of ESR1/2 mRNAs. In cancer, miRNAs have been observed to be differentially expressed between ER- and ER+ breast cancers [89]. Adams et al. reported the first evidence of the direct regulation of ERα levels by miR-206 in breast cancer [90], an observation that has since been confirmed by other studies. Other research groups have worked with breast cancer cell lines or clinical samples to demonstrate the inhibitory effect caused by miR-22 [91], let-7 [92] or miR-92 [93] overexpression on ERα/β1 levels. This inhibition typically resulted in cell proliferation inhibition and apoptosis activation [92].
Recent studies have demonstrated the cross-talk between miRNAs, long non-coding RNAs (lncRNAs) and coding genes in ER+ and ER− subtypes of breast cancer. The observed networks were closely associated with clinical outcomes and proposed as a prognostic predictor. Specifically, Xiao et al. described that LINC0092 is co-expressed with SFRP1 and RGMA and regulated by miR-449a and miR-452-5p in ER+ breast cancer with better prognosis [94].
ERα-mediated estrogen signaling inhibits the level of miR-21, miR-26a, miR-140, miR-181b and miR-206, and upregulates miR-190a, miR-191, miR-203 and miR-425. Conversely, ERβ is associated with downregulation of miR-17, miR-30a, miR-200a and miR-200b, and overexpression of miR-23b, miR-24-1 and miR-27b [95]. Additionally, specific transcription factors are able to recognize estrogen response elements and activate miRNA transcription in an ER-dependent manner, as described by Nagpal et al. [96].
Estrogen receptors are expressed in different human tissues [97]. Given the tissue specificity of miRNA expression, a complex interplay between miRNAs and ER signaling is likely to exist. In the myometrium, the increased estradiol-17β/ERα in pre-labor inhibits miR-181a, resulting in a further increase in ERα and proinflammatory signals [98]. Conversely, in hepatocellular carcinoma (HCC) miR-18a was identified to target ESR 3′UTR, thus decreasing the ERα protein and stimulating cancer cell proliferation [99]. Additionally, miR-18a and miR-19a, were among the up-regulated miRNAs in MYCN-induced neuroblastoma that targeted and repressed the expression of estrogen receptor-α (ESR1) [100], which is a marker of neuronal differentiation.
A recent study by Vidal-Gómez et al. investigated the pattern of miRNAs induced by estradiol treatment of human umbilical vein endothelial cells (HUVEC) [101]. They described a complex network of up- and down-regulated miRNAs following ERα and ERβ and G protein-coupled estrogen receptor (GPER) stimulation. Given the expression of ERβ observed in melanoma cells, as well as the presence of GPER, it can be postulated that a similar pattern of miRNA activation and repression in the tumor may exist upon estrogen release in the circulation.
Participation in negative and positive feedback loops is another common mechanism of miRNA action in cancer cells, as reported by Di Leva et al. [102]. In this study, miR-221/222 negatively regulate estrogen receptor alpha at the post-transcriptional level, which in turn represses miR-221 and -222 expression. Felicetti et al. indicated that miR-221/222 are key regulators of melanoma development and dissemination [103,104]. In melanoma, miR-221/222 directly modulate KIT oncogene, which is mutated in acral and mucosal melanoma [105]; they also target p27 (CDKN1B) and p57 (CDKN1C) cell cycle inhibitors, thus promoting cell proliferation. We hypothesize that miRNAs play a significant role in co-regulating ERα, KIT, p27 and p57 to increase melanoma cell proliferation.
Recently, Milevskiy et al. described a complex regulatory model of miR-196a locus by ERα. The expression miR-196a is induced by estrogen and was linked to the development of drug-resistance in hormone therapy-responsive breast cancer [106].
The existing scientific literature does not suggest that ERα alone can completely explain melanoma development and progression. Further melanoma-specific studies are required to assess the composite interaction between all types of estrogen receptors, miRNA expression and the impact of both on the tumor microenvironment. The role of miRNA in cancer is an emerging area of research which may provide additional insight into the diagnosis and management of many types of malignancies. Currently, the majority of miRNA research is related to accepted hormone-related malignancies. However, as the evidence of the association between sex hormones and melanoma develops, the role of miRNA may represent a novel area of melanoma research in the future.

6. Concluding Remarks

In the era of personalized medicine and diagnostic-therapy, the evaluation of the ER isoforms and GPER expressions in melanoma patients should be considered in order to improve treatment response to novel and innovative therapies. Epidemiological data demonstrate that hormone replacement therapy and the use of the oral contraceptive pill don’t affect the natural history of melanoma and, at present, no association has been found between malignant melanoma risk and use of fertility drugs. Estrogen receptors are important regulators of many molecular pathways and their molecular investigations continue to shed light on our understanding of melanoma genesis and progression in all patients independent of gender.

Author Contributions

Conceptualization, E.D., A.P. and F.S.; data curation, E.D., A.P., M.L. (Martina Lambertini), N.M., A.A., M.F. (Michelangelo Fiorentino), G.V., M.F. (Manuela Ferracin), M.L. (Mattia Lauriola), E.F., E.C., and F.S.; validation, N.M., A.A., M.F. (Michelangelo Fiorentino), G.V., M.F. (Manuela Ferracin), M.L. (Mattia Lauriola), E.F., E.C., and F.S.; investigation, E.D., A.P., M.L. (Martina Lambertini), M.F. (Michelangelo Fiorentino), M.F. (Manuela Ferracin), M.L. (Mattia Lauriola), E.F., and F.S.; writing—original draft preparation, E.D., A.P., M.L. (Martina Lambertini), N.M., A.A., M.F. (Michelangelo Fiorentino), M.F. (Manuela Ferracin), M.L. (Mattia Lauriola), E.F. and F.S.; writing—review and editing, E.D., A.P., N.M., A.A., M.F. (Manuela Ferracin), G.V., E.C., and F.S.; visualization, E.D., A.P., M.L. (Martina Lambertini), N.M., A.A., M.F. (Michelangelo Fiorentino), G.V., M.F. (Manuela Ferracin), M.L. (Mattia Lauriola), E.F., E.C., and F.S.; supervision, E.D., A.P. and F.S.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Schadendorf, D.; van Akkooi, A.C.J.; Berking, C.; Griewank, K.G.; Gutzmer, R.; Hauschild, A.; Stang, A.A.A.; Roesch, A.A.; Ugurel, S. Melanoma. Lancet 2018, 392, 971–984. [Google Scholar] [CrossRef]
  2. Robsahm, T.E.; Bergva, G.; E Hestvik, U.; Møller, B. Sex differences in rising trends of cutaneous malignant melanoma in Norway, 1954–2008. Melanoma Res. 2013, 23, 70–78. [Google Scholar] [CrossRef]
  3. Weir, H.K.; Marrett, L.D.; Cokkinides, V.; Barnholtz-Sloan, J.; Patel, P.; Tai, E.; Jemal, A.; Li, J.; Kim, J.; Ekwueme, D.U. Melanoma in adolescents and young adults (ages 15–39 years): United States, 1999–2006. J. Am. Acad. Derm. 2011, 65, S38–S49. [Google Scholar] [CrossRef]
  4. Reed, K.B.; Brewer, J.D.; Lohse, C.M.; Bringe, K.E.; Pruitt, C.N.; Gibson, L.E. Increasing incidence of melanoma among young adults: An epidemiological study in Olmsted County, Minnesota. Mayo Clin. Proc. 2012, 87, 328–334. [Google Scholar] [CrossRef]
  5. Enninga, E.A.L.; Moser, J.C.; Weaver, A.L.; Markovic, S.N.; Brewer, J.D.; Leontovich, A.A.; Hieken, T.J.; Shuster, L.; Kottschade, L.A.; Olariu, A.; et al. Survival of cutaneous melanoma based on sex, age, and stage in the United States, 1992–2011. Cancer Med. 2017, 6, 2203–2212. [Google Scholar] [CrossRef]
  6. Joosse, A.; Collette, S.; Suciu, S.; Nijsten, T.; Lejeune, F.; Kleeberg, U.R.; Coebergh, J.W.; Eggermont, A.M.; de Vries, E. Superior outcome of women with stage I/II cutaneous melanoma: Pooled analysis of four european organisation for research and treatment of cancer phase III trials. J. Clin. Oncol. 2012, 30, 2240–2247. [Google Scholar] [CrossRef]
  7. Ribero, S.; Longo, C.; Dika, E.; Fortes, C.; Pasquali, S.; Nagore, E.; Glass, D.; Robert, C.; Eggermont, A.M.; Testori, A.; et al. Members of the Melanoma Group of the EORTC. Pregnancy and melanoma: A European-wide survey to assess current management and a critical literature overview. J. Eur. Acad. Dermatol. Venereol. 2017, 31, 65–69. [Google Scholar] [CrossRef]
  8. Gandini, S.; Iodice, S.; Koomen, E.; Di Pietro, A.; Sera, F.; Caini, S. Hormonal and reproductive factors in relation to melanoma in women: Current review and meta-analysis. Eur. J. Cancer 2011, 47, 2607–2617. [Google Scholar] [CrossRef]
  9. Donley, G.M.; Liu, W.T.; Pfeiffer, R.M.; McDonald, E.C.; Peters, K.O.; Tucker, M.A.; Cahoon, E.K. Reproductive factors, exogenous hormone use and incidence of melanoma among women in the United States. Br. J. Cancer 2019, 120, 754–760. [Google Scholar] [CrossRef]
  10. Lasithiotakis, K.; Leiter, U.; Meier, F.; Eigentler, T.; Metzler, G.; Moehrle, M.; Breuninger, H.; Garbe, C. Age and gender are significant independent predictors of survival in primary cutaneous melanoma. Cancer 2008, 112, 1795–1804. [Google Scholar] [CrossRef]
  11. Joosse, A.; Collette, S.; Suciu, S.; Nijsten, T.; Patel, P.M.; Keilholz, U.; Eggermont, A.M.M.; Coebergh, J.W.W.; De Vries, E. Sex is an independent prognostic indicator for survival and relapse/progression-free survival in metastasized stage III to IV melanoma: A pooled analysis of five European organisation for research and treatment of cancer randomized controlled trials. J. Clin. Oncol. 2013, 31, 2337–2346. [Google Scholar] [CrossRef]
  12. Gamba, C.S.; Clarke, C.A.; Keegan, T.H.; Tao, L.; Swetter, S.M. Melanoma survival disadvantage in young, non-Hispanic white males compared with females. JAMA Derm. 2013, 149, 912–920. [Google Scholar] [CrossRef]
  13. Roh, M.R.; Eliades, P.; Gupta, S.; Grant-Kels, J.M.; Tsao, H. Cutaneous melanoma in women. Int. J. Womens Dermatol. 2017, 3, S11–S15. [Google Scholar] [CrossRef]
  14. Courtenay, W.H.; Keeling, R.P. Men, gender, and health: Toward an interdisciplinary approach. J. Am. Coll. Health 2000, 48, 243–246. [Google Scholar] [CrossRef]
  15. Abel, G.A.; Shelton, J.; Johnson, S.; Elliss-Brookes, L.; Lyratzopoulos, G. Cancer-specific variation in emergency presentation by sex, age and deprivation across 27 common and rarer cancers. Br. J. Cancer 2015, 112, S129–S136. [Google Scholar] [CrossRef]
  16. Micheli, A.; Ciampichini, R.; Oberaigner, W.; Ciccolallo, L.; De Vries, E.; Izarzugaza, I.; Zambon, P.; Gatta, G.; De Angelis, R. The advantage of women in cancer survival: An analysis of EUROCARE-4 data. Eur. J. Cancer 2009, 45, 1017–1027. [Google Scholar] [CrossRef]
  17. Schwartz, M.R.; Luo, L.; Berwick, M. Sex differences in melanoma. Curr. Epidemiol. Rep. 2019, 6, 112–118. [Google Scholar] [CrossRef]
  18. Fischer, J.; Jung, N.; Robinson, N.; Lehmann, C. Sex differences in immune responses to infectious diseases. Infection 2015, 43, 399–403. [Google Scholar] [CrossRef]
  19. Franconi, F.; Campesi, I. Pharmacogenomics, pharmacokinetics and pharmacodynamics: Interaction with biological differences between men and women. Br. J. Pharm. 2014, 171, 580–594. [Google Scholar] [CrossRef]
  20. Kemeny, M.M.; Busch, E.; Stewart, A.K.; Menck, H.R. Superior survival of young women with malignant melanoma. Am. J. Surg. 1998, 175, 437–444. [Google Scholar] [CrossRef]
  21. Mervic, L.; Leiter, U.; Meier, F.; Eigentler, T.; Forschner, A.; Metzler, G.; Bartenjev, I.; Büttner, P.; Garbe, C. Sex differences in survival of cutaneous melanoma are age dependent: An analysis of 7338 patients. Melanoma Res. 2011, 21, 244–252. [Google Scholar] [CrossRef] [PubMed]
  22. Molife, R.; Lorigan, P.; MacNeil, S. Gender and survival in malignant tumours. Cancer Treat. Rev. 2001, 27, 201–209. [Google Scholar] [CrossRef] [PubMed]
  23. Richardson, B.; Price, A.; Wagner, M.; Williams, V.; Lorigan, P.; Browne, S.; Miller, J.G.; Mac Neil, S. Investigation of female survival benefit in metastatic melanoma. Br. J. Cancer 1999, 80, 2025–2033. [Google Scholar] [CrossRef] [PubMed]
  24. Scoggins, C.R.; Ross, M.I.; Reintgen, D.S.; Noyes, R.D.; Goydos, J.S.; Beitsch, P.D.; Urist, M.M.; Ariyan, S.; Sussman, J.J.; Edwards, M.J.; et al. Gender-related differences in outcome for melanoma patients. Ann. Surg. 2006, 243, 693–698. [Google Scholar] [CrossRef]
  25. Unger, J.M.; Flaherty, L.E.; Liu, P.; Albain, K.S.; Sondak, V.K. Gender and other survival predictors in patients with metastatic melanoma on Southwest Oncology Group trials. Cancer 2001, 91, 1148–1155. [Google Scholar] [CrossRef]
  26. Korn, E.L.; Liu, P.-Y.; Lee, S.J.; Chapman, J.-A.W.; Niedzwiecki, D.; Suman, V.J.; Moon, J.; Sondak, V.K.; Atkins, M.B.; Eisenhauer, E.A.; et al. Meta-analysis of phase ii cooperative group trials in metastatic stage iv melanoma to determine progression-free and overall survival benchmarks for future phase II trials. J. Clin. Oncol. 2008, 26, 527–534. [Google Scholar] [CrossRef]
  27. Folkerd, E.J.; Dowsett, M. Influence of sex hormones on cancer progression. J. Clin. Oncol. 2010, 28, 4038–4044. [Google Scholar] [CrossRef] [PubMed]
  28. Bieber, A.K.; Martires, K.J.; Stein, J.A.; Grant-Kels, J.M.; Driscoll, M.S.; Pomeranz, M.K. Pigmentation and pregnancy: Knowing what is normal. Obstet. Gynecol. 2016, 129, 168–173. [Google Scholar] [CrossRef]
  29. Morvillo, V.; Lüthy, I.A.; Bravo, A.I.; Capurro, M.I.; Portela, P.; Calandra, R.S.; Mordoh, J. Androgen receptors in human melanoma cell lines IIB-MEL-LES and IIB-MEL-IAN and in human melanoma metastases. Melanoma Res. 2002, 12, 529–538. [Google Scholar] [CrossRef]
  30. Holly, E.A.; Cress, R.D.; Ahn, D.K. Cutaneous melanoma in women: Ovulatory life, menopause, and use of exogenous estrogens. Cancer Epidemiol. Biomark. Prev. 1994, 3, 661–668. [Google Scholar] [CrossRef]
  31. Karagas, M.R.; Stukel, T.A.; Dykes, J.; Miglionico, J.; Greene, M.A.; Carey, M.; Armstrong, B.; Elwood, J.M.; Gallagher, R.P.; Green, A.; et al. A pooled analysis of 10 case–control studies of melanoma and oral contraceptive use. Br. J. Cancer 2002, 86, 1085–1092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Holman, C.D.; Armstrong, B.K.; Heenan, P.J. Cutaneous malignant melanoma in women: Exogenous sex hormones and reproductive factors. Br. J. Cancer 1984, 50, 673–680. [Google Scholar] [CrossRef]
  33. Gallagher, R.P.; Elwood, J.M.; Hill, G.B.; Coldman, A.J.; Threlfall, W.J.; Spinelli, J.J. Reproductive factors, oral contraceptives and risk of malignant melanoma: Western Canada Melanoma Study. Br. J. Cancer 1985, 52, 901–907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Green, A.; Bain, C. Hormonal factors and melanoma in women. Med. J. Aust. 1985, 142, 446–448. [Google Scholar] [CrossRef] [PubMed]
  35. Zanetti, R.; Franceschi, S.; Rosso, S.; Bidoli, E.; Colonna, S. Cutaneous malignant melanoma in females: The role of hormonal and reproductive factors. Int. J. Epidemiol. 1990, 19, 522–526. [Google Scholar] [CrossRef] [PubMed]
  36. Beral, V.; Ramcharan, S.; Faris, R. Malignant melanoma and oral contraceptive use among women in California. Br. J. Cancer 1977, 36, 804–809. [Google Scholar] [CrossRef]
  37. Hannaford, P.; Villard-Mackintosh, L.; Vessey, M.; Kay, C. Oral contraceptives and malignant melanoma. Br. J. Cancer 1991, 63, 430–433. [Google Scholar] [CrossRef] [Green Version]
  38. Adam, S.A.; Sheaves, J.K.; Wright, N.H.; Mosser, G.; Harris, R.W.; Vessey, M.P. A case-control study of the possible association between oral contraceptives and malignant melanoma. Br. J. Cancer 1981, 44, 45–50. [Google Scholar] [CrossRef] [Green Version]
  39. Palmer, J.R.; Rosenberg, L.; Strom, B.L.; Harlap, S.; Zauber, A.G.; Warshauer, M.E.; Shapiro, S. Oral contraceptive use and risk of cutaneous malignant melanoma. Cancer Causes Control 1992, 3, 547–554. [Google Scholar] [CrossRef]
  40. Cabanes, P.A.; Desvignes, V.; Chanteau, M.F.; Mlika, N.; Avril, M.F. Oral contraceptive use and risk of cutaneous malignant melanoma in a case-control study of French women. Cancer Causes Control 1992, 3, 199–205. [Google Scholar]
  41. Osterlind, A.; Tucker, M.A.; Stone, B.J.; Jensen, O.M. The Danish case-control study of cutaneous malignant melanoma. III. Hormonal and reproductive factors in women. Int. J. Cancer 1988, 42, 821–824. [Google Scholar] [PubMed]
  42. Helmrich, S.P.; Rosenberg, L.; Kaufman, D.W.; Miller, D.R.; Schottenfeld, D.; Stolley, P.D.; Shapiro, S. Lack of an elevated risk of malignant melanoma in relation to oral contraceptive use. J. Natl. Cancer Inst. 1984, 72, 617–620. [Google Scholar] [PubMed]
  43. Bain, C.; Hennekens, C.H.; Speizer, F.E.; Rosner, B.; Willett, W.; Belanger, C. Oral contraceptive use and malignant melanoma. J. Natl. Cancer Inst. 1982, 68, 537–539. [Google Scholar] [PubMed]
  44. Holly, E.A.; Cress, R.D.; Ahn, D.K. Cutaneous melanoma in women. III. Reproductive factors and oral contraceptive use. Am. J. Epidemiol. 1995, 141, 943–950. [Google Scholar] [CrossRef] [PubMed]
  45. Westerdahl, J.; Olsson, H.; Måsbäck, A.; Ingvar, C.; Jonsson, N. Risk of malignant melanoma in relation to drug intake, alcohol, smoking and hormonal factors. Br. J. Cancer 1996, 73, 1126–1131. [Google Scholar] [CrossRef] [PubMed]
  46. Beral, V.; Evans, S.; Shaw, H.; Milton, G. Oral contraceptive use and malignant melanoma in Australia. Br. J. Cancer 1984, 50, 681–685. [Google Scholar] [CrossRef] [PubMed]
  47. Botteri, E.; Støer, N.C.; Sakshaug, S.; Graff-Iversen, S.; Vangen, S.; Hofvind, S.; Ursin, G.; Weiderpass, E. Menopausal hormone therapy and risk of melanoma: Do estrogens and progestins have a different role? Int. J. Cancer 2017, 141, 1763–1770. [Google Scholar] [CrossRef]
  48. Joo, B.S.; Park, S.H.; An, B.M.; Kim, K.S.; Moon, S.E.; Moon, H.S. Serum estradiol levels during controlled ovarian hyperstimulation influence the pregnancy outcome of in vitro fertilization in a concentration-dependent manner. Fertil. Steril. 2010, 93, 442–446. [Google Scholar] [CrossRef]
  49. Althuis, M.D.; Scoccia, B.; Lamb, E.J.; Althuis, M.D.; Scoccia, B.; Lamb, E.J.; Moghissi, K.S.; Westhoff, C.L.; Mabie, J.E.; Brinton, L.A. Melanoma, thyroid, cervical, and colon cancer risk after use of fertility drugs. Am. J. Obstet. Gynecol. 2005, 193, 668–674. [Google Scholar] [CrossRef]
  50. Brinton, L.A.; Moghissi, K.S.; Scoccia, B.; Lamb, E.J.; Trabert, B.; Niwa, S.; Ruggieri, D.; Westhoff, C.L. Effects of fertility drugs on cancers other than breast and gynecologic malignancies. Fertil. Steril. 2015, 104, 980–988. [Google Scholar] [CrossRef] [Green Version]
  51. Young, P.; Purdie, D.; Jackman, L.; Molloy, D.; Green, A. A study of infertility treatment and melanoma. Melanoma Res. 2001, 11, 535–541. [Google Scholar] [CrossRef] [PubMed]
  52. Venn, A.; Watson, L.; Lumley, J.; Gilles, G.; King, C.; Healy, D. Breast and ovarian cancer incidence after infertility and in vitro fertilisation. Lancet 1995, 346, 995–1000. [Google Scholar] [CrossRef]
  53. Silva Idos, S.; Wark, P.A.; McCormack, V.A.; Mayer, D.; Overton, C.; Little, V.; MacLean, A.B. Ovulation stimulation drugs and cancer risks: A long-term follow-up of a British cohort. Br. J. Cancer 2009, 100, 1824–1831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Yli-Kuha, A.-N.; Gissler, M.; Klemetti, R.; Luoto, R.; Hemminki, E. Cancer morbidity in a cohort of 9175 Finnish women treated for infertility. Hum. Reprod. 2012, 27, 1149–1155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Spaan, M.; Schaapveld, M.; Schats, R.; Kortman, M.; Belt-Dusebout, A.V.D.; Mooij, T.; Burger, C.; Van Leeuwen, F.; Lambalk, C.; Laven, J.; et al. Melanoma risk after ovarian stimulation for in vitro fertilization. Hum. Reprod. 2015, 30, 1216–1228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Eyster, K.M. The Estrogen Receptors: An Overview from Different Perspectives. Adv. Struct. Saf. Stud. 2016, 1366, 1–10. [Google Scholar]
  57. Barone, I.; Brusco, L.; Fuqua, S.A. Estrogen receptor mutations and changes in downstream gene expression and signaling. Clin. Cancer Res. 2010, 16, 2702–2708. [Google Scholar] [CrossRef] [Green Version]
  58. Lauriola, M.; Enuka, Y.; Zeisel, A.; D’Uva, G.; Roth, L.; Sharon-Sevilla, M.; Lindzen, M.; Sharma, K.; Nevo, N.; Feldman, M.; et al. Diurnal suppression of EGFR signalling by glucocorticoids and implications for tumour progression and treatment. Nat. Commun. 2014, 5, 5073. [Google Scholar] [CrossRef] [Green Version]
  59. Zhou, J.H.; Kim, K.B.; Myers, J.N.; Fox, P.S.; Ning, J.; Bassett, R.L.; Hasanein, H.; Prieto, V.G. Immunohistochemical expression of hormone receptors in melanoma of pregnant women, nonpregnant women, and men. Am. J. Derm. 2014, 36, 74–79. [Google Scholar] [CrossRef]
  60. Dika, E.; Fanti, P.A.; Vaccari, S.; Capizzi, E.; DeGiovanni, A.; Gobbi, A.; Piraccini, B.M.; Ribero, S.; Baraldi, C.; Ravaioli, G.M.; et al. Oestrogen and progesterone receptors in melanoma and nevi: An immunohistochemical study. Eur. J. Derm. 2017, 27, 254–259. [Google Scholar] [CrossRef]
  61. Glatthaar, H.; Katto, J.; Vogt, T.; Mahlknecht, U. Estrogen Receptor Alpha (ESR1) Single-Nucleotide Polymorphisms (SNPs) Affect Malignant Melanoma Susceptibility and Disease Course. Genet. Epigenet. 2016, 8, 1–6. [Google Scholar] [CrossRef] [PubMed]
  62. Marzagalli, M.; Montagnani Marelli, M.; Casati, L.; Fontana, F.; Moretti, R.M.; Limonta, P. Estrogen Receptor β in Melanoma: From Molecular Insights to Potential Clinical Utility. Front. Endocrinol. 2016, 7, 140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. De Giorgi, V.; Mavilia, C.; Massi, D.; Sestini, S.; Grazzini, M.; Brandi, M.L.; Lotti, T. The role of estrogens in melanoma and skin cancer. Carcinogenesis 2009, 30, 720. [Google Scholar] [CrossRef] [PubMed]
  64. De Giorgi, V.; Mavilia, C.; Massi, D.; Gozzini, A.; Aragona, P.; Tanini, A.; Sestini, S.; Paglierani, M.; Boddi, V.; Brandi, M.L.; et al. Estrogen receptor expression in cutaneous melanoma: A real-time reverse transcriptase-polymerase chain reaction and immunohistochemical study. Arch. Derm. 2009, 145, 30–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Taylor, A.; Al-Azzawi, F.; Ognjanovic, S.; Bao, S.; Yamamoto, S.; Garibay-Tupas, J.; Samal, B.; Bryant-Greenwood, G. Immunolocalisation of oestrogen receptor beta in human tissues. J. Mol. Endocrinol. 2000, 24, 145–155. [Google Scholar] [CrossRef] [PubMed]
  66. Fox, E.M.; Davis, R.J.; Shupnik, M.A. ERbeta in breast cancer--onlooker, passive player, or active protector? Steroids 2008, 73, 1039–1051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Hartman, J.; Ström, A.; Gustafsson, J.A. Estrogen receptor beta in breast cancer—diagnostic implications. Steroids 2009, 74, 635–641. [Google Scholar] [CrossRef]
  68. Roger, P.; Sahla, M.E.; Mäkelä, S.; Gustafsson, J.A.; Baldet, P.; Rochefort, H. Decreased expression of estrogen receptor beta protein in proliferative preinvasive mammary tumors. Cancer Res. 2001, 61, 2537–2541. [Google Scholar]
  69. Hartman, J.; Edvardsson, K.; Lindberg, K.; Zhao, C.; Williams, C.; Ström, A.; Gustafsson, J.A. Tumor repressive functions of estrogen receptor beta in SW480 colon cancer cells. Cancer Res. 2009, 69, 6100–6106. [Google Scholar] [CrossRef] [Green Version]
  70. Drummond, A.E.; Fuller, P.J. The importance of ERbeta signalling in the ovary. J. Endocrinol. 2010, 205, 15–23. [Google Scholar] [CrossRef]
  71. Thomas, C.G.; Ström, A.; Lindberg, K.; Gustafsson, J.-Å. Estrogen receptor beta decreases survival of p53-defective cancer cells after DNA damage by impairing G2/M checkpoint signaling. Breast Cancer Res. Treat. 2010, 127, 417–427. [Google Scholar] [CrossRef] [PubMed]
  72. Lindberg, K.; Ström, A.; Lock, J.G.; Gustafsson, J.A.; Haldosén, L.A.; Helguero, L.A. Expression of estrogen receptor beta increases integrin alpha1 and integrin beta1 levels and enhances adhesion of breast cancer cells. J. Cell. Physiol. 2010, 222, 156–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Schmidt, A.N.; Nanney, L.B.; Boyd, A.S.; King, L.E., Jr.; Ellis, D.L. Oestrogen receptor-beta expression in melanocytic lesions. Exp. Derm. 2006, 15, 971–980. [Google Scholar] [CrossRef] [PubMed]
  74. Lee, H.J.; Wall, B.; Chen, S. G-protein-coupled receptors and melanoma Pigment. Cell Melanoma Res. 2008, 21, 415–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Ribeiro, M.P.C.; Santos, A.E.; Custódio, J.B.A. The activation of the G protein-coupled estrogen receptor (GPER) inhibits the proliferation of mouse melanoma K1735-M2 cells. Chem. Biol. Interact. 2017, 277, 176–184. [Google Scholar] [CrossRef]
  76. Sun, M.; Xie, H.F.; Tang, Y.; Lin, S.Q.; Li, J.M.; Sun, S.N.; Hu, X.L.; Huang, Y.X.; Shi, W.; Jian, D. G protein-coupled estrogen receptor enhances melanogenesis via cAMP-protein kinase (PKA) by upregulating microphthalmia-related transcription factor-tyrosinase in melanoma. J. Steroid Biochem. Mol. Biol. 2017, 165, 236–246. [Google Scholar] [CrossRef]
  77. Natale, C.A.; Duperret, E.K.; Zhang, J.; Sadeghi, R.; Dahal, A.; O’Brien, K.T.; Cookson, R.; Winkler, J.D.; Ridky, T.W. Sex steroids regulate skin pigmentation through nonclassical membrane-bound receptors. eLife 2016, 5, e15104. [Google Scholar] [CrossRef]
  78. Fábián, M.; Rencz, F.; Krenács, T.; Brodszky, V.; Hársing, J.; Németh, K.; Balogh, P.; Kárpáti, S. Expression of G protein-coupled oestrogen receptor in melanoma and in pregnancy-associated melanoma. J. Eur. Acad. Derm. Venereol. 2017, 31, 1453–1461. [Google Scholar] [CrossRef]
  79. Lens, M.B.; Reiman, T.; Husain, A.F. Use of tamoxifen in the treatment of malignant melanoma. Cancer 2003, 98, 1355–1361. [Google Scholar] [CrossRef]
  80. Beguerie, J.R.; Xingzhong, J.; Valdez, R.P. Tamoxifen vs. non-tamoxifen treatment for advanced melanoma: A meta-analysis. Int. J. Derm. 2010, 49, 1194–1202. [Google Scholar] [CrossRef]
  81. Ricceri, F.; Fasanelli, F.; Giraudo, M.T.; Sieri, S.; Tumino, R.; Mattiello, A.; Vagliano, L.; Masala, G.; Quirós, J.R.; Travier, N.; et al. Risk of second primary malignancies in women with breast cancer: Results from the European prospective investigation into cancer and nutrition (EPIC). Int. J. Cancer. 2015, 137, 940–948. [Google Scholar] [CrossRef] [PubMed]
  82. Huber, C.; Bouchardy, C.; Schaffar, R.; Neyroud-Caspar, I.; Vlastos, G.; Le Gal, F.A.; Rapiti, E.; Benhamou, S. Antiestrogen therapy for breast cancer modifies the risk of subsequent cutaneous melanoma. Cancer Prev. Res. 2012, 5, 82–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Lens, M.; Bataille, V. Melanoma in relation to reproductive and hormonal factors in women: Current review on controversial issues. Cancer Causes Control 2008, 19, 437–442. [Google Scholar] [CrossRef] [PubMed]
  84. Chen, P.; Sheikh, S.; Ahmad, A.; Ali, S.M.; Ahmad, M.U.; Ahmad, I. Orally administered endoxifen inhibits tumor growth in melanoma-bearing mice. Cell Mol. Biol. Lett. 2018, 23, 3. [Google Scholar] [CrossRef] [Green Version]
  85. Zhao, L.; Huang, S.; Mei, S.; Yang, Z.; Xu, L.; Zhou, N.; Yang, Q.; Shen, Q.; Wang, W.; Le, X.; et al. Pharmacological activation of estrogen receptor beta augments innate immunity to suppress cancer metastasis. Proc. Natl. Acad. Sci. USA 2018, 115, E3673–E3681. [Google Scholar] [CrossRef] [Green Version]
  86. Natale, C.A.; Li, J.; Zhang, J.; Dahal, A.; Dentchev, T.; Stanger, B.Z.; Ridky, T.W. Activation of G protein-coupled estrogen receptor signaling inhibits melanoma and improves response to immune checkpoint blockade. eLife 2018, 7, e31770. [Google Scholar] [CrossRef]
  87. Volinia, S.; Calin, G.A.; Liu, C.G.; Ambs, S.; Cimmino, A.; Petrocca, F.; Visone, R.; Iorio, M.; Roldo, C.; Ferracin, M.; et al. A microrna expression signature of human solid tumors defines cancer gene targets. Proc. Natl. Acad. Sci. USA 2006, 103, 2257–2261. [Google Scholar] [CrossRef] [Green Version]
  88. Maillot, G.; Lacroix-Triki, M.; Pierredon, S.; Gratadou, L.; Schmidt, S.; Benes, V.; Roche, H.; Dalenc, F.; Auboeuf, D.; Millevoi, S.; et al. Widespread estrogen-dependent repression of micrornas involved in breast tumor cell growth. Cancer Res. 2009, 69, 8332–8340. [Google Scholar] [CrossRef]
  89. Iorio, M.V.; Ferracin, M.; Liu, C.-G.; Veronese, A.; Spizzo, R.; Sabbioni, S.; Magri, E.; Pedriali, M.; Fabbri, M.; Campiglio, M.; et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005, 65, 7065–7070. [Google Scholar] [CrossRef]
  90. Adams, B.D.; Furneaux, H.; White, B.A. The micro-ribonucleic acid (mirna) mir-206 targets the human estrogen receptor-alpha (eralpha) and represses eralpha messenger rna and protein expression in breast cancer cell lines. Mol. Endocrinol. 2007, 21, 1132–1147. [Google Scholar] [CrossRef]
  91. Pandey, D.P.; Picard, D. Mir-22 inhibits estrogen signaling by directly targeting the estrogen receptor alpha mrna. Mol. Cell Biol. 2009, 29, 3783–3790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Zhao, Y.; Deng, C.; Wang, J.; Xiao, J.; Gatalica, Z.; Recker, R.R.; Xiao, G.G. Let-7 family mirnas regulate estrogen receptor alpha signaling in estrogen receptor positive breast cancer. Breast Cancer Res. Treat. 2011, 127, 69–80. [Google Scholar] [CrossRef] [PubMed]
  93. Al-Nakhle, H.; Burns, P.A.; Cummings, M.; Hanby, A.M.; Hughes, T.A.; Satheesha, S.; Shaaban, A.M.; Smith, L.; Speirs, V. Estrogen receptor beta 1 expression is regulated by mir-92 in breast cancer. Cancer Res. 2010, 70, 4778–4784. [Google Scholar] [CrossRef] [PubMed]
  94. Xiao, B.; Zhang, W.; Chen, L.; Hang, J.; Wang, L.; Zhang, R.; Liao, Y.; Chen, J.; Ma, Q.; Sun, Z.; et al. Analysis of the mirna-mrna-lncrna network in human estrogen receptor-positive and estrogen receptor-negative breast cancer based on tcga data. Gene 2018, 658, 28–35. [Google Scholar] [CrossRef]
  95. Vrtacnik, P.; Ostanek, B.; Mencej-Bedrac, S.; Marc, J. The many faces of estrogen signaling. Biochem. Med. 2014, 24, 329–342. [Google Scholar] [CrossRef] [Green Version]
  96. Nagpal, N.; Sharma, S.; Maji, S.; Durante, G.; Ferracin, M.; Thakur, J.K.; Kulshreshtha, R. Essential role of med1 in the transcriptional regulation of er-dependent oncogenic mirnas in breast cancer. Sci. Rep. 2018, 8, 11805. [Google Scholar] [CrossRef]
  97. Bottner, M.; Thelen, P.; Jarry, H. Estrogen receptor beta: Tissue distribution and the still largely enigmatic physiological function. J. Steroid Biochem. Mol. Biol. 2014, 139, 245–251. [Google Scholar] [CrossRef]
  98. Gao, L.; Wang, G.; Liu, W.N.; Kinser, H.; Franco, H.L.; Mendelson, C.R. Reciprocal feedback between mir-181a and e-2/er alpha in myometrium enhances inflammation leading to labor. J. Clin. Endocrinol. Metab. 2016, 101, 3646–3656. [Google Scholar] [CrossRef]
  99. Liu, W.H.; Yeh, S.H.; Lu, C.C.; Yu, S.L.; Chen, H.Y.; Lin, C.Y.; Chen, D.S.; Chen, P.J. Microrna-18a prevents estrogen receptor-alpha expression, promoting proliferation of hepatocellular carcinoma cells. Gastroenterology 2009, 136, 683–693. [Google Scholar] [CrossRef]
  100. Loven, J.; Zinin, N.; Wahlstrom, T.; Muller, I.; Brodin, P.; Fredlund, E.; Ribacke, U.; Pivarcsi, A.; Pahlman, S.; Henriksson, M. Mycn-regulated micrornas repress estrogen receptor-alpha (esr1) expression and neuronal differentiation in human neuroblastoma. Proc. Natl. Acad. Sci. USA 2010, 107, 1553–1558. [Google Scholar] [CrossRef] [Green Version]
  101. Vidal-Gomez, X.; Perez-Cremades, D.; Mompeon, A.; Dantas, A.P.; Novella, S.; Hermenegildo, C. Microrna as crucial regulators of gene expression in estradiol-treated human endothelial cells. Cell. Physiol. Biochem. 2018, 45, 1878–1892. [Google Scholar] [CrossRef] [PubMed]
  102. Di Leva, G.; Gasparini, P.; Piovan, C.; Ngankeu, A.; Garofalo, M.; Taccioli, C.; Iorio, M.V.; Li, M.; Volinia, S.; Alder, H.; et al. Microrna cluster 221–222 and estrogen receptor alpha interactions in breast cancer. JNCI-J. Natl. Cancer Inst. 2010, 102, 706–721. [Google Scholar] [CrossRef] [PubMed]
  103. Felicetti, F.; De Feo, A.; Coscia, C.; Puglisi, R.; Pedini, F.; Pasquini, L.; Bellenghi, M.; Errico, M.C.; Pagani, E.; Care, A. Exosome-mediated transfer of mir-222 is sufficient to increase tumor malignancy in melanoma. J. Transl. Med. 2016, 14, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Felicetti, F.; Errico, M.C.; Segnalini, P.; Mattia, G.; Care, A. Microrna-221 and -222 pathway controls melanoma progression. Expert Rev. Anticancer 2008, 8, 1759–1765. [Google Scholar] [CrossRef] [PubMed]
  105. Godshalk, S.E.; Paranjape, T.; Nallur, S.; Speed, W.; Chan, E.; Molinaro, A.M.; Bacchiocchi, A.; Hoyt, K.; Tworkoski, K.; Stern, D.F.; et al. A variant in a microrna complementary site in the 3′ utr of the kit oncogene increases risk of acral melanoma. Oncogene 2011, 30, 1542–1550. [Google Scholar] [CrossRef] [Green Version]
  106. Milevskiy, M.J.G.; Gujral, U.; Marques, C.D.; Stone, A.; Northwood, K.; Burke, L.J.; Gee, J.M.W.; Nephew, K.; Clark, S.; Brown, M.A. Microrna-196a is regulated by er and is a prognostic biomarker in er plus breast cancer. Br. J. Cancer 2019, 120, 621–632. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The main estrogen receptors signaling pathways: in the genomic pathway, classic estrogen receptors (ER α and β) act as transcription factors upon homo- or heterodimers. ER α promotes DNA transcription, while ERβ inhibits it. In the non-genomic pathway ERs act as membrane receptors and they lead to increased activity of the RAS/BRAF/MEK axis. Figure 1 demonstrates cross-talk with EGFR receptor pathway. The G protein-coupled estrogen receptor (GPER) acts via intracellular cAMP-protein kinase (PK) and cAMP-response element-binding protein (CREB) phosphorylation.
Figure 1. The main estrogen receptors signaling pathways: in the genomic pathway, classic estrogen receptors (ER α and β) act as transcription factors upon homo- or heterodimers. ER α promotes DNA transcription, while ERβ inhibits it. In the non-genomic pathway ERs act as membrane receptors and they lead to increased activity of the RAS/BRAF/MEK axis. Figure 1 demonstrates cross-talk with EGFR receptor pathway. The G protein-coupled estrogen receptor (GPER) acts via intracellular cAMP-protein kinase (PK) and cAMP-response element-binding protein (CREB) phosphorylation.
Cells 08 01463 g001

Share and Cite

MDPI and ACS Style

Dika, E.; Patrizi, A.; Lambertini, M.; Manuelpillai, N.; Fiorentino, M.; Altimari, A.; Ferracin, M.; Lauriola, M.; Fabbri, E.; Campione, E.; et al. Estrogen Receptors and Melanoma: A Review. Cells 2019, 8, 1463. https://doi.org/10.3390/cells8111463

AMA Style

Dika E, Patrizi A, Lambertini M, Manuelpillai N, Fiorentino M, Altimari A, Ferracin M, Lauriola M, Fabbri E, Campione E, et al. Estrogen Receptors and Melanoma: A Review. Cells. 2019; 8(11):1463. https://doi.org/10.3390/cells8111463

Chicago/Turabian Style

Dika, Emi, Annalisa Patrizi, Martina Lambertini, Nicholas Manuelpillai, Michelangelo Fiorentino, Annalisa Altimari, Manuela Ferracin, Mattia Lauriola, Enrica Fabbri, Elena Campione, and et al. 2019. "Estrogen Receptors and Melanoma: A Review" Cells 8, no. 11: 1463. https://doi.org/10.3390/cells8111463

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