Next Article in Journal
Mitochondrial Genome-Encoded Long Noncoding RNA Cytochrome B (LncCytB) and Mitochondrial Ribonucleases in Diabetic Retinopathy
Previous Article in Journal
Mechanistic Insights and Therapeutic Strategies in Osteoporosis: A Comprehensive Review
Previous Article in Special Issue
Role of Mangiferin in Management of Cancers through Modulation of Signal Transduction Pathways
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Pros and Cons of Estrogens in Prostate Cancer: An Update with a Focus on Phytoestrogens

CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(8), 1636; https://doi.org/10.3390/biomedicines12081636
Submission received: 26 June 2024 / Revised: 14 July 2024 / Accepted: 20 July 2024 / Published: 23 July 2024
(This article belongs to the Special Issue Bioactive Natural Products for Novel Drugs)

Abstract

:
The role of estrogens in prostate cancer (PCa) is shrouded in mystery, with its actions going from angelic to devilish. The findings by Huggins and Hodges establishing PCa as a hormone-sensitive cancer have provided the basis for using estrogens in therapy. However, despite the clinical efficacy in suppressing tumor growth and the panoply of experimental evidence describing its anticarcinogenic effects, estrogens were abolished from PCa treatment because of the adverse secondary effects. Notwithstanding, research work over the years has continued investigating the effects of estrogens, reporting their pros and cons in prostate carcinogenesis. In contrast with the beneficial therapeutic effects, many reports have implicated estrogens in the disruption of prostate cell fate and tissue homeostasis. On the other hand, epidemiological data demonstrating the lower incidence of PCa in Eastern countries associated with a higher consumption of phytoestrogens support the beneficial role of estrogens in counteracting cancer development. Many studies have investigated the effects of phytoestrogens and the underlying mechanisms of action, which may contribute to developing safe estrogen-based anti-PCa therapies. This review compiles the existing data on the anti- and protumorigenic actions of estrogens and summarizes the anticancer effects of several phytoestrogens, highlighting their promising features in PCa treatment.

1. Introduction

Estrogens are steroid hormones mainly associated with female reproductive physiology. Nevertheless, this class of sex hormones is involved in a panoply of biological processes in both men and women [1,2,3]. Regardless of sex, estrone (E1), 17b-estradiol (E2), and estriol (E3) are the primary naturally occurring estrogens, with E2 being the most potent and prevalent [4,5]. The pioneering work of Huggins and Hodges [6] in the middle of the 20th century has provided the basis for the use of hormones in prostate cancer (PCa) therapy by establishing PCa as a hormone-sensitive cancer, stimulated by androgenic activity and inhibited by the suppression of androgen levels or estrogen administration. In fact, estrogens were used in PCa treatment between the 1940s and 1970s, with the synthetic estrogen diethylstilbestrol (DES) being an effective therapy for delaying the progression of metastatic PCa [7,8,9,10,11,12]. However, after decades of practice, estrogen therapy was discontinued due to the associated adverse effects, namely at the cardiovascular level [8,9,10].
The withdrawal of estrogens from clinical practice did not eliminate the interest in these hormones, and research efforts continued deepening the understanding of estrogenic effects in the prostate. The pros and cons of the actions of estrogens in the regulation of prostate cell fate and targeting the different cancer hallmarks have been reported over the years. In parallel to the beneficial effects in suppressing tumor growth, accumulating evidence has pointed to estrogens as causative agents of PCa, promoting cell survival and malignant transformation, leading to tumor growth and metastization [13,14,15].
An extensive amount of research has been carried out to understand the duality of estrogen’s actions in PCa. Although the detailed mechanisms are far from being completely clarified, the double-edged sword of estrogen effects has been highly linked to the separate and distinct activity of estrogen receptors (ERs), which has been elegantly reviewed in the past years [16,17,18]. At a glance, estrogen actions are mediated by the classical nuclear ERs, namely ERα and ERβ, and the membrane-bound G protein-coupled estrogen receptor (GPER) [19,20,21]. ERα and ERβ act as ligand-activated transcription factors exerting direct effects on the regulation of gene expression by binding to specific DNA sequences in the target genes (genomic signaling) [22,23], or alternatively, by interacting with other transcription factors or activating intracellular signaling cascades, indirectly affecting gene expression [24,25]. GPER is involved in rapid nongenomic estrogenic responses through the activation of signal-transduction mechanisms with the subsequent production of intracellular second messengers, cAMP regulation, and protein-kinase activation in signaling cascades, which can ultimately also lead to indirect gene expression regulation [19,20,21,26].
It has been widely accepted that ERα and ERβ drive differential responses in PCa [27,28], with ERα associated with protumorigenic effects, whereas ERβ has been implicated in the antitumorigenic actions of estrogens [29,30,31]. The complexity of estrogen actions in PCa increases considering GPER, as this membrane receptor has been shown to trigger both tumor growth and tumor regression effects [18]. Overall, despite disclosing the mechanism of action and the panoply of anticancer actions, research efforts have not found safe approaches for re-introducing estrogens in PCa therapy [32].
In line with the beneficial effects of estrogens in PCa treatment are the epidemiological studies describing the lower incidence of PCa in Eastern countries associated with a plant-based diet and a higher consumption of phytoestrogens [33,34,35], which have raised curiosity about the therapeutic potential of these compounds. At present, important lines of research have explored the use of phytoestrogens and bioactive compounds with estrogen-like activity in PCa treatment [36]. Several compounds, alone or in combined approaches, have shown promising results in controlling PCa development and progression without adverse effects [37,38,39,40,41,42,43], and have been tested in preclinical studies and clinical trials as possible estrogen therapies [44,45,46,47,48,49]. The present review revisits the effects of natural and synthetic estrogens as anticancer molecules and, opposingly, their actions in fueling PCa hallmarks such as cell survival, proliferative activity, resistance to apoptotic cell death, and invasiveness. Additionally, the actions of diverse natural compounds with estrogenic activity and promising anticancer effects are summarized. Our main goal was to compile the current information, fulfilling the scientific gaps existing in reviewing the subject. The reviews published in the last three years have only focused on one type of ER, a specific class of compounds, or stage of PCa, or more generally, have covered several types of cancer simultaneously, without exhaustively detailing the effects of estrogens in PCa. Herein, an integrated and translational perspective regarding estrogen actions in PCa is provided, and the possible contribution of phytoestrogens to therapy is discussed.

2. Estrogens in the Male

2.1. Circulating Levels

The measurement methodology is the first issue to consider in any analysis of the systematized steroid circulating levels. Among the different methods that can be used to quantify steroid levels in human blood samples, estrogens have essentially been measured by radioimmunoassay (RIA), chemiluminescence immunoassays, and liquid chromatography tandem mass spectrometry (LC-MS/MS), which have the advantage of detecting small amounts of steroids, thus reaching lower limits of quantification. Therefore, several authors have indicated LC-MS/MS as the preferred methodology for quantifying estrogen serum levels [50,51], though RIA remains the most commonly used method (Table 1).
Another important aspect in comparing studies is the units of measurement of the serum steroid concentrations, which also depends on the methodology used. The reported E2 measurements were converted to molar concentrations (nM) to allow for immediate comparisons (Table 1).
Data collected from the literature and summarized in Table 1 show that a broad range of serum E2 concentrations can be found in healthy men, from 0.028 to 0.235 nM. Overall, very low levels were obtained from healthy individuals between 30 and 90 years old. This broad age range encompasses the critical age for the development of PCa, which is 50 years old and above [52]. Moreover, the widespread values reported for E2 serum concentrations could be linked to the age group and the ethnicity or physiological conditions. Indeed, several endocrine and metabolic events, namely the increase in adiposity and body mass index, with the augmented extragonadal aromatization of testosterone, could influence the E2 levels [53,54,55]. Furthermore, circulating estrogens appeared to be inversely associated with intraprostatic inflammation [56]. However, these factors were not always considered in the available studies, mainly when comparing healthy men and PCa patients.
The relationship between PCa and E2 serum levels has been largely controversial, and the majority of studies have not found statistically significant associations between hormone concentrations and the risk of PCa [57,58,59]. Thee E2 serum levels found in PCa patients were also highly variable, ranging from 0.050 to 0.300 nM (Table 1). Although not consensual, some studies have reported a slightly significant increase in E2 levels in PCa cases (Table 1) [55,60,61,62].
Table 1. E2 serum levels in healthy men and PCa patients.
Table 1. E2 serum levels in healthy men and PCa patients.
E2 Concentration Range (nM)Methodology of MeasurementRef.
Healthy MenPCa Patients
0.1560.200 *RIA[61]
0.110–0.1600.120–0.160RIA[63]
0.114–0.1250.110–0.128 *Dextran-coated charcoal method[55]
0.110–0.1600.120–0.160RIA[63]
0.200-RIA[64]
0.093-Chemiluminescence immunoassay[65]
0.200-RIA[66]
0.066–0.2210.066–0.233-[67]
0.1250.121RIA[68]
0.070-Chemiluminescence immunoassay[69]
0.100–0.1500.050–0.300RIA[70]
0.2350.247-[71]
0.089–0.1200.086–0.106Gas chromatography-mass spectrometry[54]
0.0620.066RIA[72]
0.028–0.167-Chemiluminescence immunoassay[73]
0.1060.106RIA[74]
0.1220.122Heterogeneous competitive magnetic separation assay[75]
0.028–0.1560.063–0.068Chemiluminescence immunoassay[76]
0.1070.101–0.105Chemiluminescence immunoassay[77]
0.082–0.2340.200-[50]
0.1080.159 *Enzyme linked immunosorbent assay (ELISA)[62]
0.132-RIA[78]
0.103-RIA[79]
* statistically significant difference.

2.2. Intraprostatic Production

Estrogen biosynthesis is the last reaction on the steroidogenic pathway, and it occurs due to the activity of the heme protein aromatase/P450aro/CYP19A1 enzyme [80,81]. Aromatase in the endoplasmic reticulum [82] or mitochondria [83] catalyzes the irreversible aromatization of the A ring of the androgen precursors, generating estrogens. The naturally occurring estrogens E1, E2, and E3 (Figure 1) are produced via the aromatization of androstenedione and testosterone [5]. E1 can also be reversibly converted to E2 by the enzyme 17β-hydroxysteroid dehydrogenase, and both E1 and E2 are precursors of E3 through the involvement of CYP3A4/5 [84].
The aromatization of androgens in the male occurs mainly in the somatic cells of the testis and other peripheral organs including the prostate [85,86,87]. Prostate cells have been shown to express a functional aromatase that uses external androgen precursors such as dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate (DHEA-S), and circulating testosterone to produce estrogenic compounds (Figure 1) [80,88].
Estrogen synthesis in the human prostate was first pointed out by Matzin and Soloway [86] based on the immunohistochemical localization of aromatase in prostatic tissues. This was followed by the characterization of aromatase expression in both the epithelial and stromal cells of the human prostate [87], and later on by the demonstration that prostatic stromal cells can synthesize E2 from testosterone [89]. Over the years, the local production of estrogens in the prostate and its functional relevance have been studied. Manipulating the aromatase expression levels or activity has been shown to directly affect intraprostatic E2 levels. Moreover, the localization of aromatase in the prostate cells is “conveniently” adjacent to ERs, the mediators of the actions of estrogens in regulating prostate physiology [90,91]. Upregulation of aromatase activity increased the intracellular levels of estrogens in prostate cells concomitantly with ERα and GPER activation [85,92]. In contrast, upon aromatase inhibition, intraprostatic E2 levels were significantly reduced [93].
Altered intraprostatic estrogen biosynthesis has been reported in the neoplastic prostate. Results from the STERKPROSER trial suggest that human cancerous prostates have a higher activity of aromatase, producing more E2 compared with the noncancerous prostate tissues [60]. Moreover, an approximately 30-fold increase in aromatase expression was found in PCa metastatic tissues compared to primary tumors [94].

3. Estrogens in Prostate Cancer Therapy

3.1. Old-Times and Withdrawal

The use of estrogens in PCa therapy goes back to the 1940s. The pioneering work of Huggins and Hodges, which established PCa as a hormone-sensitive cancer, confirming its inhibited growth by the suppression of androgens levels or estrogen administration [6,7], has provided the foundations for the use of hormone therapy.
Estrogens were used in PCa treatment for several years, with the synthetic estrogen DES being a low-cost effective therapy for delaying the progression of metastatic PCa [7,8,9,10,11,12]. In the 1960s and 1970s, the Veterans Administration Cooperative Urological Research Group performed various randomized trials to evaluate the effectiveness of estrogenic therapies for PCa treatment, alone or in combination with orchiectomy [95]. This and other studies observed that estrogen therapy was able to delay the progression of PCa, accomplishing clinical responses in up to 80% of patients [8,9,10]. However, clinical trials also highlighted the adverse effects of hormone therapy, namely its hazardous effects at the cardiovascular level, with associated lethality [8]. A notorious increased risk of cardiovascular toxicity was found in up to 35% of patients receiving estrogen therapy, and thromboembolism was experienced by 15% [3,4,5].
The routes of estrogen administration are diverse (Table 2) and have evolved to accompany the monitoring of estrogens’ beneficial vs. adverse actions and minimize the extent of the harmful effects of hormones. Oral treatment is simple and convenient, and intestinal absorption is rapid [96]. However, in the intestine wall, about 70% of estradiol is metabolized into estrone, which only has one-third of the biological activity of estradiol [96]. In addition, the estrogens that reach the target organs are those that are not retained or excreted by the liver, and the concentration at these organs is about five times less than in the liver [97]. This required the use of doses that have been shown to produce toxicity, with relevant side effects such as hypertension, venous thromboembolism, pulmonary embolism, cerebrovascular accident, ischemic attack, and hypercoagulability, among others [98,99,100]. The parenteral route (intramuscular injection or subcutaneous injection) emerged to circumvent and alleviate the thromboembolic cascade of events and cardiovascular complications associated with oral administration by reducing the liver-associated toxicity and levels of several coagulation factors [101,102,103]. Nevertheless, parenteral treatment was linked to enhanced gynecomastia, intermittent claudication, and retinal thrombosis [104,105]. Several reports have demonstrated that transdermal patches (Table 2) have a similar efficacy compared with other administration routes with fewer side effects [106,107,108,109], which is interesting as they can be easily self-administered and readily withdrawn if toxicity occurs. Table 2 summarizes the information considering the administration routes and doses for different estrogenic formulations.
Despite the effectiveness in suppressing PCa growth and after decades of practice and attempts to optimize treatment approaches, estrogen therapy was withdrawn due to the associated adverse side effects.
Although the “estrogenic approach” was abandoned, research in the area has evolved over the years, disclosing the effects of estrogens and their mode of action [110].
Table 2. Estrogens in PCa treatment.
Table 2. Estrogens in PCa treatment.
Route of AdministrationCompoundDosageRef.
OralEstradiol1–2 mg 3×/day[111]
Conjugated estrogens1.25–2.5 mg 3×/day[112,113]
Ethinylestradiol0.15–3 mg/day[112,113]
Ethinylestradiol sulfonate1–2 mg 1×/week[114,115]
DES1–3 mg/day[116]
Dienestrol5 mg/day[117]
Hexestrol5 mg/day[117]
Fosfestrol100–480 mg
1–3×/day
[118,119]
Chlorotrianisene12–48 mg/day[120]
Quadrosilan900 mg/day[117]
Estramustine phosphate140–1400 mg/day[121]
Transdermal patchEstradiol2–6× 100 μg/day
Scrotal: 1× 100 μg/day
[122,123]
Intramuscular or subcutaneous injectionEstradiol benzoate1.66 mg 3×/week
Estradiol dipropionate5 mg 1×/week
Estradiol valerate10–40 mg 1×/1–2 weeks[89,124]
Estradiol undecylate100 mg 1×/4 weeks[125]
Polyestradiol phosphateAlone: 160–320 mg 1×/4 weeks
With oral EE: 40–80 mg 1×/4 weeks
[126,127]
Estrone2–4 mg 2–3×/week[91]
Intravenous injectionFosfestrol300–1200 mg 1–7×/week[118,119]
Estramustine phosphate240–450 mg/day[128]

3.2. Synthetic Estrogens

The effects of estrogens were, firstly, believed to be mostly mediated through the blockade of the hypothalamic–pituitary–gonadal axis, inhibiting gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) release through negative feedback loops [129], resulting in decreased testosterone production and tumor regression [2,130]. This fact was supported by several reports in the 1990s that were not able to identify any detectable ER levels in the epithelial compartments of human prostatic tissue [131]. However, the paradigm changed with the characterization of ERs and the demonstration that estrogens have therapeutic effects through direct action in PCa cancer cells [132,133], independently of the systemic hypothalamic–pituitary–gonadal axis.
The human ERα cDNA was first cloned in 1985 [134,135], a discovery followed more than ten years later by the identification of a second ER gene, the ERβ [21]. ERβ is the most prevalent ER subtype expressed in human prostate tissue [136] and is mainly associated with the differentiation compartment that includes the luminal cells [137]. ERα expression in the non-neoplastic prostate is restricted to stromal cells and to the androgen-independent basal cell layer that comprises prostate stem cells and the proliferation compartment of the prostate epithelium [131,138,139].
The different expression pattern of the ER subtypes between luminal cells and basal cells modulates their susceptibility to cytotoxic agents [140]. Luminal cells expressing high levels of ERβ are particularly vulnerable and activate the programmed cell death after androgen deprivation therapy, radiation, or chemotherapy. In contrast, basal cells presenting high ERα expression levels are multi-drug resistant and survive to cytotoxic conditions [140]. Accordingly, the expression of ERβ in PCa was associated with a better relapse rate than ERβ-negative tumors [141], which renders this ER subtype a putative therapeutic target [142]. Regarding ERα, its expression was demonstrated to be upregulated during malignant progression of the prostatic epithelium and is highly expressed in metastatic PCa and CRPC [143]. Moreover, it has been shown that ERα-knockout mice showed no development of high-grade prostatic intraepithelial lesions or PCa upon chronic treatment with testosterone and estradiol [144]. Therefore, ERα inhibitors might have a potential antitumor activity in PCa. Overall, nuclear ER signaling seems to play a dual role in PCa development, progression, and therapy. GPER-mediated effects should also be taken in account, as this membrane receptor has been shown to trigger both tumor growth and tumor regression effects [18]. Moreover, it is crucial to fully characterize the ERs and GPER downstream signaling pathways and the effectors involved in the modulation of PCa progression [145].
Considering the pharmacological interest and the ERs and GPER druggability, a large variety of synthetic estrogenic compounds with different structures and pharmacologic and metabolic characteristics have been investigated (Figure 2).
DES is a synthetic nonsteroid estrogen that, in contrast to the natural estrogen E2, is not markedly bound to sex hormone-binding globulin [146]. As described, it was the gold standard in endocrine therapy, though with significant side effects [147]. Ethinylestradiol is a synthetic and potent steroid estrogen since it suffers an alkylation in the 17α position, which does not allows it to be a substrate for 17β-hydroxysteroid-dehydrogenase, the enzyme that reversibly converts E2 to the less potent E1 (Figure 1) in target tissues [148,149].
In the attempt to discover better estrogen formulations to use in the treatment of PCa, novel compounds have emerged that include conjugated estrogens, polyestradiol phosphate, ethinylestradiol, the synthetic benzoate ester of estradiol (β-estradiol 3-benzoate), 17α-ethynyl-5α-androstane-3α, 17β-diol (HE3235), 8β-vinylestra-1,3,5(10)-triene-3β,17β-diol (8β-VE2), 17α-20Ζ-21-[(4-amino)phenyl]-19-norpregna-1,3,5(10),20-tetraene-3,17β-diolthe (APVE2), and the nongenotoxic estrogen 2-fluoroestradiol (2F-E2), among others (Figure 2). The detailed effects of these compounds in controlling prostate cell fate and tumor growth are presented in the subsequent sections of the review.

4. Estrogens as Prostate Carcinogens

4.1. Cell Survival and Neoplastic Transformation

Estrogens have been implicated as a cause of BPH, a condition predominantly characterized by the overgrowth of the stromal compartment. For example, E2 was shown to increase the proliferation of human BPH-derived stromal cells in an ER-dependent mechanism but had no effect on epithelial cells [150]. However, E2 (1 µM) was shown to increase the proliferation of immortalized nontumorigenic human prostate epithelial BPH-1 cells in culture for 3 days [151], although cell proliferation started being reduced after 6 days of hormone exposure [151]. Nevertheless, DNA damage was significantly higher in BPH-1 cells treated with E2 for 6 weeks, accompanied by an increased percentage of cells immunopositive for the oncogene c-Myc- and the cell cycle inducer cyclin D1 [151]. In recombinant prostate tissue, composed of urogenital mesenchyme plus BPH-1 cells grown under the kidney capsule of male athymic nude mice, testosterone and E2 treatment increased the Ki-67 proliferation index compared with the untreated animals [152].
Different types of studies, with diverse experimental approaches and using distinct estrogenic compounds, have demonstrated that estrogens can deregulate prostate cell survival and growth (Table 3), which have been linked to hyperplasia, dysplasia, metaplasia, and neoplasia.
The effects of estrogens inducing the proliferation of human prostate cells have been reported in different cell types, from non-neoplastic to androgen-sensitive and CRPC cells. E2 concentrations from 0.01 to 5 µM (Table 3) increased the proliferation of human prostate epithelial cells and androgen-sensitive LNCaP and VCaP cells [153,154,155]. In CRPC cells, namely, the DU145 and PC3 cell lines, E2 (0.01 and 1 µM) increased the anchorage-independent growth and inhibited apoptosis [14]. E2 also increased the growth and anchorage-independent growth of cells isolated from the lymph node metastasis of a PCa patient in an ERα-dependent mechanism [15].
The molecular orchestrators responsible for the proliferative actions of estrogens have also been disclosed. Besides the dependency of ERα alone, the interaction between ERα, androgen receptor (AR), and Src has been shown to be required for the rapid activation of the mitogen-activated protein kinase (MAPK) proliferation pathway in non-neoplastic cells [153]. Moreover, the augmented proliferation of LNCaP cells was associated with the increased expression of PSA, insulin-like growth factor 1 receptor (IGF-1R), IGF-1, insulin-like growth factor binding protein-3/4 (IGFBP-2), and the decreased expression of AR, ERβ, IGF-II, and IGFBP-3 [154].
The increased proliferation of prostate cell lines in response to estrogens has been translated to in vivo experiments. E2 administration (Table 3) was shown to decrease the population of rat prostate epithelial cells in phase G0/G1 while increasing the S and G2/M phase cell populations [13]. In addition, E2 caused DNA damage and induced apoptosis of prostate tissue cells, at least for short-term exposure [13].
For example, in human prostate xenografts, early exposure to estradiol benzoate (7 to 90 days) combined with secondary exposure (until 200 days) increased the proliferation of prostate epithelial cells, leading to the development of hyperplastic glands [156]. This was underpinned by the suppression of PTEN expression and likely augmented the activity of the PI3K-Akt signaling pathway, resulting in the inhibition of apoptosis and cell cycle progression [156]. Along the same line, DES has been reported as causative of prostate hyperplasia, since prenatal exposure to this compound resulted in enlarged prostates in adults [157]. Beyond the induction of cell proliferation, the effects of estrogens have been related to the development of other histological alterations in the prostate. Several studies have reported that E2, in combination with testosterone, induced dysplasia in rat dorsolateral prostate [158,159,160] as well as in the lateral, ventral, dorsal, and anterior regions of mice prostate [144].
Also, several estrogenic compounds seem to cause squamous metaplasia in the prostate of several species [161,162,163]. DES induced the metaplastic transformation of mouse prostate epithelium, mainly in the anterior region [164]. This effect was associated with the onset of cytokeratin-10 expression, the upregulation of progesterone receptor expression, and the loss of expression of the cell cycle inhibitor p27 Kip1 [164].
Yu and colleagues observed that E2 induced the neoplastic transformation of rat prostate epithelial cells [13]. These transformed cells displayed an increased expression of several putative PCa stem cell markers as well as changes in the expression of hormone receptors, namely, increased levels of ERα and the decreased expression of ERβ and AR [13,151], which indicates a change in the hormone-responsiveness accompanying tumor development. Moreover, these findings support the driven role of ERα in prostate carcinogenesis. Low doses of E2 also increased tumor growth in DU145 and PC3 cell xenograft models [14]. However, the carcinogenic effects of estrogens have mainly been demonstrated by their association with testosterone (Table 3). Rat treatment with E2, in addition to testosterone, promoted tumor development, causing the formation of DNA adducts, oxidative DNA damage, and lipid peroxidation [160]. In mice, the pathologic areas induced by the testosterone plus E2 treatment showed an increased number of PCNA-positive proliferating cells [144]. Moreover, the addition of E2 was shown to shift the incidence of prostate tumors to 100% compared only to an incidence of 35–40% when testosterone was given alone [159]. These findings implicate estrogens in prostate carcinogenesis and indicate that the known effects of androgens driving cancer may depend on testosterone aromatization to E2, which has been demonstrated in animal models. Aromatase knockout mice, unable to synthesize E2, displayed a reduced incidence of PCa compared with wild-type animals in response to testosterone administration [144]. This study also showed that the presence of a functional ERα is decisive for tumor development. E2 plus testosterone treatment was ineffective, inducing PCa in ERα knockout mice, whereas ERβ knockout animals displayed a biochemical and histological pattern of carcinogenesis similar to their wild-type counterparts [144].
Humans are exposed to estrogens since embryological life, and prenatal exposure to estrogens has been shown to have observable effects in the adult male reproductive tract including the prostate gland [165]. Studies performed in animal models have shown that exposure to low doses of estrogens during fetal development causes the enlargement of the prostate [157]. In contrast, high doses of estrogens implicate a reduction in the adult prostate weight [157]. Moreover, developmental exposure to estradiol benzoate seems to change the differentiation and epigenetic programming in a human fetal prostate xenograft model [156], which can be a mechanism contributing to carcinogenesis later in life. Research efforts are needed to ascertain the impact of fetal estrogen exposure and the development of PCa in adulthood.
Catechol estrogens are potent compounds that originate from the metabolism of estrogens. This class of compounds has been identified in the prostate tissue and has also been linked to prostate malignancy [166]. Polymorphisms in the estrogen metabolism enzymes that detoxify catechol estrogens, namely, catechol-O-methyltransferase (COMT), glutathione (GSH), and quinone reductase are associated with PCa risk [167,168,169,170,171]. Furthermore, prostate areas susceptible to carcinoma development have been shown to have less protection through the activity of these enzymes. This susceptibility and the protumorigenic effects were related to the reaction of catechol estrogen-3,4-quinones (E2-3,4-Q) with DNA, which is favored by the reduced activity of COMT, GSH, and quinone reductase [166,172]. In vitro studies showed that exposure to the catechol estrogens 2-hydroxyestradiol (2-OHE2) and 4-hydroxyestradiol (4-OHE2) increased the proliferation of BPH-1 cells [151]. This was achieved by slightly diminishing the nonproliferating cell fraction (G0/G1-phase) while increasing the cell population in S-phase [151]. Moreover, both catechol estrogens increased the expression of cyclin D1 and c-Myc, which explains the shift toward proliferative behavior [151,173]. Increased proliferative ability driven by 4-OHE2 and 2-OHE2 was accompanied by the altered expression of molecular targets in estrogen signaling pathways. These catechol estrogens increased the abundance of ERα and its downstream target IGF-1R, whereas they reduced the levels of ERβ and its downstream tumor suppressor FOXO-1. However, 4-OHE2 effects were observed to a greater extent than that of 2-OHE2 [151]. Furthermore, genotoxic effects linked with the neoplastic transformation of BPH-1 cells were observed in response to the administration of 4-OHE2 and 2-OHE2 [151].
Catechol estrogens have also been shown to disturb tissue homeostasis and prostate histology architecture. The catechol estrogen 4-OH-E2, in combination with testosterone, induced prostatic dysplasia, and the frequency of appearance of this histological phenotype was doubled by the addition of 2F-E2 to testosterone and 4-OH-E2 [160].
In sum, several reports have defended that exposure to exogenous estrogens can be tumorigenic, mainly when low doses are used. The majority of in vitro studies have reported that E2, at least for concentrations below 1 µM (Table 3), increased the proliferation of prostate cells. Also noteworthy is the fact that the altered intraprostatic estrogen biosynthesis is associated with neoplastic transformation of the prostate by disrupting downstream signaling pathways and influencing the development and proliferation of prostate epithelial cells and stroma [174].
Table 3. Effect of estrogens in regulating the survival and growth of prostate cells in vitro and in vivo.
Table 3. Effect of estrogens in regulating the survival and growth of prostate cells in vitro and in vivo.
Type of StudyCell Line/Animal ModelCompoundConcentration/DoseAssay Model/Method of AdministrationTime of TreatmentAssayEffectActivated PathwayRef.
In vitroBPH-1E21 µM6-well platesUp to 6 days/6 weeksSulforhodamine-B
Comet
↑ Proliferation
Neoplastic transformation
Genotoxic mechanism[151]
BPH-12-OHE21 µM6-well platesUp to 9 days/6 weeksSulforhodamine-B
Comet
↑ Proliferation
Neoplastic transformation
Genotoxic mechanism[151]
BPH-12-OHE210 µM6-well plates2, 4, 7 and 10 daysSulforhodamine-B↓ Cell viabilityCytotoxicity[151]
BPH-14-OHE21 µM6-well platesUp to 9 days/6 weeksSulforhodamine-B
Comet
↑ Proliferation
Neoplastic transformation
Genotoxic mechanism[151]
LNCaPE20.01 µMMulti-well plates24 hBrdU incorporation↑ ProliferationMAPK (ERK1/2) and association of AR, ERβ, and Src[175]
LNCaPE20.01–5 µM96-well plates3 daysMTS
PSA measurement
↑ PSA expression
↑ Cell growth
ER mediated[176]
LNCaPαE20.01–5 µM96-well plates3 daysMTS
PSA measurement
↑ PSA expression
↑ Cell growth
ER mediated[176]
LNCaPE20.01 µM 24 h/48 hWST-1 based↑ Proliferation
↑ PSA expression
↑ c-Myc[177]
DU145E20.01 µM12-well plates24 hSoft-agar colony formation
TUNEL
↑ Anchorage-independent growth
↓ Apoptosis
↓ FOXO1[14]
DU145E21 µM12-well plates24 hSoft-agar colony formation
TUNEL
↑ Anchorage-independent growth
↓ Apoptosis
↓ FOXO1[14]
PC3E20.01 µM12-well plates24 hSoft-agar colony formation
TUNEL
↑ Anchorage-independent growth
↓ Apoptosis
↓ FOXO1[14]
PC3E21 µM12-well plates24 hSoft-agar colony formation
TUNEL
↑ Anchorage-independent growth
↓ Apoptosis
↓ FOXO1[14]
EPNE20.01 µM60 mm
dishes
27 h/5 minBrdU incorporation↑ ProliferationMAPK (ERK1/2) and association of AR, ERα, and Src[153]
NRP-152E21 or 3 µM96-well plates/
60 mm dishes
2–6 weeks
and 3–48 h
Soft agar colony formation
Comet
Flow cytometry
Neoplastic transformationGenotoxic mechanism[13]
MDA-Pca 2bβE20.01–5 µM96-well plates3 daysMTS
PSA measurement
↑ Cell growthER mediated[176]
In vivoBALB/c miceDES *2 mg DES
(and 18 mg cholesterol)
Pellet s.c. implantUp to 3 weeksPCNA-immunohistochemistry↑ ProliferationERα[164]
Nude miceE21 or 3 µME2-NRP-152 cells s.c. injection in the flanks4–8 weeksTumor size measurement↑ Neoplastic transformationGenotoxic mechanism[13]
Athymic nude miceβ-estradiol 3-benzoate *250 μg/kg
(early exposure)
+
2.5 mg pellet (secondary exposure)
S.c injection90 days
(early exposure)
+
110 days
(secondary exposure)
Ki-67 quantification↑ ProliferationPI3K-Akt pathway[156]
Xenograft BALB/
cA-nu castrated mice using DU145 and PC3 cells
E20.18 mgPellet s.c. implant25 to 35 daysTumor volume and weight measurement
TUNEL
↑ Tumor growth↓ FOXO1
ERβ and KLF5 pathway
[14]
Athymic nude mice
Tissue recombinants composed of mouse urogenital mesenchyme plus an immortalized nontumorigenic human prostatic epithelial cell line (BPH-1) grown under the kidney capsule
E22.5 or 10 mg
(plus Testosterone)
Silastic implants1–4 monthsImmunohistochemistry
Growth indices
Determination of cancer incidence
↑ Proliferation
↑ Apoptosis
Akt pathway[152]
NBL/Cr rats4-OHE25 μg/daySilastic implants13 weeksMeasurement of DNA adducts
Measurement of 8-hydroxyguanosine
Measurement of lipid hydroperoxides
↑ Inflammation
↑ Dysplasia
-[160]
NBL/Cr rats2F-E2 *5 μg/daySilastic implants13 weeksMeasurement of DNA adducts
Measurement of 8-hydroxyguanosine
Measurement of lipid hydroperoxides
↑ Inflammation
↑ Dysplasia
-[160]
NBL/Cr ratsE2
(+Testosterone)
S.c. silastic implants91 weeksHematoxylin and eosin staining↑ Prostate adenocarcinoma development-[159]
NBL/Cr ratsDES *
(+Testosterone)
S.c. silastic implants91 weeksHematoxylin and eosin staining↑ Prostate adenocarcinoma development-[159]
Sprague-
Dawley (Hsd:SD) rats
E2
(+Testosterone)
S.c. silastic implants75 weeksHematoxylin and eosin staining↑ Prostate adenocarcinoma development-[159]
Sprague-
Dawley (Hsd:SD) rats
DES *
(+Testosterone)
S.c. silastic implants75 weeksHematoxylin and eosin staining↑ Prostate adenocarcinoma development-[159]
CD-1 miceE2
(+Testosterone)
Silastic implants4 monthsHistopathological grading
Immunohistochemistry
Histological analysis
↑ Prostate size-[144]
C57BL/6 miceE2
(+Testosterone)
Silastic implants4 monthsHistopathological grading
Immunohistochemistry
Histological analysis
↑ Prostate size
Prostatic intraepithelial neoplastic lesions’ induction
↓ α-actin
↓ E-cadherin
[144]
C57BL/
6 X J129 mice
E2
(+Testosterone)
Silastic implants4 monthsHistopathological grading
Immunohistochemistry
Histological analysis
↑ Prostate size-[144]
* synthetic compound.

4.2. Progression of Disease and Metastization

Metastization is the complex process of cancer dissemination to distant organs and is the main cause of cancer-related deaths [178]. The occurrence of metastasis mainly depends on the migration and invasion capabilities of cancer cells, which is a consequence of the epithelial–mesenchymal transition (EMT) [179]. EMT is characterized by a switch in the expression of epithelial to mesenchymal markers, namely, the reduction in E-cadherin expression and the augmentation of N-cadherin and vimentin proteins [180,181], which in the case of prostate cells has been linked with estrogenic actions, and the effect of catechol estrogens.
Estrogens could alter the phenotype of BPH-1 cells, downregulating E-cadherin and upregulating vimentin and Snail expression, thus stimulating EMT [182]. Altered expression of EMT markers and enhanced migration and invasion properties have been described in PCa cells in response to estrogens. E2 was shown to promote the migration and invasion of distinct PCa cell line models and patient-derived cells (Table 4). E2 (0.01 µM) increased the migration of PacMetUT1 (isolated from the lymph node metastasis of a PCa patient) and 22Rv1 cells [15]. Accordingly, treatment with E2 caused a change in the morphology of PacMetUT1 cells similar to the EMT process, which was confirmed by a downregulation of E-cadherin expression and upregulation of vimentin and Snail [15]. Catechol estrogens, namely 2-OHE2 and 4-OHE2, have been shown to increase the invasion of prostatic cells [151], which can disrupt the prostate histology architecture and promote tumor progression.
In vivo approaches support the outcomes of cell-based research, indicating the role of estrogens in PCa progression and metastization (Table 4). Mice receiving E2 in combination with testosterone showed a dramatic reduction in α-actin and E-cadherin expression accompanying the prostate malignant transformation [144]. Similar findings were found when applying this treatment in mice harboring recombinant prostate tissue composed of urogenital mesenchyme and immortalized BPH-1 cells [152]. Metastatic carcinoma cells were identified in renal lymph nodes, lungs, and liver, with a drastic reduction in E-cadherin expression [152].
ERα has been indicated as the “guilty” ER in mediating the pro-migration and -invasion effects of estrogens. Indeed, the enhanced migratory capabilities observed in the PCa cell lines and patient’s metastasis-derived cells were reported to depend on ERα signaling [15]. However, ERα might promote the metastatic process by favoring the “seed” of PCa cells in other organs. The “seed and soil” hypothesis proposed by Stephen Paget to explain the emergence of metastasis [183] compares the dissemination of tumor cells to the distribution of seeds, arguing that only the seeds that find the proper soil will survive. The rationale underlying this model supports that cells from a primary tumor will establish metastasis if they find a well-matched organ. PCa preferentially metastasizes to bone [184], and interestingly, it has been shown that estrogen signaling through ERα stimulates the osteoblast-like properties of PCa cells, demonstrating its important role in the formation of osteoblastic lesions [15]. This suggests that estrogen signaling can be a relevant mechanism driving the progression of the disease for more aggressive stages and the emergence of bone metastasis. Therefore, characterizing the expression of ERs, mainly ERα, in PCa cases could be a useful prognosis tool in evaluating the progression of disease. Research is needed to deeply clarify this usefulness, though the presence of ERα, or the combination of ERα-positivity with low ERβ expression, is correlated with worse biochemical recurrence, disease progression, and survival outcomes [185].
The stroma component has also been shown to play a triggering role in the metastatic effects of estrogens. One interesting work revealed that conditioned medium from E2-stimulated prostate stromal cells could promote DU145 and PC3 PCa cell migration [186]. This increased migration seems to be mediated by enolase 1 [186], a key enzyme in glycolytic metabolism, also known as pyruvate dehydrogenase phosphatase, which catalyzes the transformation of 2-phosphate-D-glycerate to phosphoric acid-pyruvate as well as the reverse conversion of phosphoric acid-pyruvate to 2-phosphate-D-glycerate for glycogen synthesis [187]. Enolase 1 expression is frequently increased in tumors [188], and due to its role in anaerobic glycolysis, it is thought to promote tumor development and progression [187]. A total of E2 0.01 µM enhanced the stability of enolase 1 in prostate stromal cells and promoted its secretion to the extracellular matrix in an ERα-dependent mechanism [186]. Studies with recombinant enolase 1 showed that this enzyme binds to the membrane of PCa cells, promoting cell migration in a paracrine manner via their plasminogen receptor activity [186]. The secretion of stromal cell-derived enolase 1 and its association with the surface of PCa cells recruits and activates plasminogen, thereby promoting the remodeling of the extracellular matrix and migration [186,189,190].
Another mechanism related to the effect of E2 promoting migration and invasion is the upregulation of the sex-determining region Y-box 4 (SOX4) [155], a developmental transcription factor overexpressed in many types of human tumors [191]. SOX4 was shown to regulate the expression of metastasis-related genes promoting EMT and PCa progression [192].
Furthermore, the augmented intraprostatic production of estrogens seems to promote the progression of CRPC and favor tumor metastasis. Aromatase knockdown and the aromatase inhibitor letrozole were shown to increase the overall survival and inhibit metastasis in castrated PC3 xenograft mice [193], which contributed to imply estrogens in PCa metastization. The estrogenic effects in the PC3 xenograft mice were proposed to occur through the activity of ERα, and its interaction with the estrogen response elements in the MMP12 gene, regulating the expression of matrix metalloproteinase 12 (MMP-12) [193], a family member of a class of proteins (MMPs) involved in the remodeling of extracellular matrix, tumor progression, and metastization [194,195]. MMP-12 knockdown suppressed the invasion of PC3 cells [193] and has been shown to mediate the interactions between PCa cells and bone marrow stromal cells during bone metastasis [196], which further supports the role of ERα as a driver of PCa metastization to the bone.
In summary, in agreement with the pro-proliferative effects, low concentrations/doses of estrogens support the increased migration and invasion of PCa cells, leading to tumor progression and mestastization.
Table 4. Effect of estrogens in promoting prostate cancer cell migration, invasion, tumor progression, and metastization.
Table 4. Effect of estrogens in promoting prostate cancer cell migration, invasion, tumor progression, and metastization.
Type of StudyCell Line/Animal ModelCompoundConcentration/DoseAssay Model/Method of AdministrationTime of TreatmentAssayEffectActivated PathwayRef.
In vitroLNCaPE20.01 µM 24 h/72 hImmunoblotting
Wound healing
Invasion
↑ Migration
↑ Invasion
EMT
↑ N-cadherin
↑ Vimentin
↓ E-cadherin
[177]
LNCaPE20.01 µM12-well plates5 daysAlkaline phosphatase activity↑ Osteoblast-like propertiesERα signaling[15]
LNCaPE20.01 µM96-well plates72 hWound healing
Invasion
↑ Migration
↑ Invasion
SOX4 up-regulation[155]
22Rv1E20.01 µM24-well Boyden
chambers
18 hMigration↑ MigrationERα signaling[15]
RWPE-1E20.01 µM96-well plates48 h/72 h
8 days
RT-PCR analysis
Western blot
EMTERα signaling[182]
C4–2E20.01 µM24-well Boyden
chambers
18 hMigration↑ MigrationERα signaling[15]
PacMetUT1 isolated from the lymph node metastasisE20.01 µM24-well Boyden
chambers
18 hMigration↑ Migration
EMT
ERα signaling[15]
DU145Conditioned medium PrSC from BPH patients or WPMY-1 cells treated with E20.01 µM15-cm dishes48 hTranswell migration
Wound healing
↑ MigrationERα signaling ENO1 effects via its plasminogen binding domain[186]
DU145E20.01 µM 24 hWound healing
Invasion
↑ Migration
↑ Invasion
ERα signaling
ERβ signaling
Galectin-3 signaling
[30]
DU145E20.01 µM 48 hInvasion analysis
Colony formation analysis (soft agar)
↑ InvasionSRC[197]
PC3E20.01 µM Invasion analysis
Colony formation analysis (soft agar)
↑ InvasionSRC[197]
PC3Conditioned medium PrSC from BPH patients or WPMY-1 cells treated with E20.01 µM15-cm dishes48 hTranswell migration
Wound healing
↑ MigrationERα signaling
ENO1 effects via its plasminogen binding domain
[186]
PC3E20.01 µMIncubation of cells 24-well, 8.0-μm pore sizeOvernightMatrigel invasion↑ InvasionERα/matrix metalloproteinase 12 axis activation[193]
PC3E20.0001 and 0.01 µM 24 hWound healing
Invasion
Colony formation analysis (soft agar)
↑ Migration
↑ Invasion
ERα and ERβ signaling
β-catenin signaling
[198]
In vivoCD-1, C57BL/6, and C57BL/
6 x J129 mice
E22.5 or 10 mg
(+Testosterone)
Silastic implants4 monthsHistological analysis
Immunohistochemistry
CarcinogenesisERα signaling[144]
NBL/Cr ratsE2 Silastic implants s.c.16 weeksMeasurement of DNA adducts
Measurement of 8-hydroxyguanosine
Measurement of lipid hydroperoxides
↑ Tumor incidenceDNA adduct
Oxidative DNA damage
Lipid peroxidation
[160]
NBL/Cr rats4-OHE25 μg/daySilastic implants13 weeksMeasurement of DNA adducts
Measurement of 8-hydroxyguanosine
Measurement of lipid hydroperoxides
Carcinogenesis-[160]
NBL/Cr rats2F-E2 *5 μg/daySilastic implants13 weeksMeasurement of DNA adducts
Measurement of 8-hydroxyguanosine
Measurement of lipid hydroperoxides
Carcinogenesis-[160]
Athymic nude mice
Tissue recombinants composed of mouse urogenital mesenchyme plus an immortalized nontumorigenic human prostatic epithelial cell line (BPH-1) grown under the kidney
capsule
E22.5 or 10 mg
(+Testosterone)
Silastic implants1–4 monthsImmunohistochemistry
Growth indices
Determination of cancer incidence
↑ Progression
↑ Metastization
Akt pathway[152]
* synthetic compound.

5. Evidence of Estrogens as Protective Agents

5.1. Antiproliferative and Proapoptotic Effects

5.1.1. Endogenous Estrogens

In vitro and in vivo evidence have shown that E2, the most potent endogenous estrogen, is capable of reducing PCa cell viability and proliferation (Figure 3 and Supplementary Table S1). This effect was reported in PCa cell lines, either in androgen-sensitive, LAPC-4 and LNCaP as well as in 22Rv1 cells and AR-negative CRPC models, namely PC3 cells [147,176,199,200,201]. Treatment with E2 concentrations up to 10 µM was shown to reduce the DHT-induced cell growth of LNCaP and LAPC-4 cells [176,200], whereas lower concentrations, ranging from 1 nM to 1 µM, were able to diminish the proliferation of 22Rv1 and PC3 cell lines [199,201]. The same effect was shared by the E2 stereo-isomer 17α-estradiol (αE2), with 0.1–10 µM concentrations suppressing the proliferation of the androgen-sensitive PCa cell lines [176,200]. Moreover, the E2 effects counteracting the proliferation and viability of PCa cells were followed by an increase in apoptosis. E2 concentrations between 1 nM and 1 µM increased apoptosis in the LAPC-4 cells and also in the CRPC cells, namely, 22Rv1 and PC3 [147,199,201]. In addition, the stimulation of PCa cells with both E2 and αE2 had other interesting effects such as reversion of the DHT- or cortisol-induced PSA expression and a reduction in the DHT-induced DNA biosynthesis [176,200].
The results obtained in vitro were translated into the in vivo setting. Wistar rats treated with a supraphysiological dose of E2 showed a marked reduction in prostate weight underpinned by decreased cell proliferation and increased apoptosis compared to rats receiving a placebo [202]. Several studies in animal tumor models (Supplementary Table S1) have also reported the effects of estrogens counteracting the growth of malignant prostate tissue (Figure 3). In rodents, E2, αE2, or estradiol benzoate treatment reduced the prostate tumor growth (size and volume) by suppressing the proliferation of tumor cells and inducing apoptosis in both androgen-sensitive and CRPC tumor models [14,133,176,200,203,204,205,206]. In addition, survival improvement was described in the mice LuCaP 35 xenograft tumor model treated with E2 [133]. Also noteworthy are other reported effects of E2 in mice including the reduction in testosterone and DHT serum concentrations and PSA levels [133,200], which further corroborates the antitumorigenic effects of this hormone.
Additionally, the antitumor role of the endogenous metabolite of E2, 2-methoxyestradiol (2-ME2), has been widely studied in PCa. Kumar et al. [207] were the first to demonstrate that 2-ME2 inhibits the growth of non-neoplastic prostate epithelial cells. This growth inhibitory effect and the reduction in cell proliferation by 2-ME2 were also observed in a variety of both androgen-sensitive and CRPC cell line models (Figure 3 and Supplementary Table S1) [207,208,209,210,211,212]. Unsurprisingly, 2-ME2 has been highly associated with apoptosis induction in PCa (Supplementary Table S1) [207,208,209,210,211,213,214,215,216,217,218].
In vivo findings in rats and mice further supported the enormous quantity of in vitro evidence of the antitumorigenic role of 2-ME2. Copenhagen X Fisher F1 male rats transplanted with Dunning R3327-PAP prostate tumors treated with 12.5 mg/kg/day of 2-ME2 showed reduced tumor growth accompanied by increased apoptosis [213]. Accordingly, in both androgen-sensitive or CRPC xenograft mice models, 2-ME2 treatment could reduce tumor growth and augment apoptosis (Supplementary Table S1) [208,210,211,218,219,220,221]. Additionally, in the TRAMP mice model, in which tumor development resembles the progression of human PCa (hyperplasia, 8–12 weeks; neoplasia, 15–18 weeks; metastasis, 24 weeks), the effect of 2-ME2 treatment in reducing tumor growth was confirmed. The administration of 2-ME2 had an important role during the various stages of PCa, reducing prostate weight, malignant transformation, and neoplastic progression as well as promoting tumor regression [210,218,221]. Moreover, these effects were accompanied by a reduction in testosterone [221] and PSA levels [210]. However, Ganapathy and colleagues [218] observed a reduction in apoptosis in TRAMP mice treated with 2-ME2, which was suggested to be related to restoring normal tissue architecture.
Some years ago, it was demonstrated that the DHT metabolites, 5α-androstane-3α,17β-diol (3α-diol) and 5α-androstane-3β,17β-diol (3β-Adiol), have estrogenic activities via the activation of ERβ [222,223,224]. Also, these estrogenic metabolites of DHT were shown to have anticancer properties. A total of 0.1 µM (and concentrations below) 3β-Adiol diminished proliferation and increased the apoptosis of both androgen-sensitive and CRPC cells [225,226,227]. Accordingly, the proliferation of established tumors in PC3 xenograft mice tumor models was substantially reduced by a 3-week treatment with 3β-Adiol [226].
ERs and GPER as well as their downstream effectors in signaling pathways are the molecular players involved in the control of prostate cell growth by estrogens [147,176,200]. However, the protective role of estrogens in PCa has mainly been attributed to ERβ, which has been shown to have antiproliferative and proapoptotic effects in PCa models [228,229].
Over the years, the downstream effectors of the ERs’ actions have been disclosed. The stem cell factor (SCF) and its tyrosine kinase receptor c-KIT, with a recognized role in controlling cell proliferation and apoptosis, are important drivers of carcinogenesis in several tissues [230,231,232]. The SCF/c-KIT system was shown to be involved in non-neoplastic prostate cell growth and a target of estrogen regulation. Treatment with supraphysiological doses of E2 strongly diminished the rat prostate weight, suppressed cell proliferation, and induced apoptosis, underpinned by the diminished expression of SCF and c-KIT [202].
In PCa cells and tissues, the actions of estrogens in controlling cell proliferation strictly depend on the control of several cell cycle regulators, namely, cyclins, cyclin-dependent kinases (CDKs), and CDK inhibitors. The antiproliferative effects of estrogenic compounds such as αE2, E2, and 2-ME2 have been related to cell cycle arrest at the S, G1, and G2/M phases. αE2 and E2 seem to reduce cyclin A and cyclin D1 expression [200] and are involved in cyclin D1 phosphorylation through p38 kinase activation [147]. The reduction in these cyclins disrupts cell cycle progression, decelerating PCa proliferation. 2-ME2 was shown to induce G2/M arrest, increasing the G2/M population, with a concomitant decrease in the G1 population [207]. This G2 block can be explained by the inhibition of CDK-1 activity, since 2-ME2 leads to the accumulation of phosphorylated-CDK-1 by inducing the expression of the inhibitory Wee 1 kinase, which mediates the phosphorylation of CDK-1 at the tyrosine 15 residue, making it inactive [207,233,234]. 2-ME2 administration was also shown to increase the expression levels of p53 and CDK inhibitor p21, which further detailed the molecular targets underlying the effect of this estrogenic compound disrupting the cell cycle (Figure 3) [211,235].
Concerning the mechanisms involved in the proapoptotic effects of estrogens, both the extrinsic and intrinsic pathways of apoptosis are activated by E2 and estrogenic metabolites (Figure 3). E2- and 2-ME2-induced PCa cell apoptosis was shown to be mediated by the membrane death receptor Fas and its ligand (Fas L), with the subsequent activation of caspase-8 [202,215,218]. In fact, E2 and its metabolite 2-ME2 increased the expression of Fas and Fas L as well as caspase-8 [202]. Activation of the extrinsic pathway of apoptosis upon Fas/Fas L binding causes the formation of the death inducing signaling complex (DISC) by the recruitment of several molecules including the Fas-associated death domain (FADD) and procaspase-8, which becomes activated [236,237,238]. 2-ME2 was shown to augment DISC formation by inhibiting Akt, sensitizing PCa cells to Fas-mediated apoptosis [215]. The antiapoptotic protein Fas-associated death domain interleukin-1 converting enzyme-like inhibitory protein (FLIP) is a key inhibitor of Fas-induced apoptosis by competing with procaspase-8 for FADD binding, thus preventing the recruitment of caspase-8 to FADD, or by inhibiting caspase-8 activation through DISC [239,240]. Several studies have reported that PCa cells treated with 2-ME2 displayed diminished FLICE-inhibitory protein (FLIP) expression (Figure 3) [214,215,218].
2-ME2 proapoptotic effects through the activation of the intrinsic pathway of apoptosis encompass the regulation of several members of the Bcl-2 family, altering the ratio of and antiapoptotic proteins. This seems to occur due to the indirect effect of 2-ME2 reducing the levels of phosphorylated Akt as well as by other mechanisms [213,217,220,241,242,243,244,245,246]. Exposure to 2-ME2 increased the levels of the proapoptotic proteins Bax and Bim and reduced the expression of Bcl-2, an antiapoptotic member of the family [213,218,220,247]. The proapoptotic Bid, which promotes the leakage of cytochrome C from mitochondria, connects the intrinsic and extrinsic apoptotic pathways as it is activated by caspase-8 [248,249]. A study showed that 2-ME2 could induce Bid cleavage with consequent reduction in Bcl-2 and caspase-8 activation [218].
The 2-ME2 induction of apoptosis in PCa cells also seems to be correlated with the reduced levels of X-linked inhibitor of apoptosis protein (XIAP), a member of the inhibitor of apoptosis family of proteins (IAP) that inhibits caspase activity [235].
The proapoptotic actions of the 3β-Adiol metabolite were shown to induce the apoptosis of PCa cells through the intrinsic pathway with a reduction in Bcl-2, increase in Bax expression, and involvement of caspase-9, which culminates in the augmented activity of caspases-3/7 [225]. Furthermore, the 3β-Adiol activation of apoptosis by the mitochondrial pathway seems to be triggered by the increased transcription of the transcription factor Forkhead box class O 3a (FOXO3a), which causes an increase in the expression of p53 upregulated modulator of apoptosis (PUMA) (Figure 3) [225].

5.1.2. Synthetic Estrogens

Several synthetic compounds with estrogenic activity have been developed over the years with the perspective of being used as anticancer molecules, suppressing PCa cell growth. With this rationale, the synthesized compounds were generally developed as ERβ agonists, as this receptor has been assigned to have antiproliferative and proapoptotic properties. Examples include the 8β-VE2 and APVE2 compounds (Figure 3 and Supplementary Table S1).
High doses of 8β-VE2 induced apoptosis in the PCa cell lines and the prostate of rodent tumor models (Supplementary Table S1), which was related to the interference in androgen/AR signaling. Androgens are the main players promoting PCa cell survival, and androgen deprivation is well-known to block prostate cell growth [250,251]. For this reason, androgen deprivation therapy (ADT) is a gold standard in PCa treatment. A recent study demonstrated that stimulating ERβ activity with the specific agonist 8β-VE2 diminished survival and increased the apoptosis of VCaP cells [252]. This human PCa cell line overexpresses the AR and its splice variants are linked with the resistance to ADT. Interestingly, 8β-VE2 treatment reduced the overexpression of AR and AR variants in VCaP cells under androgen deprivation [252], which renders this compound interesting in overcoming resistance to treatments. Results in mice models of BPH and PCa treated with 8β-VE2 demonstrated that ERβ causes apoptosis through the activation of the extrinsic pathway and TNFα mediation in an androgen-independent manner [253,254]. Other studies have reported that 8β-VE2 induces the apoptosis of PCa cells by activating both the extrinsic and intrinsic pathways [225,253]. 8β-VE2 has been shown to increase the expression of caspase-8, with the subsequent activation of caspase-3, in a TNFα mediated process [253]. Concerning the intrinsic pathway, 8β-VE2 reduced Bcl-2 expression and increased the cleavage of caspase-9 [225]. Furthermore, 8β-VE2-induced apoptosis by the intrinsic pathway seems to be mediated by the increased levels of FOXO3a [225]. The APVE2 compound was shown to reduce PCa cell growth by arresting the cell cycle at the G2/M phase and inducing apoptosis, with effects perceived at nanomolar concentrations (Supplementary Table S1) [255]. Studies concerning the antiproliferative and proapoptotic actions of APVE2 are scarce, and the mechanism is highly unknown.
Another well-known synthetic estrogen is DES, which has been widely used for CRPC treatment [98]. This compound diminishes PCa cell viability [256], reduces proliferation, and augments apoptosis (Figure 3) [147,257,258]. DES treatment depleted the population of CRPC cells in the G1 and S-phase, with an accumulation of cells in the G2/M phase [257,259]. DES has been shown to decrease the phospho-ERK levels and c-Jun N-terminal kinase (JNK) activation, increase the phosphorylation of the cell-cycle inhibitor protein p16INK4A, and activate p38 kinase, which then phosphorylates cyclin D1, tagging it for degradation and arresting cell cycle. This process was shown to be mediated by ERβ as well as by GPER [147].
Furthermore, the actions of DES suppressing cell division include the inhibition of telomerase activity, which is highly upregulated in dividing cells such as malignant cells [258] and the disruption of microtubule organization [259]. PCa cells stimulated with DES showed a decreased density of microtubules [259], with the suppression of β-tubulin isotypes I, II, and IV, and α-tubulin isotypes I and IV as well as the diminished expression of glyceraldehyde-3-phosphate dehydrogenase [259], which is a microtubule-associated protein that plays a role in microtubule bundling [260].
In addition, DES has been shown to inhibit AR transactivation activity in the PC3 and DU145 PCa cell lines, which indicates its antiandrogenic effects by attenuating the levels of AR-mediated transcription [261]. Other mechanisms associated with DES include the inhibition of HIF1α activation and changes in a panoply of metabolites including lactate, phosphocreatine, and reduced glutathione [262]. This suggests that DES can have a putative role in the metabolic rewiring of PCa cells.
HE3235 is another synthetic estrogen, which was suggested as potential clinical use for treatment of CRPC. HE3235 significantly decelerated tumor growth, decreased AR expression, lowered intratumoral testosterone and DHT levels, and inhibited the development of bone tumors [263,264].
Not surprisingly, the review of the existing knowledge concerning the role of estrogens in controlling PCa growth showed the consistent and powerful effects of these hormones in counteracting PCa cell proliferation (Figure 3). Indeed, these beneficial effects have been known for decades, with estrogens being used in PCa therapy, and only disregarded because of the important adverse effects reported. Notwithstanding, the evidence of the antitumorigenic role of estrogens has prompted the appearance of studies evaluating the effect of these hormones in combination with both chemotherapeutic drugs and natural compounds [201,211,212,220,265] as a strategy that could potentiate the action of each compound alone, allowing for a reduction in the doses used, and therefore, overcoming the problems related with collateral effects.

5.2. Suppression of Metastization

Tumor progression is a complex process encompassing several biological mechanisms that determine the molecular, morphological, and functional changes in cancer cells and tumor microenvironment. These changes result in the reorganization of the extracellular matrix, the promotion of angiogenesis, inflammation, and modulation of the immune system [266], which altogether drive tumor progression, invasion, and metastization.
Loss of cell–cell adhesion, which depends on the disruption of cell-to-cell and cell–matrix interactions, with the involvement of several molecular players, is critical for the occurrence of cell migration and invasion. E2 and 2-ME2 seem to interfere with tumor progression by upregulating and stabilizing the β-catenin protein, an important molecule in cell–cell adhesion [217,247,267]. β-catenin is a key component of the cadherin/catenin complex that mediates calcium-dependent homophilic interactions between cell adhesion molecules [268]. Furthermore, 2-ME2 has been shown to reduce the migratory ability of PCa cells by increasing the interaction of β-catenin with E-cadherin [247]. E-cadherin is the most abundant adhesion molecule in epithelial adherens junctions, and its loss is frequently associated with migration, invasion, and metastatic dissemination [269]. The estrogenic metabolite 3β-Adiol increased E-cadherin expression and cell adhesion, which resulted in a reduction in migration and invasion [226,227]. In vivo results confirmed that 3β-Adiol decreased PCa progression with the ability to reduce metastasis formation [226]. Furthermore, it was reported that 3β-Adiol controls the progression of PCa through the activation of ERβ [226,227]. Accordingly, studies in CRPC cells reported that ERβ activation leads to the increase in E-cadherin and β-catenin expression and the reduced expression of N-cadherin [267,270].
Considering MMPs, other important molecules involved in tumor cell invasion and metastasis, Kanagaraj et al. [199] reported that E2 was able to reduce the expression levels of MMP-2 and MMP-9. These findings are highly interesting as both MMPs are prostate gland secretion products, and their higher levels are related to the aggressive behavior of PCa and metastization [271,272].
The angiogenic process is essential for tumor cell survival, growth and metastasis by providing nutrients and oxygen to the growing tumor cells [273]. Angiogenesis comprises endothelial cell proliferation and migration, which can be activated by tumor cells via paracrine factors or direct cell-to-cell communication [274,275]. The vascular endothelial growth factor (VEGF) is a crucial angiogenic factor with mitogenic and antiapoptotic effects. It regulates angiogenesis by inducing the proliferation, migration, and permeability of endothelial cells [276]. VEGF expression is highly enhanced in developing tumors [277]. LAPC-4 or LNCaP cells in conditioned media were shown to induce the growth of a murine endothelial cell line (MEC), and VEGF was the main factor responsible for this paracrine stimulation [278]. E2 administration inhibited the paracrine effect of DHT in stimulating MEC proliferation [278]. Accordingly, both E2 and its stereoisomer αE2 reduced the number of microvessels in the prostate tumor tissues of the LAPC-4 or LNCaP xenograft models [278]. Additionally, E2 seems to suppress angiogenesis by inhibiting the ERβ- and KLF5-mediated expression of platelet-derived growth factor subunit A (PDGFA) [14]. This factor plays an essential role in regulating cell migration and chemotaxis [279], and its overactivity can trigger carcinogenesis [280]. The E2 metabolite 2-ME2 has also displayed antiangiogenic properties. 2-ME2 administration to LNCaP and PC3 xenografts reduced the prostate microvessel density [211,219,220], which occurred by decreasing the VEGF expression [220].
Estrogenic compounds have also been shown to control inflammation in PCa. This is the case of 2-ME2, which inhibited the progression of PCa in TRAMP mice by increasing the expression of TNF-α-stimulated gene 6 (TSG-6), a secreted glycoprotein with anti-inflammatory action [221]. No further evidence exists of the anti-inflammatory actions of estrogens either in vitro or in vivo. Nevertheless, a study in noncancer patients showed that circulating estrogens were inversely associated with intraprostatic inflammation [56], which suggests that these hormones can have a role in controlling prostate inflammation and PCa progression.

6. The Phytoestrogen Scope

Among all the natural products that have been used in improving human health as either preventive supplements or treatments, phytoestrogens are a class of compounds of particular interest. The relevance of phytoestrogens in human health relies on their wide distribution in several types of plants and fruits and their potent and diverse biological effects. Phytoestrogens have recognized antioxidant, antiproliferative, antiangiogenic, and proapoptotic activity [36], making them highly attractive for exploitation in the context of anticancer therapy. Furthermore, many epidemiological studies support the protective role of phytoestrogens in malignancy. For example, the incidence of breast and PCa is much lower in Asian people when compared to Westerners [281]. The average daily consumption of phytoestrogens in Asia is estimated to range from 20 to 50 mg, whereas in the United States and Europe, it is predicted to be 0.153 mg and 0.491 mg, respectively [282]. This astonishing difference is justified by the dietary habits of the Asian population, with a plant-based diet and much less consumption of meat and animal-derivative products [283], and it has been postulated to explain the lower incidence and mortality rates of PCa in Eastern countries [284,285].

6.1. Classification and Structure

Phytoestrogens are a group of natural biologically active compounds derived from plants and fruits, which, because of their analogous structure to the principal estrogen, E2, display estrogenic and/or antiestrogenic activity [286]. A great group of phytoestrogens are the phenolic compounds, encompassing two main classes of molecules: flavonoids and nonflavonoids. The first group includes flavones, isoflavones, flavanones, flavonols, and coumestans [287,288,289]. Stilbenoids (and lignans) are included in the nonflavonoid class [287,288]. Supplementary Table S2 lists the types of phytoestrogens with an identified role in modifying human prostate cell fate.

6.2. Sources and Metabolism

Phytoestrogens are abundant in nature and are mainly present in the fruits, vegetables, and whole grains usually consumed by humans [290]. Moreover, they are also found in several edible and/or medicinal plants [290]. Plant extracts with recognized estrogen-like activity comprise soy, red clover, kudzu, hops, licorice, rhubarb, yam, and chasteberry, among others [291].
After consumption, phytoestrogens are metabolized by intestinal bacteria, absorbed, conjugated in the liver, disseminated in plasma, and expelled in urine [292]. Gut metabolism has been shown to play a crucial role in determining the effectiveness of the actions of phytoestrogens. For some members of this class of compounds, the estrogenic effect is mainly due to the metabolites generated by digestion, rather than by the original phytoestrogen molecule [293]. For example, daidzein is a phytoestrogen that is converted to a more active metabolite, equol, with enhanced estrogenic activity compared with the original molecule [294]. However, the effect of equol is variable, depending on individual specificities since the ability for this conversion and the extension of this reaction appears to be restricted to approximately a third of the population [294]. Moreover, the bioavailability and uptake of some isoflavones require some metabolic processes that allow absorption into the peripheral circulation such as hydrolysis and reconjugation to glucuronic and sulfuric acids [295,296].
Despite some limitations on bioavailability and bioaccessibility, the potential of phytoestrogens as anticancer molecules is unquestionable, and continuous research would help in identifying new preventive or treatment approaches.

6.3. Mechanisms of Action

As above-mentioned, the molecular structure of phytoestrogens is very similar to E2, and they can bind ERα and ERβ. However, the binding affinity seems to be higher for ERβ [297,298]. After ligand binding, the receptors are translocated to the nucleus, where they bind to specific regulatory regions, the estrogens’ response elements, modulating gene transcription, and consequently, the cell protein network and activity [299]. Besides their ability to bind to ERs, phytoestrogens can modulate the activity of other transcription factors and pathways, namely serotoninergic and IGF-1 receptor signaling [300,301,302]. The effect of these compounds can also be exerted by binding free radicals, inducing DNA methylation and histone modification and modulating tyrosine kinase, cAMP/protein kinase A, cGMP/NO, PI3K/Akt, and MAP kinase activity [303,304,305,306].
In addition to classical ERs, phytoestrogens have also been shown to activate GPER [307].

6.4. Phytoestrogens Actions against Prostate Cancer

6.4.1. Apigenin

Apigenin, also known as 4′,5,7,-trihydroxyflavone, is a nontoxic and nonmutagenic flavonoid widely present in common fruits and vegetables [308]. This phytoestrogen has been proven to have anti-inflammatory and anticarcinogenic effects in preclinical studies. Several types of cancer are sensitive to apigenin, namely leukemia, breast, colon, lungs, skin, thyroid, and prostate [309,310,311].
The effects of apigenin have been shown to inhibit PCa growth, reducing cell proliferation and inducing cell cycle arrest at the G2-M phase (Figure 4) [312,313,314,315]. In LNCaP and DU145 cells, cell cycle inhibition by apigenin has been linked to a marked decrease in the expression of cyclins D1, D2, and E and their activating partners CDK2, 4, and 6 [316,317]. This phytoestrogen also increased the expression of tumor suppressor protein p53 and cell cycle inhibitor p21 [313,316,317,318,319,320,321] and caused the dephosphorylation and inactivation of retinoblastoma protein (Rb) (Figure 4) [322].
The antiproliferative effect of apigenin seems to be mediated through the activation of ERβ or the suppression of the IGF axis, one of the well-known crucial pathways for cell proliferation [323,324,325]. Although no studies were performed on PCa, apigenin also acts through GPER [326].
Apigenin’s actions are also significant in relation to the control of NF-κB [327,328] and MAPK and Akt/PI3K survival pathways [322,329]. Inactivation of the Akt pathway, and stimulation of the production of reactive oxygen species (ROS), culminated in the apoptosis of PCa cells [321,330,331]. Akt is known to phosphorylate the Bad protein, which renders the antiapoptotic Bcl-2 protein free and activated and cells resistant to death. Thus, Akt inactivation is promptly linked to increased apoptosis as Bcl-2 is inactivated. Apigenin treatment has been shown to shift the Bax/Bcl-2 ratio toward apoptosis by increasing Bax and reducing Bcl-2 expression (Figure 4) [316,318,320,321].
Existent reports also indicate that apigenin can induce apoptotic cell death triggered at the cell membrane. Stimulation of PCa cells with apigenin strongly induced the expression of DR5 and TNF-related apoptosis-inducing ligand (TRAIL) death receptors [332,333] as well as caspase-8, caspase-10, caspase-9, and caspase-3 [317,330,331,332].
Of note, apigenin treatment significantly decreased the viability of cancer cells and augmented apoptosis, in contrast to the low magnitude of effects observed in non-neoplastic cells [312,332], indicating a selectivity over PCa cells.
Apigenin also inhibited the cellular processes related to cancer progression such as EMT, migration, and invasion [334]. Exposure to apigenin resulted in decreased cell motility and the reversion of EMT [334,335] as well as the blockade of β-catenin signaling [336] and inhibition of the migration and invasive potential of PCa cells [337]. These effects were shown to be dose- and time-dependent [337].
In LNCaP and PC3 cells, apigenin treatment also inhibited the expression of VEGF and hypoxia-inducible factor α (HIFα), angiogenesis, and hypoxia markers, respectively [338,339,340]. HIFα is a known driver of the hyperglycolytic phenotype of cancer cells in response to hypoxia by regulating the expression of several targets in the glycolytic flux, namely, glucose transporters (GLUTs), as in the case of GLUT1 [341]. Accordingly, in the presence of apigenin, PCa cells displayed a decreased expression of HIFα, accompanied by the diminution of GLUT expression [342,343], which likely limited the nutrient availability, contributing to suppress proliferative activity. Exposure of PCa cells to apigenin also diminished the activity of fatty acid synthase (FASN) [342,344], a target metabolic regulator whose expression is highly increased in PCa [345]. Overall, these findings indicate the potential of apigenin to counteract the metabolic rewiring in PCa.
The beneficial properties of apigenin could be observed in animal models [346], and one study reported that a herbal extract containing apigenin was able to enhance the chemotherapeutic effect of docetaxel in PCa [347]. Furthermore, apigenin combined therapy enhanced the efficacy of abiraterone acetate [348] and doxorubicin [349], and sensitized the PCa cells to radiation [350]. Moreover, apigenin sensitized PCa stem cells to therapy, which is relevant as this cell population is generally resistant to therapy and is believed to be involved in the nonresponse of PCa to anticancer drugs and disease recurrence [37,38]. Overall, the present data support the interest in this natural agent.

6.4.2. Chrysin

Chrysin, also known as 5,7-dihydroxyflavone, is a flavone that can be extracted from honey, propolis, and blue passion flowers [351]. This agent is considered as a valuable resource for health purposes as it shows anticancer, antiangiogenenic, anti-inflammatory, antidiabetic, antibacterial, antiaging, and antiallergic properties [352,353,354,355,356,357,358]. Chrysin exhibited some binding activity to the nuclear ERs and GPER [359,360,361]; however, there is no evidence of the ERβ- or GPER-mediated actions of this compound in PCa. Nevertheless, chrysin suppressed proliferation and induced the apoptosis of PC3 cells [362,363,364]. One of the mechanisms by which this substance induces apoptosis is through the production of ROS [363]. HIF-1α is another signaling pathway that seems to have a role in chrysin anticancer activity [365]. In DU145 cells, chrysin decreased HIFα expression by inhibiting its protein synthesis (Figure 4) [352].
In parallel to the effects controlling PCa cell fate, a report showed that the use of chrysin as a complement to docetaxel increased the therapeutic efficacy of this anticancer drug, mitigating some side effects such as edema [366].

6.4.3. Biochanin A

Biochanin A, 5,7-dihydroxy-4′-methoxyisoflavone, is an O-methylated isoflavone that can be found, for example, in red clover, soy, alfalfa sprouts, peanuts, and other legumes [367]. This flavonoid has been shown to induce a dose-dependent inhibition of LNCaP, DU145, and PC3 cell proliferation and viability [319,368]. Furthermore, it has been suggested that ERβ could mediate the antiproliferative effect of biochanin A, as this compound stimulated the receptor expression in PC3 cells [368]. Although biochanin A action through GPER has been reported [369], no studies have been caried out in PCa.
The capability of biochanin A to suppress cell division, with the accumulation of PCa cells in the G0 phase [370], was related to the decreased expression of cyclins B and E and oncogene c-Myc as well as with the increased expression of cell cycle inhibitor p21 [319,368,370]. Akt and ERK were other proliferative and survival signaling pathways associated with the actions of biochanin A in PCa cells [368].
In addition, this isoflavone was able to increase the apoptotic rate of LNCaP and PC3 cells [319,368,370,371], which was suggested to be mediated by the TRAIL cell death receptor [371]. Also, biochanin A modulated the expression levels of mitochondrial regulators Bax and Bcl-2, controlling apoptosis (Figure 4) [368].
The administration of biochanin A in vivo reduced the incidence and tumor size in an LNCaP xenograft model [370]. Studies exploring the potential of biochanin A as a possible therapy in PCa are inexistent. However, a report showed that it reduced the PSA production by PC3 cells [372].

6.4.4. Daidzein

Soybean is the major source of daidzein, a natural isoflavone chemically known as 7-hydroxy-3-(4-hydroxyphenyl)-4Hchromen-4-one [373]. Other legumes and fruits can also be important sources of daidzein [373].
Similar to other flavonoids, daidzein has antiproliferative effects in PCa cells (Figure 4). Exposure of PC3 cells to a soy isoflavone concentrate highly enriched in daidzein caused the accumulation of cells in the G2/M phase of the cell cycle, underpinned by the increased expression of p21 [374].
BPH-1, LNCaP, and PC3 cells treated with daidzein exhibited higher apoptotic rates than the control untreated groups [375,376,377]. Concerning the activated apoptotic pathway, this isoflavone increased the expression of DR4 and DR5 death receptors and thus triggered TRAIL-mediated apoptosis [377]. In PC3 cells, treatment with daidzein increased the expression of the proapoptotic Bax protein [375], also implicating the intrinsic apoptosis pathway.
Additionally, several studies have reported that daidzein can reduce PCa growth [378,379,380,381,382] by suppressing migration, invasion, and angiogenesis (Figure 4) [383,384,385].
This isoflavone has been shown to interact with ERβ with some selectivity [364,386], and novel daidzein analogs showed anticarcinogenic activity in PCa cells through ERβ mediation [387]. Also, GPER seems to be involved in the daidzein mechanism of action, with reports existing in other cancer types [388,389]. Further investigation is needed to clarify whether ERs mediate the daidzein effects in PCa.
The remarkable beneficial effects of daidzein counteracting the cancer hallmarks were tested in clinical trials. Men diagnosed with PCa consuming a daily phytoestrogen-enriched diet containing daidzein showed the favorable evolution of PSA levels and free/total PSA ratio [390]. Other significant observed effects included tumor growth inhibition and a chemopreventive action in prostate carcinogenesis, with nontoxic effects [391,392,393,394,395,396,397,398,399,400,401,402,403,404].

6.4.5. Equol

Equol, (3S)-isoflavan-4,7′-diol, is a metabolite of the isoflavone daidzein produced by the activity of intestinal bacteria [405,406]. Only about 30 to 50% of the population can convert daidzein to equol, with a much higher prevalence found in Asian countries compared with Western ones [407,408]. Equol has two diastereoisomers, R-equol and S-equol, with the last one being the natural form. Equol has been shown to have a modest binding affinity for ERs, though it has been suggested that it mimics estrogen’s effects via ERβ and GPER [409,410,411]. Equol was reported to have stronger bioactivity than daidzein and genistein, namely regarding antioxidant activity [412].
The antiproliferative activity of equol in PCa (Figure 4) has been demonstrated in LNCaP, CxR, 22Rv1, and PC3 cells. Equol administration resulted in the inhibition of cell proliferation and diminished cell growth [413,414,415]. Moreover, equol increased the number of LNCaP, LAPC-4, 22Rv1, DU145, and PC3 cells arrested in G0 [413,414]. However, the mechanisms by which equol controls cell proliferation and the dependency on ERs still need clarification.
Equol was also shown to increase the apoptosis of PCa cells (Figure 4) [377,413,414,416]. Despite the information provided by several studies, the mechanisms underlying the proapoptotic effect of equol is highly unknown. However, in LNCaP cells, the presence of this compound was shown to increase the expression of death receptors, specifically the TRAIL receptor [377]. It is also possible to speculate that the mitochondrial pathway can trigger death-induction by equol, as high concentrations of this compound have been related to a significant increment in DNA damage [416].
Although the information available is very limited, equol has been shown to inhibit the invasion of DU145 cells probably by downregulating the expression of MMP-2 and MMP-9 [417].
Reports in animal models and PCa patients also support the beneficial effects of equol delaying prostate tumor growth [391,397,418,419,420,421,422]. Feeding rats with soy flour (containing daidzein, which is converted to intestinal metabolite equol) inhibited the growth of Dunning R3327 prostate adenocarcinoma [391]. In the TRAMP mice, prostate carcinogenesis induced by a high-fat diet occurred concomitantly with the lowered equol serum concentrations because of the adverse effects over equol-producing bacterium [421], which demonstrates the relevance of this bioactive compound in prostate physiology. Equol’s effects include the reduction in PSA levels and blockage of the PSA increase in response to DHT stimulation both in vitro and in vivo [420]. These findings were mirrored in human patients. Men with higher levels of equol in their diet presented reduced PSA levels [397,423] and lower PCa risk and incidence [397,418,422].

6.4.6. Formononetin

The isoflavone formononetin, 7-hydroxy-4′-methoxyisoflavone, can mainly be found as a component of red clover plants [424]. This type of plant is a component of traditional Chinese herbal medicine and has been widely used in China for thousands of years. Formononetin seems to promote cell cycle arrest in PC3 cells by downregulating the Akt pathway and cyclin D1 and CDK4 expression [425]. Moreover, the exposure of LNCaP, DU145, and PC3 cells to formononetin resulted in the induction of apoptotic cell death, which was shown to occur by several mechanisms, namely the augmentation of the Bax/Bcl-2 ratio, suppression of the p38/Akt pathway through the downregulation of the IGF-1/IGF-1R signaling pathway, and the inactivation of ERK1/2 mitogen-activated protein kinase (Figure 4) [426,427,428,429].
A recent study showed a synergistic antitumor effect in PCa cells through the combination of formononetin with docetaxel in one nano-sized drug delivery system [430].
Formononetin can interact with ERβ [364] and GPER [431], with ER-mediated effects reported in breast cancer [432]. No evidence of this relationship exists in PCa, and further research is needed to implicate ERs in the antiproliferative and proapoptotic actions of formononetin over prostate cells.

6.4.7. Genistein

Genistein, chemically known as 4′,5,7-trihydroxyisoflavone, is the main isoflavone present in the human diet, as it is predominantly included in the composition of soybeans, peas, lentils, and other beans [433]. This compound has been shown to interact with both ER isoforms, being one of the most potent phytoestrogens stimulating the transcriptional activity of both ERα and ERβ [364]. However, genistein interaction with ERα was shown to be one-thousandth of the potency of E2, whereas for ERβ, the potency was one-third of that of E2 [364]. Therefore, it is the selective binding to ERβ that sustains the genistein anticancer activity [364,434,435]. Nevertheless, genistein action through GPER has been reported in other types of cancer except PCa [436,437].
Multiple biological processes in PCa cells have been shown to be modulated by the activity of genistein (Figure 4). This compound seems to decrease the viability and proliferation of LNCaP, 22RV1, DU145, and PC3 cells [380,438,439,440,441,442,443,444,445,446,447,448,449,450], which has been linked to the downregulated expression of cyclin B and the upregulation of the cell cycle inhibitor p21 [438,439,445]. Furthermore, in LNCaP, DU145, and PC3 cells, genistein arrested the cell cycle in G2/M [438,439,440,441,444,445]. Curiously, cell growth inhibition by genistein was associated with suppressing telomerase activity and inhibiting stemness by targeting the hedgehog pathway [442,443]. Another mechanism linked to the genistein anticancer activity was its ability to downregulate oncogene MDM2 expression, both at the transcriptional and posttranslational levels [451].
Genistein also strongly modulates the survival of PCa cells. It inhibits the activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NFkβ), mediated by Akt signaling [452]. Genistein also inhibited the radiation-induced activation of the NFkβ pathway in PCa cells [438] and Akt phosphorylation, which resulted in increased apoptosis [444,453]. Another study reported that this flavonoid could abrogate epidermal growth factor (EGF)-induced activation of Akt and inhibit the Akt kinase activity [452].
The reported in vitro findings were mirrored in human PCa cases. By analyzing radical prostatectomy specimens, it was shown that the consumption of genistein was related to the increase in apoptotic rate [394,453], together with the upregulation of p53 [453].
Genistein can also control the metastatic spread of PCa cells. This natural compound has been demonstrated to inhibit the contact-stimulated migration of PCa cells [383,444,454] and suppress MMP-2 activity and cell invasion [444,455,456,457,458]. Furthermore, it reversed the EMT in PCa cells by increasing E-cadherin expression and reducing that of vimentin at the mRNA and protein level [459,460]. Moreover, genistein-treated DU145 and PC3 cells displayed a decreased activity of focal adhesion kinase [461], which was suggested to control the aggressiveness of PCa [462].
Additionally, the angiogenic process in PCa seems to be modulated by genistein, since treatment with this flavonoid suppressed VEGF expression [463,464].
Interestingly, the stimulation of LNCaP, LAPC-4, VCap, and PC3 cells with genistein also decreased or even inhibited PSA expression [465,466].
Genistein, like other plant-derived compounds, can induce epigenetic alterations in human cells, altering the landscape of active molecular targets [467]. In PCa, studies showed that the exposure of LNCaP and LAPC-4 cells to this natural agent reduced the methylation of the ERβ promoter, thus increasing the receptor expression [468], which can be an additional mechanism enhancing the estrogenic effects of genistein.
This phytoestrogen can also modulate DNA methylation in the promoter regions of retinoic acid receptor β (RARβ2), glutathione S-transferase P1 (GSTP1), Ras association domain family 1 (RASSF1A), and ephrin B2 (EPHB2) genes, which seems to be protective against PCa [469,470]. Moreover, this natural compound increased the expression of the tumor suppressor gene BTG3 in the RWPE-1, DU145, and PC3 cells as well as the sFRP1 and Smad4 genes via demethylation and histone modification [444,471].
Noteworthy, genistein has also been reported to modulate the expression of regulatory microRNAs. It reduced the expression of the oncogenic miRNAs miR-1260b and miR-151, which are upregulated in PCa [444,472]. On the other hand, genistein treatment increased the tumor suppressor miR-574-3p, which is downregulated in PCa [473].
Studies in animal models have confirmed the beneficial properties of dietary genistein in suppressing the development of PCa [474,475,476,477]. A low-fat diet with soy protein and isoflavones, namely genistein, reduced the tumor growth in LNCaP xenografts [478]. Furthermore, genistein in the diet seems to reduce the incidence of poorly differentiated PCa and improve survival in TRAMP mice [479,480,481,482]. This antitumor role of genistein is supported by reports that showed a reduction in the proliferation and increase in apoptosis in the ventral prostate of Simian Virus-40 T-antigen (SV-40 Tag) targeted probasin promoter rat model, a transgenic model developing PCa spontaneously [476], and in the cancerous prostate dorsolateral prostate of Lobund-Wistar rats [475]. At the molecular level, genistein reduced the cyclin D1 levels post-transcriptionally, disrupting cell cycle progression, and thus reducing proliferation [479]. One of the molecular mechanisms related to these genistein effects is PTEN/Akt axis regulation. Genistein in the diet upregulated PTEN, inhibiting the activation of Akt and restoring the activation of GSK-3β [475,479]. Furthermore, genistein in the diet retained cadherin-1 expression through the decrease in snail-1 transcription [479], which can be critical in maintaining the integrity of prostatic epithelial cells and retarding cancer progression.
Curiously, this natural compound reduced the AR mRNA and protein levels [475,483] as well as ERα and ERβ expression in the rat prostate [483]. Moreover, genistein was shown to downregulate the AR levels in LNCaP cells through the activity of ERβ [484], which highlights that aside from their direct actions, estrogens could modulate the response to androgens. In addition, genistein decreased steroid receptor coactivator-3 (SRC-3) levels in the ventral prostate of SV-40 Tag rats [476]. SRC-3 is an important regulator of PCa proliferation and survival and is essential for the progression of prostate tumorigenesis in the TRAMP model [485]. Another interesting effect of genistein is its ability to reduce intraprostatic DHT levels together with the diminished expression of 5α-redutase-2 [474], corroborating the benefit of genistein to prevent prostate diseases such as PCa.
The protective role of genistein against PCa development is also supported by the reduction in IGF-1 [474,476], a player that has been associated with the increased risk of an advanced stage of PCa [486], and the inhibition of osteopontin [480], an extracellular matrix protein secreted by infiltrating macrophages and prostate tumors cells themselves [487,488,489], which is related to increased tumorigenicity and metastatic ability [490].
From a therapeutic perspective, the beneficial effects of genistein, when joined with classical chemotherapy and antiandrogenic drugs, are worth noting. In fact, this isoflavonoid has been tested in clinical trials with PCa patients with promising results [397,491,492,493,494,495,496,497].
The combination of genistein with polysaccharides enhanced the activity of docetaxel, bicalutamide, and Src kinase inhibitors in both androgen-sensitive and CRPC cells, namely, in LNCaP, CWR22Rv1, and PC3 cells [498]. Furthermore, this natural agent increased the efficacy of cabazitaxel in C4-2, ARCaPM, and PC3 cells [499] and paclitaxel in LNCaP and DU145 cells [500]. Genistein has also been shown to potentiate the effect of radiation, diminishing PCa cell proliferation and tumor growth [501,502,503,504,505]. The use of BIO 300, a nanosuspension of genistein, sensitized human PCa xenografts to radiation therapy [506].
Additionally, several reports have demonstrated that genistein combined with SB715992, an experimental inhibitor of kinesin spindle proteins that play an essential role in mitotic spindle formation, can improve the outcomes in PCa patients [507].
A limitation found in clinical trials for using genistein in treatment is its reduced bioavailability upon oral delivery. Using nanostructured delivery systems carrying genistein is a possible solution to increase its bioavailability and enhance anticancer action against PCa [508]. Some approaches have been tested with good results, namely, a nanoliposomal formulation encapsulating celecoxib and genistein was shown to inhibit the cyclooxygenase-2 (COX-2) pathway and GLUT1 expression, preventing PCa cell proliferation [449]; another liposomal formulation containing plumbagin and genistein inhibited proliferation and induced the apoptosis of PCa cells [509,510], while nanoparticles loaded with genistein and doxorubicin reduced the development of PCa metastasis by amplifying oxidative stress damage [511], and genistein-gold nanoparticle conjugates displayed antioxidant and antitumorigenic effects in PCa [512].
Despite genistein’s beneficial effects against PCa, reports that demonstrated a dual effect of this compound in PCa cells and in vivo [513,514,515,516,517] should be given attention. Moreover, it is critical to deeply understand the mechanisms underlying genistein actions in PCa and establish the ideal threshold dose to avoid dual effects before disseminating its use in treatment.

6.4.8. Naringenin

Naringenin, 4′,5,7-trihydroxyflavanone, is a bioactive compound mainly found in grapefruit and orange [518,519], which has been demonstrated to have antiproliferative and proapoptotic activity in PCa cells (Figure 4) [520]. Information concerning the effects of naringenin and its mechanism of action in PCa is still scarce. However, naringenin-reduced oxidative stress [521] and induced apoptotic cell death mediated by the PI3K/Akt and MAPK signaling pathways [520]. This biologically active substance is also able to repress the migration and invasion of PCa cells by increasing the expression of E-cadherin and diminishing the vimentin expression levels [522,523]. On the other hand, the exposure of LNCaP cells to naringenin stimulated DNA repair, preventing mutagenic changes as a consequence of oxidative damage [524], which raises curiosity about the role of this compound against early events in human prostate carcinogenesis.
Naringenin interaction with ERs has been demonstrated, with a stronger binding affinity and higher transcriptional activity for ERβ [364]. It has also been shown to modulate the receptors’ expression levels in breast tumors, decreasing ERα expression while increasing ERβ [525]. Thus, it is reasonable to assume that the actions of naringenin in PCa could be mediated by ERs, with a relevant role for ERβ. Additional investigation is needed to clarify this relationship and further explore the actions of naringenin.

6.4.9. Kaempferol

Kaempferol, 3,4,5,7-tetrahydroxyflavone, is a secondary metabolite found in many plants, plant-derived foods, and traditional medicines [526], which seems to have an important role against PCa (Figure 4). The beneficial effects of kaempferol have been demonstrated by a set of in vitro and in vivo experimental evidence [527,528,529,530,531]. Several studies have reported the antiproliferative effects of kaempferol [381,532,533] by increasing the proportion of PCa cells blocked in the G2/M phase [381,533] and its ability to inhibit the DHT-stimulated growth of LNCaP cells [532,534,535]. Moreover, the proapoptotic properties of kaempferol have been described [530,532,536,537]. This compound augmented the apoptosis of PCa cells, which was accompanied by the increased expression of TRAIL, enhanced levels of caspase-8, caspase-9, and caspase-3, and the inhibition of the NFkβ pathway [530,536,537].
A study addressing the effect of a mix of flavonoid compounds including kaempferol in a preclinical murine model demonstrated the ability of these compounds to target AR signaling and inhibit PCa growth [531]. Despite the known selectivity of kaempferol for ERs [364], no ER-mediated effects were reported in PCa. Nevertheless, as shown in other tissues, it seems to mainly act by antagonizing ERα activity [538,539].

6.4.10. Myricetin

Myricetin, 3,3′,4′,5,5′,7-hexahydroxyflavone, is a family member of flavonoids normally found in vegetables, fruits, nuts, berries, tea, and red wine [540,541]. This phytoestrogen has recognized antioxidant properties [542], and has been demonstrated to inhibit proliferation and induce apoptosis in PC3 cells [543]. Moreover, myricetin effectively suppressed the in vivo progression of PCa by disrupting the interaction of the protooncogene protein proviral integration site for Moloney murine leukemia virus-1 (PIM1) with C-X-C chemokine receptor type 4 (CXCR4) [544]. PIM1 is overexpressed in PCa, playing an important role in tumorigenesis, castration resistance, and metastasis [545,546,547,548,549]. PIM1 promotes the phosphorylation and surface expression of CXCR4, supporting the CXCL12–CXCR4 axis responsible for cancer cell proliferation and metastasis [547,550].
An interesting study showed that PLGA-encapsulated myricetin formulation combined with enzalutamide enhanced the effects of enzalutamide alone in a xenograft model of PCa [551].
This compound was shown to bind ERs competing with E2 and was able to reduce ERα expression in breast cancer cells [552,553]. Nevertheless, there is no evidence of the involvement of ERs in mediating the myricetin effects in PCa.

6.4.11. Quercetin

Quercetin, also known as 3,3′,4′,5,7-pentahydroxyflavone, is a polyphenol belonging to flavonoids and is commonly found in several fruits and vegetables, namely capers, lovage, dill, cilantro, onions, apples, and berries [554]. The main biological properties of this phytoestrogen include antioxidant, antiallergic, anti-inflammatory, and antiviral activities [555,556,557]. Additionally, it has been proven that quercetin can be useful against several types of cancer such as lung, liver, breast, colon, cervical, and prostate [309,558].
Quercetin can reduce the viability and proliferation of LNCaP, LAPC-4, DU145, and PC3 cells [533,559,560,561,562,563,564] in part by diminishing the action of growth factors [565,566]. Quercetin has been shown to arrest cell cycle progression in all phases of the cycle [559,564,567,568]. In LNCaP cells, quercetin arrested the cell cycle in the S phase, significantly reducing the number of cells in G1 [564]. The control of cell cycle progression and cell proliferation was shown to depend on the regulation of several molecular targets (Figure 4). Quercetin strongly inhibited the expression of cyclin B1 and CDK1 and increased the levels of p21 and hypophosphorylated Rb protein in a dose-dependent manner in PC3 cells [559,568] and targeted the PI3K/Akt signaling pathway [569]. Moreover, the inhibition of PCa growth by quercetin can be mediated by the reduction in AR signaling, as quercetin demonstrated the ability to suppress AR expression and inhibit receptor function [561,562,566,570]. Considering ERs, although quercetin has been shown to act through these receptors in other tissues [364,436,571], no reports exist concerning the involvement of ERs in PCa cell responses.
Research efforts have been made to clarify quercetin’s actions and the molecular players involved in stimulating PCa cell apoptosis. Quercetin induced the TRAIL-mediated apoptosis of LNCaP, DU145 and PC3 cells by increasing the expression of TRAIL and DR5 cell death receptors [536,572,573]. Moreover, the exposure of PCa cells to quercetin promoted the activation of caspase-3, caspase-8, and caspase-9 and affected the expression of several apoptosis-related proteins. It decreased the levels of heat shock protein 90 [574] and antiapoptotic proteins Bid and Bcl-2 and increased the expression of the proapoptotic protein Bax [543,567,572,575,576,577,578].
Another mechanism reported to diminish the survival of DU145 cells in response to quercetin is the induction of ROS production, which will lead to an increase in oxidative stress, culminating with cell death [579].
Quercetin also affects PCa cell migration and invasion, stemness, and angiogenesis [580,581]. It synergizes with epigallocatechin gallate to inhibit PCa stem cell characteristics, EMT, invasion, and migration [582]. Accordingly, the expression of prostate stem cell antigen is downregulated by quercetin in DU145 cells [583,584]. Moreover, quercetin decreased the migration of PC3 cells induced by TGF-β [585]. In PC3 cells, quercetin also reversed the EGF-induced EMT and invasiveness via activation of the EGFR/PI3K/Akt pathway [586] and downregulated the expression of MMP-2 and MMP-9 [587]. Furthermore, this phytoestrogen demonstrated the capability of reducing angiogenesis by targeting the VEGFR-2-regulated Akt/mTOR signaling pathways [588].
Interestingly, the actions of quercetin also seem to target the immune response, which can be highly relevant for treatment purposes, particularly considering immunotherapy. It stimulated GM-CSF production in PC3 cells, and the conditioned medium of quercetin-treated PC3 cells increased the chemotaxis of human dendritic cells, suggesting that quercetin treatment can promote the recruitment of dendritic cells to the tumor site [589].
Noteworthy, quercetin sensitized PCa cells to chemotherapy and antiandrogenic drugs. In combination with docetaxel, paclitaxel, doxorubicin, or enzalutamide, quercetin increased the sensitivity of PCa cells to treatment, leading to a notorious anticarcinogenic effect compared with the administration of the anticancer drugs alone both in vitro and in vivo [44,45,46,47,590,591,592].
Several robust findings have pointed out the mechanisms that stimulated the effects of chemotherapeutics in suppressing cell proliferation and invasion, increasing apoptosis, and reducing tumor growth in the presence of quercetin such as the inhibition of the PI3K/Akt and signal transducer and activator of transcription (Stat) 3 signaling pathways, increased expression of cleaved caspase-7 [46], activation of the mitochondrial/ROS pathway, decreased expression of the multidrug resistance-related protein (MRP1) [45], and reduced blood concentrations of growth factors like VEGF and EGF [46].
Resistance to the next-generation antiandrogen, enzalutamide, has been proposed to depend to a large extent of the AR splice variant AR-V7 [593]. This constitutively active variant is generated by the alternative splicing of the AR involving the activity of splicing factors such as hnRNPA1 [594,595]. Quercetin reduced the expression of hnRNPA1, and consequently, that of AR-V7, sensitizing resistant PCa cells to enzalutamide treatment [47]. Furthermore, quercetin can reverse docetaxel-resistance in vitro and in vivo through the AR and PI3K/Akt signaling pathways, supporting its clinical use in docetaxel-resistant PCa [596]. Quercetin also increased PCa radiosensitivity by targeting the radiation-induced ARv7 [597], supporting the possibility of applying radiotherapy combined with quercetin in PCa treatment. Studies in rodents further confirmed the potential of quercetin to prevent PCa growth by suppressing cell survival and proliferation and the expression of antiapoptotic proteins [598,599]. Quercetin supplementation reverted the augmented levels of proliferative and antiapoptotic markers as well as the decreased expression of proapoptotic markers resultant of N-methyl-N-nitrosourea plus testosterone induction of prostate carcinogenesis [598]. In this carcinogen-induced PCa model, quercetin also increased the expression of antioxidant enzymes and decreased the expression of insulin-like-growth factor receptor 1(IGFIR), Akt, and AR [598].
The beneficial use of quercetin was also reported in PCa patients [39,40]. One study evaluated the association between serum vitamin D and dietary quercetin and PCa risk in African men. In individuals with vitamin D deficiency, the augmented levels of quercetin in diet were linked to a lower risk of PCa [40]. It has also been reported that metastatic PCa patients receiving increasing doses of quercetin displayed longer PSA doubling time [39]. However, the enormous benefits of quercetin in suppressing PCa growth have some limitations in clinical practice and therapy. As with other natural compounds, quercetin displays reduced water solubility and low bioavailability, which largely conditions the potential of therapeutic application. To solve this problem, a few studies have evaluated the use of nanoparticles to deliver quercetin directly to PCa cells. The use of cationic PEGylated niosome-containing quercetin and quercetin-loaded nanomicelles assembled from DSPE-PEG2000 showed promising results regarding tumor regression [600,601]. More recently, several approaches such as LHRH-conjugated, PEGylated, poly-lactide-co-glycolide nanocapsules [602], carboxylated graphene oxide as a nanocarrier [603], chitosan-coated, quercetin-loaded PLGA nanoparticles [604], and others [605] have shown promising results in controlling PCa growth and progression, opening up new avenues of research for exploiting treatment approaches.

6.4.12. Coumestrol

Coumestrol, 3,9-dihydroxypterocarp-6a(11a)-en-6-one, is a natural organic compound found in soybean, legumes, Brussels sprouts, alfalfa, and spinach [606,607]. Recently, this natural compound was shown to exhibit anticancer activity against several types of cancer [608,609]. However, only a few studies have investigated the effects of coumestrol in PCa cells. Treatment with coumestrol inhibited the proliferation of LNCaP and PC3 cells and stimulated apoptosis by inducing DNA damage and mitochondrial dysfunction as a consequence of the loss of mitochondrial membrane potential and increased ROS production [416,610]. Furthermore, PCa cells treated with coumestrol showed elevated levels of the proapoptotic protein Bad and cleaved caspase-3 and caspase-9 [610].
The antiproliferative and prosurvival properties of coumestrol seem to be mediated by the modulation of the PI3K/AKT and ERK1/2 and JNK MAPK cell signaling pathways, which have also been implicated in the decreased migration of LNCaP and PC3 cells in the presence of this phytoestrogen (Figure 4) [610]. Furthermore, the capability of coumestrol in reducing PCa progression can be related to HIFα suppression. In hypoxic PC3 cells, coumestrol suppressed HIFα by inhibiting ROS-mediated sphingosine kinase 1 [611], a key enzyme that converts sphingosine into sphingosine 1-phosphate [612], a sphingolipid metabolite and a lipid mediator crucial in several biologic processes of carcinogenesis [613,614,615].
In humans, coumestrol intake is associated with a reduced risk of developing PCa [616]. Of note, coumestrol’s actions have been related to its ability to bind ERs, with the capability of the transcriptional activation of both ERα and ERβ at concentrations of 1–10 nM [364,617]. Indeed, like genistein, coumestrol is among the phytoestrogens with a higher estrogenic potency in transactivation assays compared with E2 [364]. In PCa, evidence for the ER-mediated effects of coumestrol is scarce. Nevertheless, the intake of phytoestrogens like coumestrol substantially reduced the PCa risk among men with specific polymorphic variation in the promoter region of the ERβ gene [618], which is suggestive of this receptor’s involvement.

6.4.13. Resveratrol

Resveratrol, also known as trans-3,40,5-trihydroxystilbene, is a stilbene polyphenol highly abundant in red grapes, peanuts, and other fruits [619]. Generally, red wine still comprises resveratrol in amounts ranging from 0.1 to 14.3 mg/L [620].
This natural agent is considered as a phytoestrogen due to its ability to bind ERs and GPER, competing with natural estrogens and altering the normal biological response including an anticarcinogenic effect [621,622,623]. Several studies have shown that resveratrol targets cancer cells without producing harmful effects on nonmalignant cells [48,49,624,625,626]. For this reason, it has been considered as an ideal molecule for anticancer therapy.
Resveratrol has obtained attention as a chemopreventive agent targeting multiple signaling pathways and affecting several cellular processes, namely cell viability, proliferation, apoptosis, survival, migration and invasion, and metabolism. A large number of studies have shown that resveratrol reduced the viability and proliferation of both androgen-sensitive and CRPC cells, namely, LNCaP, 22Rv1, DU145, and PC3 cells. [256,627,628,629,630,631,632]. It increased the number of cells in the G0/G1 arresting phase and diminished the number of cells in the G2 phase [630,631,632], likely by reducing the expression of cyclin D1, B1, and E and increasing the expression of some cell cycle inhibitors such as p21 and p27 [631,633]. Interestingly, resveratrol seems to reduce PC3 and 22Rv1 cell survival without affecting nonmalignant PNT1A cells [631]. In fact, the antiproliferative effect of resveratrol has been associated with its ability to reduce ERα and increase ERβ expression [634,635]. Other mechanisms associated with the effects of resveratrol in reducing PCa cell proliferation include the inhibition of the: (i) AR and CXCR4 pathway [625]; (ii) nuclear factor NFkβ, which is also associated with cell survival; and mTORC1 pathway, which promotes cancer cell growth, survival, invasion, and angiogenesis [636,637].
The induction of PCa cell apoptosis by resveratrol is dependent on modulating the expression of the pro- and antiapoptotic proteins (Figure 4). Resveratrol can upregulate the expression of proapoptotic genes BAX, BID, and BAK and downregulate the antiapoptotic ones, MCL-1, BCL-2, and BCL-XL, in several human and rodent PCa cell lines, namely, LNCaP, C4-2B, DU145, PC3, as well as TRAMP-C1, TRAMP-C2, and TRAMP-C3 [633,638,639,640,641]. The consequent alterations in the apoptosis-related proteins culminated with the increased activity of several caspase family members, namely, caspase-7 and caspase-3 [256,632,638,639,641,642]. Furthermore, resveratrol sensitized the LNCaP cells and PC3 xenografts to TRAIL-mediated apoptosis by increasing the expression of TRAIL receptors [643,644]. Other mechanisms involved in resveratrol-induced apoptosis include the increased expression of p53, the generation of ROS, the downregulation of the PI3K/Akt survival pathway, and autophagy induction [633,638,639,640,645]. Moreover, the activation of apoptotic targets was also achieved by the activation of FOXO transcription factors [643].
Concerning PCa progression, resveratrol was shown to control cell migration, invasion, and EMT. Resveratrol treatment reduced the migration and invasion of LNCaP, DU145, PC3, PC3M-MM2, and MAT-LyLu cells [627,646,647,648]. Moreover, this natural agent could revert EMT in LNCaP and PC3 cells by increasing the expression of the epithelial marker E-cadherin, and decreasing the expression of vimentin, a mesenchymal marker [649]. In vivo studies in TRAMP and SV-40 Tag rats confirmed the results obtained in cell lines and the valuable effects of resveratrol in suppressing PCa growth [476,635].
The effect of resveratrol extends to other features of cancer cells such as metabolic rewiring and the capacity to survive in hypoxic microenvironments. Resveratrol has been shown to inhibit HIFα expression, repressing the progression of LNCaP xenograft tumors [650]. Furthermore, in hypoxic LNCaP cells at low androgen levels (mimicking CRPC), resveratrol inhibited hypoxia-induced nuclear accumulation of β-catenin, inhibiting its mediated AR signaling [650]. Concerning the metabolic response, resveratrol decreased the uptake of glutamine and deregulated lipid metabolism in human (LNCaP, DU145, PC3, C4-2B) and murine (TRAMP and HMVp2) PCa cells as well as in LNCaP and HMVP2 xenografts [639,650,651], which delayed the progression of PCa.
Overall, due to all the above-mentioned beneficial properties of resveratrol, several approaches have been developed to test its therapeutic effectiveness, alone or in combined therapy, together with the optimal delivery method, using different coated nanoparticles targeting PCa cells [652,653,654,655,656,657]. Alginate nanoparticles containing resveratrol and curcumin exhibited cytotoxic effects on DU145 cells [652]. Also, resveratrol-loaded poly(lactic-co-glycolic acid) nanoparticles reduced the LNCaP cell viability, arrested cell cycle at the G1-S transition phase, enhanced ROS levels, and promoted apoptosis, causing the externalization of phosphatidylserine, DNA nicking, and the loss of mitochondrial membrane potential [657]. Other similar resveratrol-loaded nanoparticles based on a poly(epsilon-caprolactone) and poly(d,l-lactic-co-glycolic acid)–poly(ethylene glycol) blend also displayed cytotoxicity in LNCaP, DU145, and PC3 cells [653], and resveratrol-loaded poly(2-hydroxyethyl methacrylate)-chitosan-based nanoparticles also showed cytotoxicity in PC3 cells [658]. Planetary-ball-milled nanoparticles encapsulated with resveratrol in combination with docetaxel and conjugated with folic acid increased the apoptotic cell number in PC3 and docetaxel-resistant PC3 cells, reducing the expression of NF-kB p65, COX-2, and pro- (BAX, BAK) and antiapoptotic (BCL-2, BCL-XL) genes and downregulating survivin as well as increasing cleaved caspase-3 expression [654]. Moreover, functionalized mesoporous silica nanoparticles encapsulating resveratrol displayed antiproliferative effects and sensitized PCa cells to docetaxel in a hypoxic cell environment [655].
The combination of resveratrol with docetaxel was also tested using liposomes and showed antitumor efficiency against PCa, reducing proliferation and increasing apoptosis [659]. Resveratrol-loaded solid lipid nanoparticles were another approach tested in PCa cells as a potential carrier for delivery chemotherapeutics at the tumor site [660]. Other combinations, even without using nanocarriers, showed the enhanced anticarcinogenic effects of cisplatin, docetaxel, doxorubicin, and resveratrol in PCa cells [661,662,663].
Various reports have also shown that resveratrol enhances the response to ionizing radiation in LAPC4, DU145, and PC3 cells [631,664,665,666,667,668].
The results obtained with resveratrol in preclinical studies support testing this phytoestrogen in clinical trials concerning its safety, tolerability, and effective dose in PCa patients [39,41]. A pilot study assessing the effects of a phytotherapeutic intervention containing turmeric, resveratrol, green tea, and broccoli sprouts revealed that its use is feasible in men with biochemically recurrent PCa and a moderate PSA rise rate [41]. In a phase I study in nonmetastatic biochemically recurrent PCa patients, 4000 mg of a pulverized muscadine grape skin containing ellagic acid, quercetin, and resveratrol was safe in augmenting PSA doubling time, with no serious adverse effects being reported in the study time-frame [39]. This formulation was further investigated in a randomized, multicenter, placebo-controlled, dose-evaluating phase II trial. However, no significant difference in PSA doubling time was observed between the control and treatment groups [669].
Besides the beneficial effects of resveratrol and the promising results from preclinical studies and clinical trials, it remains crucial to better understand its detailed mechanism of action and the ideal dose as a therapeutic agent against PCa.

6.4.14. Diosgenin

Diosgenin, also known as 25R-spirost-5-en-3β-ol, is a steroidal sapogenin with a structure similar to cholesterol and other steroids, being used by the pharmaceutical industry in the synthesis of sex hormones and corticosteroids for application as anti-inflammatory, androgenic, and contraceptive drugs [670,671]. This natural compound is commonly found in a panoply of plants, namely, in species from the Dioscorea, Trigonella, Costus, and Smilax genera, particularly in tubers and seeds [672,673,674,675].
This steroidal sapogenin has been shown to have antimicrobial, antioxidant, anti-inflammatory, antidiabetic, and antiobesity activity [676]. Furthermore, diosgenin’s role in cancer is well-described. It has a potential antitumor effect in many types of cancer, namely, in leukemia [677,678,679,680] and gastric [681,682], lung [683,684,685], breast [686,687], liver [688,689,690], renal [691], colon [692,693,694], pancreas [695], bone [696], cervical [697], skin [698,699], and oral [700] cancers.
The anticancer effects of diosgenin have also been confirmed in PCa (Figure 4) [701,702,703]. This sapogenin reduced the PC3 cell viability in a dose-dependent manner [703]. Along the same line, diosgenin and a fenugreek extract containing diosgenin as the bioactive compound inhibited the proliferation of PCa cells [702,704], induced cell cycle arrest in the G2/M phase, and increased the sub-G1 and G2/M phase cell population [704,705].
Diosgenin also augmented apoptosis in DU145 and PC3 cells [702,704], underpinned by the increased expression of caspase-9 [702]. The fenugreek extract also reduced the expression of the mutant p53 protein in DU145 cells [705]. In addition, diosgenin can activate autophagy [702], a cell death process distinct from apoptosis that contributes to the maintenance of the stability of the intracellular environment and the normal physiological functions of the cell, which have been related to tumor growth inhibition [706,707,708]. DU145 cells treated with diosgenin showed a large number of autophagosomes including autophagosomes containing mitochondria [702].
The molecular mediators of diosgenin’s actions in suppressing cell survival and growth have been explored. In PC3 cells, diosgenin increased the expression of cell cycle inhibitor p21 and inhibited the phosphorylation of EGFR [705]. Furthermore, it was confirmed that diosgenin inhibits several signaling pathways, namely, the PI3K/Akt, JNK, and ERK pathways [701,702,709]. Exposure of PCa cells to diosgenin was shown to reduce PI3K, Akt, mTOR, JNK1/2 and ERK1/2 phosphorylation [701,702,705,709] by regulating the TGF-β and the HGF pathways.
Concerning tumor progression, the steroid sapogenin diosgenin reduced the migration and invasion capabilities of PC3 cells [701,704], inhibiting the expression and activation of MMP-2, MMP-7, and MMP-9, which have a crucial role in extracellular matrix degradation [701]. In DU145 cells, diosgenin inhibited HGF-induced motility, invasion, and EMT by increasing E-cadherin and decreasing vimentin and Mdm2 expression [709]. The role of diosgenin in inhibiting PCa tumor progression is also supported by its ability to suppress angiogenesis, as it blocked the expression of VEGF in PC3 cells and the tube formation of endothelial cells [701].
Few studies have investigated the anticancer activity of diosgenin in animal models. However, a study showed that diosgenin treatment diminished the prostate-to-body weight ratio and serum PSA levels in a rat model of BPH, established by the subcutaneous injection of testosterone propionate [710]. Reduced prostate size could be related to a proapoptotic effect of diosgenin, increasing Bax and p53 expression, and reducing the expression of Bcl-2 [710]. Furthermore, diosgenin-treated rats displayed an elevated activity of SOD and GPx and reduced the MDA levels [710], suggesting that diosgenin can mitigate oxidative stress in the hyperplastic prostate. Moreover, diosgenin reduced the tumor growth and metastasis in xenograft [711] and TRAMP mice by regulating NF-κB/STAT3 signaling [712].
Regarding the panoply of diosgenin effects against PCa, modulating many cellular processes, several studies evaluating the effect of diosgenin analogs and/or derivatives have emerged. Most of these compounds were revealed to have antiproliferative and proapoptotic activity in PCa, namely, in DU145 and PC3 cells [713,714,715,716,717], with some displaying advantages relative to diosgenin.

7. Lessons Learned, Conclusions, and Future Directions

Estrogens, the female sex hormones, have been shown to play a role in men’s physiology. The adult prostate is one of the male reproductive organs that is highly dependent on the actions of estrogens, which oscillate from anticarcinogenic, supporting the use of these hormones in therapy, to protumorigenic, accelerating PCa growth. However, the relationship between PCa risk and E2 serum levels remains controversial, with most studies not finding statistically significant associations. This lack of relationship can be a consequence of the broad range of serum E2 concentrations found in men, which is a result of a set of different variables not always controlled for in the different studies. E2 concentrations have been shown to vary with age group, ethnicity, and endocrine and metabolic conditions including adiposity and body mass index as well as with specificities in aromatase expression and activity. The intraprostatic production of estrogens cannot be ignored in the scenario of their influence on prostate physiology and carcinogenesis. External androgen precursors or circulating testosterone can be converted to estrogens locally in the prostate by aromatase. Indeed, it has been shown that increasing the aromatase activity with augmented E2 production accompanies the development of PCa (noncancerous prostate < primary tumors < metastatic tumors). Nevertheless, further research is needed to clarify the impact of intraprostatic estrogen biosynthesis in prostate pathology, as clinical trials using aromatase inhibitors have failed to confirm its utility [718,719]. Additionally, it would be reasonable to assess the effect of estrogen therapy in men with prostate cancer and low or high circulating estrogen levels in an analogy to breast cancer therapy in pre- and postmenopausal women.
As mentioned, and after the discontinuity of using estrogens in PCa treatment, research efforts over the years have produced a vast amount of information establishing the dual role of estrogens in prostate carcinogenesis, balancing from guardians to guilty. The enormous amount of data herein reviewed have detailed the biological processes and molecular events orchestrating the actions of estrogens as prostate carcinogens. Naturally occurring and synthetic hormones can augment cell proliferation, resistance to apoptosis, migration and invasion, and other events involved in tumor growth and progression. These procarcinogenic effects have mainly been related to the differential activation of ERα. In contrast, the role of estrogens in counteracting PCa development and metastization by reducing cell growth, promoting cell cycle arrest, increasing apoptosis, and suppressing migration, invasiveness, and angiogenesis has mainly implicated ERβ as the mediator. Nevertheless, the complexity of the actions of estrogens is far from being enlightened. For example, it remains to be disclosed whether differential responses in human patients may rely on the physiological condition, which may disrupt ERα and ERβ expression, changing prostate cells to a cancer-like phenotype. This hypothesis is gaining strength considering the reports showing that ER expression is altered in the prostate of diabetic [720] and obese [721] patients. Of note, diabetes markedly reduced ERβ and GPER expression in PCa patients [720].
More research is also needed to clarify the role of GPER in PCa, as the last decades have witnessed the emergence of reports showing that it can trigger both pro- and anticancer responses. It will also be of paramount importance to further investigate the relationship between GPER, aromatase expression, and intraprostatic estrogen production.
Another relevant issue in the landscape of prostate responses to estrogens is related to the fact that different concentrations/doses of estrogens can trigger distinct and sometimes opposing effects. Generally, data support that lower concentrations/doses are associated with the procarcinogenic effects of these hormones, with the higher doses being antitumorigenic. This was the basis of estrogen therapy in PCa, with the high hormone doses causing well-characterized adverse effects. A great pharmacological challenge in PCa therapy relies on developing new approaches allowing for the use of reduced doses of estrogens, receptor-specific agonists, or alternative estrogen-like compounds with similar properties to estrogens, but fewer side effects. In recent years, many studies have investigated the effect of natural compounds with estrogen-like activity in controlling PCa. This review summarized the promising properties of a panoply of phytoestrogens against PCa, reported both in vitro and in vivo. Overall, distinct substances of this class of compounds have been shown to control PCa cell proliferation and tumor growth, reduce migration and invasion, suppress EMT, angiogenesis, and metastization, and also regulate the bioenergetic metabolism of PCa cells. Several molecular targets have been discovered alongside identifying the actions of phytoestrogens and disclosing their mechanisms of action. These effects encompass the reduction in cyclins and CDK expression, the increased levels of p53 and decrease in those of p21, the activation of TRAIL receptors and caspase cascades, the reduction in β-catenin and MMP expression, and the inhibition of the Akt/PI3K and NFκB pathways, while activating MAPK signaling. Based on their tumor-suppressor properties, phytoestrogens such as chrysin, daidzein, genistein, quercetin, coumestrol, resveratrol, and equol have been pointed out as possible alternative therapies for PCa, and have been tested on humans. Equol, quercetin, coumestrol, and resveratrol are among the most promising agents as they have been shown to reduce the PSA levels and lower PCa risk and incidence when used in the diet. It is also quite challenging to demonstrate that quercetin can promote the recruitment of dendritic cells to the tumor site, which opens up new perspectives of research for evaluating phytoestrogens as anticancer molecules.
In summary, the discussion in the present review systematized the role of estrogens in prostate carcinogenesis and provided the scientific fundamentals for the evolution of PCa treatment based on the use of natural estrogenic substances. Using natural bioactive compounds such as phytoestrogens in PCa therapy is an exciting and powerful research and clinical question. Even if these compounds are not chosen as a first-line therapy, they can be combined with conventional treatments such as chemotherapy, radiation therapy, and immunotherapy, increasing their efficacy and/or mitigating the systemic adverse effects. Ongoing research efforts and others yet to come will expand the treatment options available for PCa patients.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biomedicines12081636/s1, Table S1: Anti-proliferative and pro-apoptotic effects of estrogens in prostate cells; Table S2: Phytoestrogens with a role in PCa.

Author Contributions

M.I.F., T.M.A.C., and J.M.-M. researched the data for the article. All authors contributed substantially to the discussion of the content. M.I.F., T.M.A.C., J.M.-M., and S.S. wrote the article. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by FEDER funds through the POCI—COMPETE 2020—Operational Program Competitiveness and Internationalization in Axis I-Strengthening research, technological development, and innovation (Project Nos. 007491 and 029114) and National Funds by FCT-Foundation for Science and Technology/MCTES (CICS-UBI projects UIDB/00709/2020 and UIDP/00709/2020). Cardoso H.J. and Figueira M.I. were recipients of FCT fellowships SFRH/BD/111351/2015 and SFRH/BD/104671/2014, respectively.

Conflicts of Interest

All authors declare no competing interests.

References

  1. Hamilton, K.J.; Hewitt, S.C.; Arao, Y.; Korach, K.S. Estrogen Hormone Biology. Curr. Top. Dev. Biol. 2017, 125, 109–146. [Google Scholar] [CrossRef] [PubMed]
  2. Schulster, M.; Bernie, A.M.; Ramasamy, R. The role of estradiol in male reproductive function. Asian J. Androl. 2016, 18, 435–440. [Google Scholar] [CrossRef] [PubMed]
  3. Cauley, J.A. Estrogen and bone health in men and women. Steroids 2015, 99, 11–15. [Google Scholar] [CrossRef] [PubMed]
  4. Fuentes, N.; Silveyra, P. Estrogen receptor signaling mechanisms. Adv. Protein Chem. Struct. Biol. 2019, 116, 135–170. [Google Scholar] [CrossRef] [PubMed]
  5. Miller, W.L.; Auchus, R.J. The Molecular Biology, Biochemistry, and Physiology of Human Steroidogenesis and Its Disorders. Endocr. Rev. 2011, 32, 81–151. [Google Scholar] [CrossRef] [PubMed]
  6. Huggins, C.; Hodges, C.V. Studies on prostatic cancer. I. The effect of castration, of estrogen and androgen injection on serum phosphatases in metastatic carcinoma of the prostate. CA A Cancer J. Clin. 1972, 22, 232–240. [Google Scholar] [CrossRef] [PubMed]
  7. Huggins, C.; Hodges, C.V. The Effect of Castration, of Estrogen and of Androgen Injection on Serum Phosphatases in Metastatic Carcinoma of the Prostate. Cancer Res. 1941, 1, 293–297. [Google Scholar]
  8. Group, V.A.C.-o.U.R. Treatment and survival of patients with cancer of the prostate. Surg. Gynecol. Obstet. 1967, 124, 1011–1017. [Google Scholar]
  9. Bailar, J.C., III; Byar, D.P.; Veterans Administration Cooperative Urological Research Group. Estrogen treatment for cancer of the prostate. Early results with 3 doses of diethylstilbestrol and placebo. Cancer 1970, 26, 257–261. [Google Scholar] [CrossRef]
  10. Byar, D.P. The Veterans Administration Cooperative Urological Research Group’s studies of cancer of the prostate. Cancer 1973, 32, 1126–1130. [Google Scholar] [CrossRef]
  11. Denmeade, S.R.; Isaacs, J.T. A history of prostate cancer treatment. Nat. Rev. Cancer 2002, 2, 389–396. [Google Scholar] [CrossRef] [PubMed]
  12. Oh, W.K. The evolving role of estrogen therapy in prostate cancer. Clin. Prostate Cancer 2002, 1, 81–89. [Google Scholar] [CrossRef] [PubMed]
  13. Yu, S.; Zhang, Y.; Yuen, M.T.; Zou, C.; Danielpour, D.; Chan, F.L. 17-Beta-estradiol induces neoplastic transformation in prostatic epithelial cells. Cancer Lett. 2011, 304, 8–20. [Google Scholar] [CrossRef] [PubMed]
  14. Nakajima, Y.; Osakabe, A.; Waku, T.; Suzuki, T.; Akaogi, K.; Fujimura, T.; Homma, Y.; Inoue, S.; Yanagisawa, J. Estrogen Exhibits a Biphasic Effect on Prostate Tumor Growth through the Estrogen Receptor beta-KLF5 Pathway. Mol. Cell. Biol. 2016, 36, 144–156. [Google Scholar] [CrossRef] [PubMed]
  15. Mishra, S.; Tai, Q.; Gu, X.; Schmitz, J.; Poullard, A.; Fajardo, R.J.; Mahalingam, D.; Chen, X.; Zhu, X.; Sun, L.Z. Estrogen and estrogen receptor alpha promotes malignancy and osteoblastic tumorigenesis in prostate cancer. Oncotarget 2015, 6, 44388–44402. [Google Scholar] [CrossRef] [PubMed]
  16. Ramírez-de-Arellano, A.; Pereira-Suárez, A.L.; Rico-Fuentes, C.; López-Pulido, E.I.; Villegas-Pineda, J.C.; Sierra-Diaz, E. Distribution and Effects of Estrogen Receptors in Prostate Cancer: Associated Molecular Mechanisms. Front. Endocrinol. 2021, 12, 811578. [Google Scholar] [CrossRef] [PubMed]
  17. Qu, L.G.; Wardan, H.; Davis, I.D.; Pezaro, C.; Sluka, P. Effects of estrogen receptor signaling on prostate cancer carcinogenesis. Transl. Res. 2020, 222, 56–66. [Google Scholar] [CrossRef] [PubMed]
  18. Figueira, M.I.; Cardoso, H.J.; Socorro, S. The Role of GPER Signaling in Carcinogenesis: A Focus on Prostate Cancer. In Recent Trends in Cancer Biology Spotlight on Signaling Cascades and microRNAs Cell Signaling Pathways and microRNAs in Cancer Biology; Fayyaz, S., Farooqi, A.A., Eds.; Springer International Publishing: Cham, Switzerland, 2018; pp. 59–117. [Google Scholar]
  19. Jensen, E.V.; DeSombre, E.R. Estrogen-receptor interaction. Science 1973, 182, 126–134. [Google Scholar] [CrossRef] [PubMed]
  20. Filardo, E.J.; Quinn, J.A.; Bland, K.I.; Frackelton, A.R., Jr. Estrogen-induced activation of Erk-1 and Erk-2 requires the G protein-coupled receptor homolog, GPR30, and occurs via trans-activation of the epidermal growth factor receptor through release of HB-EGF. Mol. Endocrinol. 2000, 14, 1649–1660. [Google Scholar] [CrossRef] [PubMed]
  21. Kuiper, G.; Enmark, E.; Pelto-Huikko, M.; Nilsson, S.; Gustafsson, J.-A. Cloning of a novel receptor expressed in rat prostate and ovary. Proc. Natl. Acad. Sci. USA 1996, 93, 5925–5930. [Google Scholar] [CrossRef]
  22. Marino, M.; Galluzzo, P.; Ascenzi, P. Estrogen signaling multiple pathways to impact gene transcription. Curr. Genom. 2006, 7, 497–508. [Google Scholar] [CrossRef] [PubMed]
  23. O’Malley, B.W. A life-long search for the molecular pathways of steroid hormone action. Mol. Endocrinol. 2005, 19, 1402–1411. [Google Scholar] [CrossRef] [PubMed]
  24. Göttlicher, M.; Heck, S.; Herrlich, P. Transcriptional cross-talk, the second mode of steroid hormone receptor action. J. Mol. Med. 1998, 76, 480–489. [Google Scholar] [CrossRef] [PubMed]
  25. Aranda, A.; Pascual, A. Nuclear hormone receptors and gene expression. Physiol. Rev. 2001, 81, 1269–1304. [Google Scholar] [CrossRef] [PubMed]
  26. Lösel, R.; Wehling, M. Nongenomic actions of steroid hormones. Nat. Rev. Mol. Cell Biol. 2003, 4, 46–56. [Google Scholar] [CrossRef] [PubMed]
  27. Ellem, S.J.; Risbridger, G.P. The dual, opposing roles of estrogen in the prostate. Ann. N. Y. Acad. Sci. 2009, 1155, 174–186. [Google Scholar] [CrossRef] [PubMed]
  28. Risbridger, G.P.; Ellem, S.J.; McPherson, S.J. Estrogen action on the prostate gland: A critical mix of endocrine and paracrine signaling. J. Mol. Endocrinol. 2007, 39, 183–188. [Google Scholar] [CrossRef] [PubMed]
  29. Attia, D.M.; Ederveen, A.G. Opposing roles of ERα and ERβ in the genesis and progression of adenocarcinoma in the rat ventral prostate. Prostate 2012, 72, 1013–1022. [Google Scholar] [CrossRef]
  30. Souza, D.S.; Macheroni, C.; Vicente, C.M.; Cavalheiro, R.P.; Campo, V.L.; Porto, C.S. Estrogen receptors regulate galectin-3 in androgen-independent DU-145 prostate cancer cells. Oncol. Rep. 2023, 49, 8530. [Google Scholar] [CrossRef]
  31. Xiao, L.; Luo, Y.; Tai, R.; Zhang, N. Estrogen receptor β suppresses inflammation and the progression of prostate cancer. Mol. Med. Rep. 2019, 19, 3555–3563. [Google Scholar] [CrossRef]
  32. Moorthy, H.K.; Laxman Prabhu, G.G.; Venugopal, P. The resurgence of estrogens in the treatment of castration-resistant prostate cancer. Indian J. Urol. IJU J. Urol. Soc. India 2019, 35, 189–196. [Google Scholar] [CrossRef]
  33. Kimura, T.; Egawa, S. Epidemiology of prostate cancer in Asian countries. Int. J. Urol. 2018, 25, 524–531. [Google Scholar] [CrossRef] [PubMed]
  34. Loeb, S.; Fu, B.C.; Bauer, S.R.; Pernar, C.H.; Chan, J.M.; Van Blarigan, E.L.; Giovannucci, E.L.; Kenfield, S.A.; Mucci, L.A. Association of plant-based diet index with prostate cancer risk. Am. J. Clin. Nutr. 2022, 115, 662–670. [Google Scholar] [CrossRef] [PubMed]
  35. He, J.; Wang, S.; Zhou, M.; Yu, W.; Zhang, Y.; He, X. Phytoestrogens and risk of prostate cancer: A meta-analysis of observational studies. World J. Surg. Oncol. 2015, 13, 231. [Google Scholar] [CrossRef]
  36. Sirotkin, A.V.; Harrath, A.H. Phytoestrogens and their effects. Eur. J. Pharmacol. 2014, 741, 230–236. [Google Scholar] [CrossRef] [PubMed]
  37. Erdogan, S.; Doganlar, O.; Doganlar, Z.B.; Serttas, R.; Turkekul, K.; Dibirdik, I.; Bilir, A. The flavonoid apigenin reduces prostate cancer CD44+ stem cell survival and migration through PI3K/Akt/NF-κB signaling. Life Sci. 2016, 162, 77–86. [Google Scholar] [CrossRef]
  38. Erdogan, S.; Turkekul, K.; Serttas, R.; Erdogan, Z. The natural flavonoid apigenin sensitizes human CD44+ prostate cancer stem cells to cisplatin therapy. Biomed. Pharmacother. 2017, 88, 210–217. [Google Scholar] [CrossRef]
  39. Paller, C.J.; Rudek, M.A.; Zhou, X.C.; Wagner, W.D.; Hudson, T.S.; Anders, N.; Hammers, H.J.; Dowling, D.; King, S.; Antonarakis, E.S. A phase I study of muscadine grape skin extract in men with biochemically recurrent prostate cancer: Safety, tolerability, and dose determination. Prostate 2015, 75, 1518–1525. [Google Scholar] [CrossRef] [PubMed]
  40. Paller, C.J.; Kanaan, Y.M.; Beyene, D.A.; Naab, T.J.; Copeland, R.L.; Tsai, H.L.; Kanarek, N.F.; Hudson, T.S. Risk of prostate cancer in African-American men: Evidence of mixed effects of dietary quercetin by serum vitamin D status. Prostate 2015, 75, 1376–1383. [Google Scholar] [CrossRef]
  41. van Die, M.D.; Williams, S.G.; Emery, J.; Bone, K.M.; Taylor, J.M.; Lusk, E.; Pirotta, M.V. A Placebo-Controlled Double-Blinded Randomized Pilot Study of Combination Phytotherapy in Biochemically Recurrent Prostate Cancer. Prostate 2017, 77, 765–775. [Google Scholar] [CrossRef]
  42. Singh, C.K.; Chhabra, G.; Ndiaye, M.A.; Siddiqui, I.A.; Panackal, J.E.; Mintie, C.A.; Ahmad, N. Quercetin-Resveratrol Combination for Prostate Cancer Management in TRAMP Mice. Cancers 2020, 12, 2141. [Google Scholar] [CrossRef] [PubMed]
  43. Sharma, N.; Raut, P.W.; Baruah, M.M.; Sharma, A. Combination of quercetin and 2-methoxyestradiol inhibits epithelial-mesenchymal transition in PC-3 cell line via Wnt signaling pathway. Future Sci. OA 2021, 7, Fso747. [Google Scholar] [CrossRef] [PubMed]
  44. Shu, Y.; Xie, B.; Liang, Z.; Chen, J. Quercetin reverses the doxorubicin resistance of prostate cancer cells by downregulating the expression of c-met. Oncol. Lett. 2018, 15, 2252–2258. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, P.; Henning, S.M.; Heber, D.; Vadgama, J.V. Sensitization to docetaxel in prostate cancer cells by green tea and quercetin. J. Nutr. Biochem. 2015, 26, 408–415. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, P.; Henning, S.M.; Magyar, C.E.; Elshimali, Y.; Heber, D.; Vadgama, J.V. Green tea and quercetin sensitize PC-3 xenograft prostate tumors to docetaxel chemotherapy. J. Exp. Clin. Cancer Res. 2016, 35, 73. [Google Scholar] [CrossRef] [PubMed]
  47. Tummala, R.; Lou, W.; Gao, A.C.; Nadiminty, N. Quercetin targets hnRNPA1 to overcome enzalutamide resistance in prostate cancer cells. Mol. Cancer Ther. 2017, 16, 2770–2779. [Google Scholar] [CrossRef] [PubMed]
  48. Marcsek, Z.; Kocsis, Z.; Szende, B.; Tompa, A. Effect of formaldehyde and resveratrol on the viability of Vero, HepG2 and MCF-7 cells. Cell Biol. Int. 2007, 31, 1214–1219. [Google Scholar] [CrossRef]
  49. Reagan-Shaw, S.; Mukhtar, H.; Ahmad, N. Resveratrol imparts photoprotection of normal cells and enhances the efficacy of radiation therapy in cancer cells. Photochem. Photobiol. 2008, 84, 415–421. [Google Scholar] [CrossRef]
  50. Gido, S.; Jenny, A.V.; Wiebke, A.; Johannes, H. Circulating steroid hormone variations throughout different stages of prostate cancer. Endocr.-Relat. Cancer 2017, 24, R403–R420. [Google Scholar] [CrossRef]
  51. Wang, Q.; Rangiah, K.; Mesaros, C.; Snyder, N.W.; Vachani, A.; Song, H.; Blair, I.A. Ultrasensitive quantification of serum estrogens in postmenopausal women and older men by liquid chromatography-tandem mass spectrometry. Steroids 2015, 96, 140–152. [Google Scholar] [CrossRef]
  52. MacKintosh, F.R.; Sprenkle, P.C.; Walter, L.C.; Rawson, L.; Karnes, R.J.; Morrell, C.H.; Kattan, M.W.; Nawaf, C.B.; Neville, T.B. Age and Prostate-Specific Antigen Level Prior to Diagnosis Predict Risk of Death from Prostate Cancer. Front. Oncol. 2016, 6, 157. [Google Scholar] [CrossRef] [PubMed]
  53. Ho, S.M.; Lee, M.T.; Lam, H.M.; Leung, Y.K. Estrogens and prostate cancer: Etiology, mediators, prevention, and management. Endocrinol. Metab. Clin. North Am. 2011, 40, 591–614. [Google Scholar] [CrossRef] [PubMed]
  54. Neuzillet, Y.; Raynaud, J.P.; Radulescu, C.; Fiet, J.; Giton, F.; Dreyfus, J.F.; Ghoneim, T.P.; Lebret, T.; Botto, H. Sexual steroids in serum and prostatic tissue of human non-cancerous prostate (STERPROSER trial). Prostate 2017, 77, 1512–1519. [Google Scholar] [CrossRef] [PubMed]
  55. Ahluwalia, B.; Jackson, M.A.; Jones, G.W.; Williams, A.O.; Rao, M.S.; Rajguru, S. Blood hormone profiles in prostate cancer patients in high-risk and low-risk populations. Cancer 1981, 48, 2267–2273. [Google Scholar] [CrossRef] [PubMed]
  56. Chadid, S.; Barber, J.R.; Nelson, W.G.; Gurel, B.; Lucia, M.S.; Thompson, I.M.; Goodman, P.J.; Stanczyk, F.Z.; Parnes, H.L.; Lippman, S.M.; et al. The association between serum sex steroid hormone concentrations and intraprostatic inflammation in men without prostate cancer and irrespective of clinical indication for biopsy in the placebo arm of the Prostate Cancer Prevention Trial. Prostate 2020, 80, 895–905. [Google Scholar] [CrossRef] [PubMed]
  57. Yao, S.; Till, C.; Kristal, A.R.; Goodman, P.J.; Hsing, A.W.; Tangen, C.M.; Platz, E.A.; Stanczyk, F.Z.; Reichardt, J.K.; Tang, L.; et al. Serum estrogen levels and prostate cancer risk in the prostate cancer prevention trial: A nested case-control study. Cancer Causes Control CCC 2011, 22, 1121–1131. [Google Scholar] [CrossRef] [PubMed]
  58. Mohr, B.A.; Feldman, H.A.; Kalish, L.A.; Longcope, C.; McKinlay, J.B. Are serum hormones associated with the risk of prostate cancer? Prospective results from the Massachusetts Male Aging Study. Urology 2001, 57, 930–935. [Google Scholar] [CrossRef] [PubMed]
  59. Dorgan, J.F.; Albanes, D.; Virtamo, J.; Heinonen, O.P.; Chandler, D.W.; Galmarini, M.; McShane, L.M.; Barrett, M.J.; Tangrea, J.; Taylor, P.R. Relationships of serum androgens and estrogens to prostate cancer risk: Results from a prospective study in Finland. Cancer Epidemiol. Biomark. Prev. A Publ. Am. Assoc. Cancer Res. Cosponsored By Am. Soc. Prev. Oncol. 1998, 7, 1069–1074. [Google Scholar]
  60. Meunier, M.E.; Neuzillet, Y.; Raynaud, J.P.; Radulescu, C.; Ghoneim, T.; Fiet, J.; Giton, F.; Rouanne, M.; Dreyfus, J.F.; Lebret, T.; et al. Sex steroids in serum and prostatic tissue of human cancerous prostate (STERKPROSER trial). Prostate 2019, 79, 272–280. [Google Scholar] [CrossRef]
  61. Grosman, H.; Fabre, B.; Mesch, V.; Lopez, M.A.; Schreier, L.; Mazza, O.; Berg, G. Lipoproteins, sex hormones and inflammatory markers in association with prostate cancer. Aging Male Off. J. Int. Soc. Study Aging Male 2010, 13, 87–92. [Google Scholar] [CrossRef]
  62. Usoro, A.J.; Obot, A.S.; Ekaidem, I.S.; Akaiso, O.E.; Udoh, A.E.; Akinloye, O. Serum Testosterone, 17beta-Estradiol and PSA Levels in Subjects with Prostate Disorders. Indian J. Clin. Biochem. IJCB 2015, 30, 59–65. [Google Scholar] [CrossRef] [PubMed]
  63. Barrett-Connor, E.; Garland, C.; McPhillips, J.B.; Khaw, K.-T.; Wingard, D.L. A Prospective, Population-based Study of Androstenedione, Estrogens, and Prostatic Cancer. Cancer Res. 1990, 50, 169–173. [Google Scholar]
  64. Belanger, A.; Candas, B.; Dupont, A.; Cusan, L.; Diamond, P.; Gomez, J.L.; Labrie, F. Changes in serum concentrations of conjugated and unconjugated steroids in 40- to 80-year-old men. J. Clin. Endocrinol. Metab. 1994, 79, 1086–1090. [Google Scholar] [CrossRef] [PubMed]
  65. Cao, J.; Chen, T.M.; Hao, W.J.; Li, J.; Liu, L.; Zhu, B.P.; Li, X.Y. Correlation between sex hormone levels and obesity in the elderly male. Aging Male Off. J. Int. Soc. Study Aging Male 2012, 15, 85–89. [Google Scholar] [CrossRef] [PubMed]
  66. Chen, C.; Weiss, N.S.; Stanczyk, F.Z.; Lewis, S.K.; DiTommaso, D.; Etzioni, R.; Barnett, M.J.; Goodman, G.E. Endogenous sex hormones and prostate cancer risk: A case-control study nested within the Carotene and Retinol Efficacy Trial. Cancer Epidemiol. Biomark. Prev. A Publ. Am. Assoc. Cancer Res. Cosponsored By Am. Soc. Prev. Oncol. 2003, 12, 1410–1416. [Google Scholar]
  67. Roddam, A.W.; Allen, N.E.; Appleby, P.; Key, T.J. Endogenous sex hormones and prostate cancer: A collaborative analysis of 18 prospective studies. J. Natl. Cancer Inst. 2008, 100, 170–183. [Google Scholar] [CrossRef] [PubMed]
  68. Gann, P.H.; Hennekens, C.H.; Ma, J.; Longcope, C.; Stampfer, M.J. Prospective study of sex hormone levels and risk of prostate cancer. J. Natl. Cancer Inst. 1996, 88, 1118–1126. [Google Scholar] [CrossRef]
  69. Hagiuda, J.; Ishikawa, H.; Marumo, K. Serum oestradiol levels in male partners of infertile couples. Andrologia 2015, 47, 669–673. [Google Scholar] [CrossRef]
  70. Hammond, G.L.; Kontturi, M.; Vihko, P.; Vihko, R. Serum steroids in normal males and patients with prostatic diseases. Clin. Endocrinol. 1978, 9, 113–121. [Google Scholar] [CrossRef]
  71. Hsing, A.W.; Comstock, G.W. Serological precursors of cancer: Serum hormones and risk of subsequent prostate cancer. Cancer Epidemiol. Biomark. Prev. A Publ. Am. Assoc. Cancer Res. Cosponsored By Am. Soc. Prev. Oncol. 1993, 2, 27–32. [Google Scholar]
  72. Nomura, A.; Heilbrun, L.K.; Stemmermann, G.N.; Judd, H.L. Prediagnostic serum hormones and the risk of prostate cancer. Cancer Res. 1988, 48, 3515–3517. [Google Scholar] [CrossRef] [PubMed]
  73. Pellitero, S.; Olaizola, I.; Alastrue, A.; Martinez, E.; Granada, M.L.; Balibrea, J.M.; Moreno, P.; Serra, A.; Navarro-Diaz, M.; Romero, R.; et al. Hypogonadotropic hypogonadism in morbidly obese males is reversed after bariatric surgery. Obes. Surg. 2012, 22, 1835–1842. [Google Scholar] [CrossRef] [PubMed]
  74. Platz, E.A.; Leitzmann, M.F.; Rifai, N.; Kantoff, P.W.; Chen, Y.C.; Stampfer, M.J.; Willett, W.C.; Giovannucci, E. Sex steroid hormones and the androgen receptor gene CAG repeat and subsequent risk of prostate cancer in the prostate-specific antigen era. Cancer Epidemiol. Biomark. Prev. A Publ. Am. Assoc. Cancer Res. Cosponsored By Am. Soc. Prev. Oncol. 2005, 14, 1262–1269. [Google Scholar] [CrossRef] [PubMed]
  75. Salonia, A.; Abdollah, F.; Capitanio, U.; Suardi, N.; Gallina, A.; Castagna, G.; Clementi, M.C.; Briganti, A.; Rigatti, P.; Montorsi, F. Circulating sex steroids and prostate cancer: Introducing the time-dependency theory. World J. Urol. 2013, 31, 267–273. [Google Scholar] [CrossRef] [PubMed]
  76. Schnoeller, T.J.; Steinestel, J.; Zengerling, F.; Schrader, A.J.; Jentzmik, F. Serum 17beta-estradiol fails as a marker in identification of aggressive tumour disease in patients with localized prostate cancer. World J. Urol. 2015, 33, 1979–1984. [Google Scholar] [CrossRef] [PubMed]
  77. Severi, G.; Morris, H.A.; MacInnis, R.J.; English, D.R.; Tilley, W.; Hopper, J.L.; Boyle, P.; Giles, G.G. Circulating steroid hormones and the risk of prostate cancer. Cancer Epidemiol. Biomark. Prev. A Publ. Am. Assoc. Cancer Res. Cosponsored By Am. Soc. Prev. Oncol. 2006, 15, 86–91. [Google Scholar] [CrossRef] [PubMed]
  78. Vermeulen, A.; Kaufman, J.M.; Deslypere, J.P.; Thomas, G. Attenuated luteinizing hormone (LH) pulse amplitude but normal LH pulse frequency, and its relation to plasma androgens in hypogonadism of obese men. J. Clin. Endocrinol. Metab. 1993, 76, 1140–1146. [Google Scholar] [CrossRef] [PubMed]
  79. Zumoff, B.; Strain, G.W.; Kream, J.; O’Connor, J.; Levin, J.; Fukushima, D.K. Obese young men have elevated plasma estrogen levels but obese premenopausal women do not. Metab. Clin. Exp. 1981, 30, 1011–1014. [Google Scholar] [CrossRef] [PubMed]
  80. Simpson, E.R.; Davis, S.R. Minireview: Aromatase and the Regulation of Estrogen Biosynthesis—Some New Perspectives. Endocrinology 2001, 142, 4589–4594. [Google Scholar] [CrossRef] [PubMed]
  81. Cooke, P.S.; Nanjappa, M.K.; Ko, C.; Prins, G.S.; Hess, R.A. Estrogens in Male Physiology. Physiol. Rev. 2017, 97, 995–1043. [Google Scholar] [CrossRef]
  82. Miller, W.L. Molecular Biology of Steroid Hormone Synthesis*. Endocr. Rev. 1988, 9, 295–318. [Google Scholar] [CrossRef] [PubMed]
  83. Krolik, M.; Milnerowicz, H. The effect of using estrogens in the light of scientific research. Adv. Clin. Exp. Med. Off. Organ Wroc. Med. Univ. 2012, 21, 535–543. [Google Scholar]
  84. Farkas, S.; Szabó, A.; Hegyi, A.E.; Török, B.; Fazekas, C.L.; Ernszt, D.; Kovács, T.; Zelena, D. Estradiol and Estrogen-like Alternative Therapies in Use: The Importance of the Selective and Non-Classical Actions. Biomedicines 2022, 10, 861. [Google Scholar] [CrossRef] [PubMed]
  85. Williams, G. Aromatase up-regulation, insulin and raised intracellular oestrogens in men, induce adiposity, metabolic syndrome and prostate disease, via aberrant ER-alpha and GPER signalling. Mol. Cell. Endocrinol. 2012, 351, 269–278. [Google Scholar] [CrossRef] [PubMed]
  86. Matzkin, H.; Soloway, M.S. Immunohistochemical evidence of the existence and localization of aromatase in human prostatic tissues. Prostate 1992, 21, 309–314. [Google Scholar] [CrossRef]
  87. Takase, Y.; Lévesque, M.-H.; Luu-The, V.; El-Alfy, M.; Labrie, F.; Pelletier, G. Expression of Enzymes Involved in Estrogen Metabolism in Human Prostate. J. Histochem. Cytochem. 2006, 54, 911–921. [Google Scholar] [CrossRef] [PubMed]
  88. Ellem, S.J.; Risbridger, G.P. Aromatase and regulating the estrogen:androgen ratio in the prostate gland. J. Steroid Biochem. Mol. Biol. 2010, 118, 246–251. [Google Scholar] [CrossRef] [PubMed]
  89. Machioka, K.; Mizokami, A.; Yamaguchi, Y.; Izumi, K.; Hayashi, S.; Namiki, M. Active estrogen synthesis and its function in prostate cancer-derived stromal cells. Anticancer Res. 2015, 35, 221–227. [Google Scholar] [PubMed]
  90. Prossnitz, E.R.; Barton, M. Signaling, physiological functions and clinical relevance of the G protein-coupled estrogen receptor GPER. Prostaglandins Other Lipid Mediat. 2009, 89, 89–97. [Google Scholar] [CrossRef]
  91. Takizawa, I.; Lawrence, M.G.; Balanathan, P.; Rebello, R.; Pearson, H.B.; Garg, E.; Pedersen, J.; Pouliot, N.; Nadon, R.; Watt, M.J.; et al. Estrogen receptor alpha drives proliferation in PTEN-deficient prostate carcinoma by stimulating survival signaling, MYC expression and altering glucose sensitivity. Oncotarget 2015, 6, 604–616. [Google Scholar] [CrossRef]
  92. Morais-Santos, M.; Werneck-Gomes, H.; Campolina-Silva, G.H.; Santos, L.C.; Mahecha, G.A.B.; Hess, R.A.; Oliveira, C.A. Basal Cells Show Increased Expression of Aromatase and Estrogen Receptor alpha in Prostate Epithelial Lesions of Male Aging Rats. Endocrinology 2018, 159, 723–732. [Google Scholar] [CrossRef] [PubMed]
  93. Cheboub, A.; Regouat, N.; Djidjik, R.; Slimani, A.; Hadj-Bekkouche, F. Short-term aromatase inhibition induces prostatic alterations in adult wistar rat: A biochemical, histopathological and immunohistochemical study. Acta Histochem. 2019, 121, 151441. [Google Scholar] [CrossRef] [PubMed]
  94. Montgomery, R.B.; Mostaghel, E.A.; Vessella, R.; Hess, D.L.; Kalhorn, T.F.; Higano, C.S.; True, L.D.; Nelson, P.S. Maintenance of intratumoral androgens in metastatic prostate cancer: A mechanism for castration-resistant tumor growth. Cancer Res. 2008, 68, 4447–4454. [Google Scholar] [CrossRef] [PubMed]
  95. Byar, D.P.; Corle, D.K. Hormone therapy for prostate cancer: Results of the Veterans Administration Cooperative Urological Research Group studies. NCI Monogr. A Publ. Natl. Cancer Inst. 1988, 7, 165–170. [Google Scholar] [CrossRef]
  96. Lyrenäs, S.; Carlström, K.; Bäckström, T.; Von Schoultz, B. A comparison of serum oestrogen levels after percutaneous and oral administration of oestradiol-17β. BJOG Int. J. Obstet. Gynaecol. 1981, 88, 181–187. [Google Scholar] [CrossRef]
  97. de Liguieres, B.; Basdevant, A. Differential metabolic tolerance between oral and percutaneous administration of estradiol in postmenopausal women. Osteoporosis 1987, 2, 1120–1131. [Google Scholar]
  98. Bosset, P.-O.; Albiges, L.; Seisen, T.; de la Motte Rouge, T.; Phé, V.; Bitker, M.-O.; Rouprêt, M. Current role of diethylstilbestrol in the management of advanced prostate cancer. BJU Int. 2012, 110, E826–E829. [Google Scholar] [CrossRef] [PubMed]
  99. Takezawa, Y.; Nakata, S.; Kobayashi, M.; Kosaku, N.; Fukabori, Y.; Yamanaka, H. Moderate dose diethylstilbestrol diphosphate therapy in hormone refractory prostate cancer. Scand. J. Urol. Nephrol. 2001, 35, 283–287. [Google Scholar] [PubMed]
  100. de Voogt, H.J.; Smith, P.H.; Pavone-Macaluso, M.; de Pauw, M.; Suciu, S.; Members of the European Organization for Research on Treatment of Cancer Urological Group. Cardiovascular side effects of diethylstilbestrol, cyproterone acetate, medroxyprogesterone acetate and estramustine phosphate used for the treatment of advanced prostatic cancer: Results from European Organization for Research on Treatment of Cancer trials 30761 and 30762. J. Urol. 1986, 135, 303–307. [Google Scholar]
  101. von Schoultz, B.; Carlström, K.; Collste, L.; Eriksson, A.; Henriksson, P.; Pousette, Å.; Stege, R. Estrogen therapy and liver function—Metabolic effects of oral and parenteral administration. Prostate 1989, 14, 389–395. [Google Scholar] [CrossRef]
  102. Henriksson, P.; Blombäck, M.; Eriksson, A.; Stege, R.; Carlström, K. Effect of parenteral oestrogen on the coagulation system in patients with prostatic carcinoma. Br. J. Urol. 1990, 65, 282–285. [Google Scholar] [CrossRef] [PubMed]
  103. Hedlund, P.O.; Henriksson, P. Parenteral estrogen versus total androgen ablation in the treatment of advanced prostate carcinoma: Effects on overall survival and cardiovascular mortality. Urology 2000, 55, 328–332. [Google Scholar] [CrossRef] [PubMed]
  104. Hedlund, P.O.; Ala-Opas, M.; Brekkan, E.; Damber, J.E.; Damber, L.; Hagerman, I.; Haukaas, S.; Henriksson, P.; Iversen, P.; Pousette, Å. Parenteral estrogen versus combined androgen deprivation in the treatment of metastatic prostatic cancer-Scandinavian Prostatic Cancer Group (SPCG) Study No. 5. Scand. J. Urol. Nephrol. 2002, 36, 405–413. [Google Scholar] [CrossRef] [PubMed]
  105. Olov Hedlund, P.; Damber, J.-E.; Hagerman, I.; Haukaas, S.; Henriksson, P.; Iversen, P.; Johansson, R.; Klarskov, P.; Lundbeck, F.; Rasmussen, F. Parenteral estrogen versus combined androgen deprivation in the treatment of metastatic prostatic cancer: Part 2. Final evaluation of the Scandinavian Prostatic Cancer Group (SPCG) Study No. 5. Scand. J. Urol. Nephrol. 2008, 42, 220–229. [Google Scholar] [CrossRef] [PubMed]
  106. Langley, R.E.; Cafferty, F.H.; Alhasso, A.A.; Rosen, S.D.; Sundaram, S.K.; Freeman, S.C.; Pollock, P.; Jinks, R.C.; Godsland, I.F.; Kockelbergh, R. Cardiovascular outcomes in patients with locally advanced and metastatic prostate cancer treated with luteinising-hormone-releasing-hormone agonists or transdermal oestrogen: The randomised, phase 2 MRC PATCH trial (PR09). Lancet Oncol. 2013, 14, 306–316. [Google Scholar] [CrossRef] [PubMed]
  107. Langley, R.E.; Godsland, I.F.; Kynaston, H.; Clarke, N.W.; Rosen, S.D.; Morgan, R.C.; Pollock, P.; Kockelbergh, R.; Lalani, E.N.; Dearnaley, D. Early hormonal data from a multicentre phase II trial using transdermal oestrogen patches as first-line hormonal therapy in patients with locally advanced or metastatic prostate cancer. BJU Int. 2008, 102, 442–445. [Google Scholar] [CrossRef]
  108. Stein, M.; Goodin, S.; Doyle-Lindrud, S.; Silberberg, J.; Kane, M.; Metzger, D.; Eddy, S.; Shih, W.; DiPaola, R.S. Transdermal estradiol in castrate and chemotherapy resistant prostate cancer. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2012, 18, CR260. [Google Scholar] [CrossRef] [PubMed]
  109. Phillips, I.; Shah, S.I.; Duong, T.; Abel, P.; Langley, R.E. Androgen deprivation therapy and the re-emergence of parenteral estrogen in prostate cancer. Oncol. Hematol. Rev. 2014, 10, 42. [Google Scholar] [CrossRef]
  110. Silva, É.D.; Ferreira, U.; Matheus, W.; Faria, E.F.; Silva, G.D.; Saito, M.; De Souza, A.A.; Laranjo, A.; Clark, O.; Magna, L.A. Goserelin versus leuprolide in the chemical castration of patients with prostate cancer. Int. Urol. Nephrol. 2012, 44, 1039–1044. [Google Scholar] [CrossRef]
  111. Otto, C.; Rohde-Schulz, B.; Schwarz, G.; Fuchs, I.; Klewer, M.; Brittain, D.; Langer, G.; Bader, B.; Prelle, K.; Nubbemeyer, R.; et al. G protein-coupled receptor 30 localizes to the endoplasmic reticulum and is not activated by estradiol. Endocrinology 2008, 149, 4846–4856. [Google Scholar] [CrossRef]
  112. Thomas, J.A.; Keenan, E.J. Principles of Endocrine Pharmacology; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2012. [Google Scholar]
  113. Oettel, M. Estrogens and antiestrogens in the male. In Estrogens and Antiestrogens II; Springer: Berlin/Heidelberg, Germany, 1999; pp. 505–571. [Google Scholar]
  114. Höfling, G.; Heynemann, H. Die orale Östrogentherapie des fortgeschrittenen Prostatakarzinoms—Anlaß für eine Neubewertung? Der. Urol. B 1998, 38, 165–170. [Google Scholar] [CrossRef]
  115. Hinkelbein, W.; Miller, K.; Wiegel, T. Prostatakarzinom Urologische und Strahlentherapeutische Aspekte; Springer: Berlin/Heidelberg, Germany, 1999. [Google Scholar]
  116. Lupelescu, A. (Ed.) Hormones and hormonotherapy. In Hormones and Vitamins in Cancer Treatment; CRC Press: Boca Raton, FL, USA, 1990; pp. 33–90. [Google Scholar]
  117. Weinstein, R.S. Prostate Cancer|International Perspectives in Urology; Jacobi, G.H., Hohenfellner, R., Eds.; Williams & Wilkins Co.: Baltimore, MD, USA, 1983; Volume 3. [Google Scholar]
  118. Sweetman, S.C. Sex hormones and their modulators. In Martindale: The Complete Drug Reference; The Pharmaceutical Press: London, UK, 2009; p. 2097. [Google Scholar]
  119. Hager, H. Hagers Handbuch der Pharmazeutischen Praxis Band 8 Stoffe EO; Springer: Berlin/Heidelberg, Germany, 2013. [Google Scholar]
  120. Dao, T.L. Pharmacology and Clinical utility of Hormones in Hormone related Neoplasms. In Antineoplastic and Immunosuppressive Agents; Springer: Berlin/Heidelberg, Germany, 1975; pp. 170–192. [Google Scholar]
  121. Perry, C.M.; McTavish, D. Estramustine phosphate sodium. Drugs Aging 1995, 7, 49–74. [Google Scholar] [CrossRef]
  122. Ockrim, J.; Lalani, E.-N.; Abel, P. Therapy insight: Parenteral estrogen treatment for prostate cancer—A new dawn for an old therapy. Nat. Rev. Clin. Oncol. 2006, 3, 552. [Google Scholar] [CrossRef] [PubMed]
  123. Ockrim, J.; Lalani, E.-N.; Laniado, M.; Carter, S.S.C.; Abel, P. Transdermal estradiol therapy for advanced prostate cancer—Forward to the past? J. Urol. 2003, 169, 1735–1737. [Google Scholar] [CrossRef] [PubMed]
  124. Kohli, M.; Alikhan, M.; Spencer, H.; Carter, G. Phase I trial of intramuscular estradiol valerate (I/ME) in hormone refractory prostate cancer. J. Clin. Oncol. 2004, 22, 4726. [Google Scholar] [CrossRef]
  125. Norman, G.; Dean, M.; Langley, R.; Hodges, Z.; Ritchie, G.; Parmar, M.; Sydes, M.; Abel, P.; Eastwood, A. Parenteral oestrogen in the treatment of prostate cancer: A systematic review. Br. J. Cancer 2008, 98, 697. [Google Scholar] [CrossRef] [PubMed]
  126. Cox, R.L.; Crawford, E.D. Estrogens in the treatment of prostate cancer. J. Urol. 1995, 154, 1991–1998. [Google Scholar] [CrossRef]
  127. Carlström, K.; Stege, R.; Henriksson, P.; Grande, M.; Gunnarsson, P.O.; Pousette, A. Possible bone-preserving capacity of high-dose intramuscular depot estrogen as compared to orchidectomy in the treatment of patients with prostatic carcinoma. Prostate 1997, 31, 193–197. [Google Scholar] [CrossRef]
  128. Bergenheim, A.T.; Henriksson, R. Pharmacokinetics and pharmacodynamics of estramustine phosphate. Clin. Pharmacokinet. 1998, 34, 163–172. [Google Scholar] [CrossRef]
  129. Radovick, S. Estrogenic regulation of the GnRH neuron. Front. Endocrinol. 2012, 3, 52. [Google Scholar] [CrossRef]
  130. Warde, P.; Mason, M.; Ding, K.; Kirkbride, P.; Brundage, M.; Cowan, R.; Gospodarowicz, M.; Sanders, K.; Kostashuk, E.; Swanson, G. Combined androgen deprivation therapy and radiation therapy for locally advanced prostate cancer: A randomised, phase 3 trial. Lancet 2011, 378, 2104–2111. [Google Scholar] [CrossRef] [PubMed]
  131. Ehara, H.; Koji, T.; Deguchi, T.; Yoshii, A.; Nakano, M.; Nakane, P.K.; Kawada, Y. Expression of estrogen receptor in diseased human prostate assessed by non-radioactive in situ hybridization and immunohistochemistry. Prostate 1995, 27, 304–313. [Google Scholar] [CrossRef] [PubMed]
  132. Domińska, K.; Kowalski, A.; Ochędalski, T.; Rębas, E. Effects of testosterone and 17β-estradiol on angiotensin-induced changes in tyrosine kinase activity in the androgen-independent human prostate cancer cell line, DU145. Int. J. Mol. Med. 2017, 40, 1573–1581. [Google Scholar] [CrossRef] [PubMed]
  133. Montgomery, B.; Nelson, P.S.; Vessella, R.; Kalhorn, T.; Hess, D.; Corey, E. Estradiol suppresses tissue androgens and prostate cancer growth in castration resistant prostate cancer. BMC Cancer 2010, 10, 244. [Google Scholar] [CrossRef] [PubMed]
  134. Walter, P.; Green, S.; Greene, G.; Krust, A.; Bornert, J.-M.; Jeltsch, J.-M.; Staub, A.; Jensen, E.; Scrace, G.; Waterfield, M. Cloning of the human estrogen receptor cDNA. Proc. Natl. Acad. Sci. USA 1985, 82, 7889–7893. [Google Scholar] [CrossRef] [PubMed]
  135. Bonkhoff, H.; Fixemer, T.; Hunsicker, I.; Remberger, K. Estrogen Receptor Expression in Prostate Cancer and Premalignant Prostatic Lesions. Am. J. Pathol. 1999, 155, 641–647. [Google Scholar] [CrossRef] [PubMed]
  136. Adams, J.Y.; Leav, I.; Lau, K.M.; Ho, S.M.; Pflueger, S.M. Expression of estrogen receptor beta in the fetal, neonatal, and prepubertal human prostate. Prostate 2002, 52, 69–81. [Google Scholar] [CrossRef] [PubMed]
  137. Makela, S.; Strauss, L.; Kuiper, G.; Valve, E.; Salmi, S.; Santti, R.; Gustafsson, J.A. Differential expression of estrogen receptors alpha and beta in adult rat accessory sex glands and lower urinary tract. Mol. Cell. Endocrinol. 2000, 164, 109–116. [Google Scholar] [CrossRef]
  138. Schulze, H.; Claus, S. Histological localization of estrogen receptors in normal and diseased human prostates by immunocytochemistry. Prostate 1990, 16, 331–343. [Google Scholar] [CrossRef]
  139. Hiramatsu, M.; Maehara, I.; Orikasa, S.; Sasano, H. Immunolocalization of oestrogen and progesterone receptors in prostatic hyperplasia and carcinoma. Histopathology 1996, 28, 163–168. [Google Scholar] [CrossRef]
  140. Bonkhoff, H. Estrogen receptor signaling in prostate cancer: Implications for carcinogenesis and tumor progression. Prostate 2018, 78, 2–10. [Google Scholar] [CrossRef] [PubMed]
  141. Linja, M.J.; Savinainen, K.J.; Tammela, T.L.J.; Isola, J.J.; Visakorpi, T. Expression of ERα and ERβ in prostate cancer. Prostate 2003, 55, 180–186. [Google Scholar] [CrossRef] [PubMed]
  142. Maggiolini, M.; Recchia, A.G.; Carpino, A.; Vivacqua, A.; Fasanella, G.; Rago, V.; Pezzi, V.; Briand, P.-A.; Picard, D.; Ando, S. Oestrogen receptor beta is required for androgen-stimulated proliferation of LNCaP prostate cancer cells. J. Mol. Endocrinol. 2004, 32, 777–791. [Google Scholar] [CrossRef] [PubMed]
  143. Härkönen, P.L.; Mäkelä, S.I. Role of estrogens in development of prostate cancer. J. Steroid Biochem. Mol. Biol. 2004, 92, 297–305. [Google Scholar] [CrossRef] [PubMed]
  144. Ricke, W.A.; McPherson, S.J.; Bianco, J.J.; Cunha, G.R.; Wang, Y.; Risbridger, G.P. Prostatic hormonal carcinogenesis is mediated by in situ estrogen production and estrogen receptor alpha signaling. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2008, 22, 1512–1520. [Google Scholar] [CrossRef] [PubMed]
  145. Butt, A.J.; McNeil, C.M.; Musgrove, E.A.; Sutherland, R.L. Downstream targets of growth factor and oestrogen signalling and endocrine resistance: The potential roles of c-Myc, cyclin D1 and cyclin E. Endocr.-Relat. Cancer 2005, 12, S47–S59. [Google Scholar] [CrossRef] [PubMed]
  146. Pugeat, M.M.; Dunn, J.F.; Nisula, B.C. Transport of steroid hormones: Interaction of 70 drugs with testosterone-binding globulin and corticosteroid-binding globulin in human plasma. J. Clin. Endocrinol. Metab. 1981, 53, 69–75. [Google Scholar] [CrossRef] [PubMed]
  147. Koong, L.Y.; Watson, C.S. Direct estradiol and diethylstilbestrol actions on early- versus late-stage prostate cancer cells. Prostate 2014, 74, 1589–1603. [Google Scholar] [CrossRef] [PubMed]
  148. Barreiros, L.; Queiroz, J.F.; Magalhães, L.M.; Silva, A.M.; Segundo, M.A. Analysis of 17-β-estradiol and 17-α-ethinylestradiol in biological and environmental matrices—A review. Microchem. J. 2016, 126, 243–262. [Google Scholar] [CrossRef]
  149. Zhang, H.; Cui, D.; Wang, B.; Han, Y.-H.; Balimane, P.; Yang, Z.; Sinz, M.; Rodrigues, A.D. Pharmacokinetic drug interactions involving 17α-ethinylestradiol. Clin. Pharmacokinet. 2007, 46, 133–157. [Google Scholar] [CrossRef]
  150. Ho, C.K.; Nanda, J.; Chapman, K.E.; Habib, F.K. Oestrogen and benign prostatic hyperplasia: Effects on stromal cell proliferation and local formation from androgen. J. Endocrinol. 2008, 197, 483–491. [Google Scholar] [CrossRef] [PubMed]
  151. Mosli, H.A.; Tolba, M.F.; Al-Abd, A.M.; Abdel-Naim, A.B. Catechol estrogens induce proliferation and malignant transformation in prostate epithelial cells. Toxicol. Lett. 2013, 220, 247–258. [Google Scholar] [CrossRef] [PubMed]
  152. Ricke, W.A.; Ishii, K.; Ricke, E.A.; Simko, J.; Wang, Y.; Hayward, S.W.; Cunha, G.R. Steroid hormones stimulate human prostate cancer progression and metastasis. Int. J. Cancer 2006, 118, 2123–2131. [Google Scholar] [CrossRef] [PubMed]
  153. Chieffi, P.; Kisslinger, A.; Sinisi, A.A.; Abbondanza, C.; Tramontano, D. 17beta-estradiol-induced activation of ERK1/2 through endogenous androgen receptor-estradiol receptor alpha-Src complex in human prostate cells. Int. J. Oncol. 2003, 23, 797–801. [Google Scholar] [PubMed]
  154. Arnold, J.T.; Le, H.; McFann, K.K.; Blackman, M.R. Comparative effects of DHEA vs. testosterone, dihydrotestosterone, and estradiol on proliferation and gene expression in human LNCaP prostate cancer cells. Am. J. Physiol. Endocrinol. Metab. 2005, 288, E573–E584. [Google Scholar] [CrossRef] [PubMed]
  155. Yang, M.; Wang, J.; Wang, L.; Shen, C.; Su, B.; Qi, M.; Hu, J.; Gao, W.; Tan, W.; Han, B. Estrogen induces androgen-repressed SOX4 expression to promote progression of prostate cancer cells. Prostate 2015, 75, 1363–1375. [Google Scholar] [CrossRef] [PubMed]
  156. Saffarini, C.M.; McDonnell-Clark, E.V.; Amin, A.; Huse, S.M.; Boekelheide, K. Developmental exposure to estrogen alters differentiation and epigenetic programming in a human fetal prostate xenograft model. PLoS ONE 2015, 10, e0122290. [Google Scholar] [CrossRef] [PubMed]
  157. Vom Saal, F.S.; Timms, B.G.; Montano, M.M.; Palanza, P.; Thayer, K.A.; Nagel, S.C.; Dhar, M.D.; Ganjam, V.; Parmigiani, S.; Welshons, W.V. Prostate enlargement in mice due to fetal exposure to low doses of estradiol or diethylstilbestrol and opposite effects at high doses. Proc. Natl. Acad. Sci. USA 1997, 94, 2056–2061. [Google Scholar] [CrossRef] [PubMed]
  158. Leav, I.; Merk, F.B.; Kwan, P.W.-L.; Ho, S.-M. Androgen-supported estrogen-enhanced epithelial proliferation in the prostates of intact noble rats. Prostate 1989, 15, 23–40. [Google Scholar] [CrossRef]
  159. Bosland, M.C.; Ford, H.; Horton, L. Induction at high incidence of ductal prostate adenocarcinomas in NBL/Cr and Sprague-Dawley Hsd:SD rats treated with a combination of testosterone and estradiol-17 beta or diethylstilbestrol. Carcinogenesis 1995, 16, 1311–1317. [Google Scholar] [CrossRef]
  160. Ozten, N.; Vega, K.; Liehr, J.; Huang, X.; Horton, L.; Cavalieri, E.L.; Rogan, E.G.; Bosland, M.C. Role of Estrogen in Androgen-Induced Prostate Carcinogenesis in NBL Rats. Horm. Cancer 2019, 10, 77–88. [Google Scholar] [CrossRef] [PubMed]
  161. Andersson, H.; Tisell, L.E. Morphology of rat prostatic lobes and seminal vesicles after long-term estrogen treatment. Acta Pathol. Microbiol. Et Immunol. Scandinavica. Sect. A Pathol. 1982, 90, 441–448. [Google Scholar] [CrossRef] [PubMed]
  162. Aumuller, G.; Funke, P.J.; Hahn, A.; Hoffbauer, G.; Tunn, U.; Neumann, F. Phenotypic modulation of the canine prostate after long-term treatment with androgens and estrogens. Prostate 1982, 3, 361–373. [Google Scholar] [CrossRef] [PubMed]
  163. Sugimura, Y.; Cunha, G.R.; Yonemura, C.U.; Kawamura, J. Temporal and spatial factors in diethylstilbestrol-induced squamous metaplasia of the developing human prostate. Hum. Pathol. 1988, 19, 133–139. [Google Scholar] [CrossRef] [PubMed]
  164. Risbridger, G.P.; Wang, H.; Frydenberg, M.; Cunha, G. The metaplastic effects of estrogen on mouse prostate epithelium: Proliferation of cells with basal cell phenotype. Endocrinology 2001, 142, 2443–2450. [Google Scholar] [CrossRef] [PubMed]
  165. Mills, J.L.; Bongiovanni, A.M. Effect of prenatal estrogen exposure on male genitalia. Pediatrics 1978, 62, 1160–1165. [Google Scholar] [CrossRef]
  166. Cavalieri, E.L.; Devanesan, P.; Bosland, M.C.; Badawi, A.F.; Rogan, E.G. Catechol estrogen metabolites and conjugates in different regions of the prostate of Noble rats treated with 4-hydroxyestradiol: Implications for estrogen-induced initiation of prostate cancer. Carcinogenesis 2002, 23, 329–333. [Google Scholar] [CrossRef] [PubMed]
  167. Tanaka, Y.; Sasaki, M.; Shiina, H.; Tokizane, T.; Deguchi, M.; Hirata, H.; Hinoda, Y.; Okayama, N.; Suehiro, Y.; Urakami, S.; et al. Catechol-O-methyltransferase gene polymorphisms in benign prostatic hyperplasia and sporadic prostate cancer. Cancer Epidemiol. Biomark. Prev. A Publ. Am. Assoc. Cancer Res. Cosponsored By Am. Soc. Prev. Oncol. 2006, 15, 238–244. [Google Scholar] [CrossRef] [PubMed]
  168. Mo, Z.; Gao, Y.; Cao, Y.; Gao, F.; Jian, L. An updating meta-analysis of the GSTM1, GSTT1, and GSTP1 polymorphisms and prostate cancer: A HuGE Review. Prostate 2009, 69, 662–688. [Google Scholar] [CrossRef]
  169. Nock, N.L.; Bock, C.; Neslund-Dudas, C.; Beebe-Dimmer, J.; Rundle, A.; Tang, D.; Jankowski, M.; Rybicki, B.A. Polymorphisms in glutathione S-transferase genes increase risk of prostate cancer biochemical recurrence differentially by ethnicity and disease severity. Cancer Causes Control 2009, 20, 1915. [Google Scholar] [CrossRef]
  170. Zhang, Q.; Zheng, M.; Qi, X.-L.; Liu, F.; Mao, Z.-J.; Zhang, D.-H. Effect of NQO1 C609T polymorphism on prostate cancer risk: A meta-analysis. OncoTargets Ther. 2014, 7, 907–914. [Google Scholar] [CrossRef] [PubMed]
  171. Lévesque, É.; Laverdière, I.; Audet-Walsh, É.; Caron, P.; Rouleau, M.; Fradet, Y.; Lacombe, L.; Guillemette, C. Steroidogenic Germline Polymorphism Predictors of Prostate Cancer Progression in the Estradiol Pathway. Clin. Cancer Res. 2014, 20, 2971–2983. [Google Scholar] [CrossRef] [PubMed]
  172. Cavalieri, E.; Rogan, E. Catechol quinones of estrogens in the initiation of breast, prostate, and other human cancers: Keynote lecture. Ann. N. Y. Acad. Sci. 2006, 1089, 286–301. [Google Scholar] [CrossRef] [PubMed]
  173. Mosli, H.A.; Al-Abd, A.M.; El-Shaer, M.A.; Khedr, A.; Khedr, A.; Gazzaz, F.S.; Abdel-Naim, A.B. Local inflammation influences oestrogen metabolism in prostatic tissue. BJU Int. 2012, 110, 274–282. [Google Scholar] [CrossRef] [PubMed]
  174. Rahman, H.P.; Hofland, J.; Foster, P.A. In touch with your feminine side: How oestrogen metabolism impacts prostate cancer. Endocr.-Relat. Cancer 2016, 23, R249–R266. [Google Scholar] [CrossRef] [PubMed]
  175. Migliaccio, A.; Castoria, G.; Di Domenico, M.; de Falco, A.; Bilancio, A.; Lombardi, M.; Barone, M.V.; Ametrano, D.; Zannini, M.S.; Abbondanza, C.; et al. Steroid-induced androgen receptor-oestradiol receptor beta-Src complex triggers prostate cancer cell proliferation. EMBO J. 2000, 19, 5406–5417. [Google Scholar] [CrossRef] [PubMed]
  176. Qiao, Y.; Wang, L.; Cai, L.Q.; Tan, C.; Imperato-McGinley, J.; Zhu, Y.S. Inhibition of aberrant androgen receptor induction of prostate specific antigen gene expression, cell proliferation and tumor growth by 17α-estradiol in prostate cancer. J. Urol. 2011, 185, 305–314. [Google Scholar] [CrossRef] [PubMed]
  177. Erzurumlu, Y.; Dogan, H.K.; Catakli, D.; Aydogdu, E.; Muhammed, M.T. Estrogens drive the endoplasmic reticulum-associated degradation and promote proto-oncogene c-Myc expression in prostate cancer cells by androgen receptor/estrogen receptor signaling. J. Cell Commun. Signal. 2023, 17, 793–811. [Google Scholar] [CrossRef]
  178. Lambert, A.W.; Pattabiraman, D.R.; Weinberg, R.A. Emerging Biological Principles of Metastasis. Cell 2017, 168, 670–691. [Google Scholar] [CrossRef]
  179. Tsai, J.H.; Yang, J. Epithelial-mesenchymal plasticity in carcinoma metastasis. Genes Dev 2013, 27, 2192–2206. [Google Scholar] [CrossRef]
  180. Loh, C.-Y.; Chai, J.Y.; Tang, T.F.; Wong, W.F.; Sethi, G.; Shanmugam, M.K.; Chong, P.P.; Looi, C.Y. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019, 8, 1118. [Google Scholar] [CrossRef] [PubMed]
  181. Fontana, F.; Raimondi, M.; Marzagalli, M.; Sommariva, M.; Limonta, P.; Gagliano, N. Epithelial-To-Mesenchymal Transition Markers and CD44 Isoforms Are Differently Expressed in 2D and 3D Cell Cultures of Prostate Cancer Cells. Cells 2019, 8, 143. [Google Scholar] [CrossRef]
  182. Shi, X.; Peng, Y.; Du, X.; Liu, H.; Klocker, H.; Lin, Q.; Shi, J.; Zhang, J. Estradiol promotes epithelial-to-mesenchymal transition in human benign prostatic epithelial cells. Prostate 2017, 77, 1424–1437. [Google Scholar] [CrossRef] [PubMed]
  183. Paget, S. The Distribution of Secondary Growths in Cancer of the Breast. Lancet 1889, 133, 571–573. [Google Scholar] [CrossRef]
  184. Thobe, M.N.; Clark, R.J.; Bainer, R.O.; Prasad, S.M.; Rinker-Schaeffer, C.W. From prostate to bone: Key players in prostate cancer bone metastasis. Cancers 2011, 3, 478–493. [Google Scholar] [CrossRef]
  185. Megas, G.; Chrisofos, M.; Anastasiou, I.; Tsitlidou, A.; Choreftaki, T.; Deliveliotis, C. Estrogen receptor (α and β) but not androgen receptor expression is correlated with recurrence, progression and survival in post prostatectomy T3N0M0 locally advanced prostate cancer in an urban Greek population. Asian J. Androl. 2015, 17, 98–105. [Google Scholar] [CrossRef] [PubMed]
  186. Yu, L.; Shi, J.; Cheng, S.; Zhu, Y.; Zhao, X.; Yang, K.; Du, X.; Klocker, H.; Yang, X.; Zhang, J. Estrogen promotes prostate cancer cell migration via paracrine release of ENO1 from stromal cells. Mol. Endocrinol. 2012, 26, 1521–1530. [Google Scholar] [CrossRef] [PubMed]
  187. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef]
  188. Altenberg, B.; Greulich, K.O. Genes of glycolysis are ubiquitously overexpressed in 24 cancer classes. Genomics 2004, 84, 1014–1020. [Google Scholar] [CrossRef]
  189. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef]
  190. Jaiswal, R.K.; Varshney, A.K.; Yadava, P.K. Diversity and functional evolution of the plasminogen activator system. Biomed. Pharmacother. 2018, 98, 886–898. [Google Scholar] [CrossRef]
  191. Vervoort, S.J.; van Boxtel, R.; Coffer, P.J. The role of SRY-related HMG box transcription factor 4 (SOX4) in tumorigenesis and metastasis: Friend or foe? Oncogene 2013, 32, 3397–3409. [Google Scholar] [CrossRef] [PubMed]
  192. Wang, L.; Zhang, J.; Yang, X.; Chang, Y.W.Y.; Qi, M.; Zhou, Z.; Zhang, J.; Han, B. SOX4 is associated with poor prognosis in prostate cancer and promotes epithelial-mesenchymal transition in vitro. Prostate Cancer Prostatic Dis. 2013, 16, 301–307. [Google Scholar] [CrossRef] [PubMed]
  193. Liang, Z.; Cao, J.; Tian, L.; Shen, Y.; Yang, X.; Lin, Q.; Zhang, R.; Liu, H.; Du, X.; Shi, J.; et al. Aromatase-induced endogenous estrogen promotes tumour metastasis through estrogen receptor-alpha/matrix metalloproteinase 12 axis activation in castration-resistant prostate cancer. Cancer Lett. 2019, 467, 72–84. [Google Scholar] [CrossRef] [PubMed]
  194. Sarkar, S.; Nuttall, R.K.; Liu, S.; Edwards, D.R.; Yong, V.W. Tenascin-C stimulates glioma cell invasion through matrix metalloproteinase-12. Cancer Res. 2006, 66, 11771–11780. [Google Scholar] [CrossRef]
  195. Chung, I.C.; Chen, L.C.; Chung, A.K.; Chao, M.; Huang, H.Y.; Hsueh, C.; Tsang, N.M.; Chang, K.P.; Liang, Y.; Li, H.P.; et al. Matrix metalloproteinase 12 is induced by heterogeneous nuclear ribonucleoprotein K and promotes migration and invasion in nasopharyngeal carcinoma. BMC Cancer 2014, 14, 348. [Google Scholar] [CrossRef]
  196. Nabha, S.M.; dos Santos, E.B.; Yamamoto, H.A.; Belizi, A.; Dong, Z.; Meng, H.; Saliganan, A.; Sabbota, A.; Bonfil, R.D.; Cher, M.L. Bone marrow stromal cells enhance prostate cancer cell invasion through type I collagen in an MMP-12 dependent manner. Int. J. Cancer 2008, 122, 2482–2490. [Google Scholar] [CrossRef]
  197. Lombardi, A.P.G.; Cavalheiro, R.P.; Porto, C.S.; Vicente, C.M. Estrogen Receptor Signaling Pathways Involved in Invasion and Colony Formation of Androgen-Independent Prostate Cancer Cells PC-3. Int. J. Mol. Sci. 2021, 22, 1153. [Google Scholar] [CrossRef]
  198. Lombardi, A.P.G.; Vicente, C.M.; Porto, C.S. Estrogen Receptors Promote Migration, Invasion and Colony Formation of the Androgen-Independent Prostate Cancer Cells PC-3 Through β-Catenin Pathway. Front. Endocrinol. 2020, 11, 184. [Google Scholar] [CrossRef]
  199. Kanagaraj, P.; Vijayababu, M.R.; Ilangovan, R.; Senthilkumar, K.; Venkataraman, P.; Aruldhas, M.M.; Arunakaran, J. Effect of 17β-estradiol on apoptosis, IGF system components and gelatinases A and B in prostate cancer cells (PC-3). Clin. Chim. Acta 2007, 377, 70–78. [Google Scholar] [CrossRef]
  200. Qiao, Y.; Zhang, Z.K.; Cai, L.Q.; Tan, C.; Imperato-McGinley, J.L.; Zhu, Y.S. 17alpha-estradiol inhibits LAPC-4 prostatic tumor cell proliferation in cell cultures and tumor growth in xenograft animals. Prostate 2007, 67, 1719–1728. [Google Scholar] [CrossRef] [PubMed]
  201. Yu, J.D.; Yang, K.; Mao, Q.Q.; Kong, D.B.; Zheng, X.Y.; Xie, L.P. Estrogen in combination with 5-azacitidine up-regulates p75NTR expression and induces apoptosis in 22Rv1 prostate cancer cells. Mol. Med. Rep. 2009, 2, 831–836. [Google Scholar] [CrossRef] [PubMed]
  202. Figueira, M.I.; Correia, S.; Vaz, C.V.; Cardoso, H.J.; Gomes, I.M.; Marques, R.; Maia, C.J.; Socorro, S. Estrogens down-regulate the stem cell factor (SCF)/c-KIT system in prostate cells: Evidence of antiproliferative and proapoptotic effects. Biochem. Pharmacol. 2016, 99, 73–87. [Google Scholar] [CrossRef] [PubMed]
  203. Corey, E.; Quinn, J.E.; Emond, M.J.; Buhler, K.R.; Brown, L.G.; Vessella, R.L. Inhibition of androgen-independent growth of prostate cancer xenografts by 17beta-estradiol. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2002, 8, 1003–1007. [Google Scholar]
  204. Landström, M.; Bergh, A.; Tomic, R.; Damber, J.E. Estrogen treatment combined with castration inhibits tumor growth more effectively than castration alone in the Dunning R3327 rat prostatic adenocarcinoma. Prostate 1990, 17, 57–68. [Google Scholar] [CrossRef]
  205. Landström, M.; Eklöv, S.; Colosetti, P.; Nilsson, S.; Damber, J.E.; Bergh, A.; Funa, K. Estrogen induces apoptosis in a rat prostatic adenocarcinoma: Association with an increased expression of TGF-beta 1 and its type-I and type-II receptors. Int. J. Cancer 1996, 67, 573–579. [Google Scholar] [CrossRef]
  206. Westin, P.; Brändström, A.; Damber, J.E.; Bergh, A. Castration plus oestrogen treatment induces but castration alone suppresses epithelial cell apoptosis in an androgen-sensitive rat prostatic adenocarcinoma. Br. J. Cancer 1995, 72, 140–145. [Google Scholar] [CrossRef] [PubMed]
  207. Kumar, A.P.; Garcia, G.E.; Slaga, T.J. 2-methoxyestradiol blocks cell-cycle progression at G2/M phase and inhibits growth of human prostate cancer cells. Mol. Carcinog. 2001, 31, 111–124. [Google Scholar] [CrossRef] [PubMed]
  208. Qadan, L.R.; Perez-Stable, C.M.; Anderson, C.; D’Ippolito, G.; Herron, A.; Howard, G.A.; Roos, B.A. 2-Methoxyestradiol induces G2/M arrest and apoptosis in prostate cancer. Biochem. Biophys. Res. Commun. 2001, 285, 1259–1266. [Google Scholar] [CrossRef]
  209. Ray, G.; Dhar, G.; Van Veldhuizen, P.J.; Banerjee, S.; Saxena, N.K.; Sengupta, K.; Banerjee, S.K. Modulation of cell-cycle regulatory signaling network by 2-methoxyestradiol in prostate cancer cells is mediated through multiple signal transduction pathways. Biochemistry 2006, 45, 3703–3713. [Google Scholar] [CrossRef]
  210. Horny, C.; Balasubashini, M.S.; Komanduri, K.; Ganapathy, M.; Yeh, I.T.; Ghosh, R.; Kumar, A.P. 2-methoxyestradiol Prevents LNCaP Tumor Development in Nude Mice: Potential Role of G2/M Regulatory Proteins. J. Cell Death 2009, 2, 1–8. [Google Scholar] [CrossRef] [PubMed]
  211. Reiner, T.; de las Pozas, A.; Gomez, L.A.; Perez-Stable, C. Low dose combinations of 2-methoxyestradiol and docetaxel block prostate cancer cells in mitosis and increase apoptosis. Cancer Lett. 2009, 276, 21–31. [Google Scholar] [CrossRef] [PubMed]
  212. Ghosh, R.; Ganapathy, M.; Alworth, W.L.; Chan, D.C.; Kumar, A.P. Combination of 2-methoxyestradiol (2-ME2) and eugenol for apoptosis induction synergistically in androgen independent prostate cancer cells. J Steroid Biochem. Mol. Biol. 2009, 113, 25–35. [Google Scholar] [CrossRef] [PubMed]
  213. Bu, S.; Blaukat, A.; Fu, X.; Heldin, N.E.; Landström, M. Mechanisms for 2-methoxyestradiol-induced apoptosis of prostate cancer cells. FEBS Lett. 2002, 531, 141–151. [Google Scholar] [CrossRef] [PubMed]
  214. Yun, H.; Xie, J.; Olumi, A.F.; Ghosh, R.; Kumar, A.P. Activation of AKR1C1/ERβ induces apoptosis by downregulation of c-FLIP in prostate cancer cells: A prospective therapeutic opportunity. Oncotarget 2015, 6, 11600–11613. [Google Scholar] [CrossRef] [PubMed]
  215. Shimada, K.; Nakamura, M.; Ishida, E.; Kishi, M.; Matsuyoshi, S.; Konishi, N. The molecular mechanism of sensitization to Fas-mediated apoptosis by 2-methoxyestradiol in PC3 prostate cancer cells. Mol. Carcinog. 2004, 39, 1–9. [Google Scholar] [CrossRef] [PubMed]
  216. Davoodpour, P.; Bergström, M.; Landström, M. Effects of 2-methoxyestradiol on proliferation, apoptosis and PET-tracer uptake in human prostate cancer cell aggregates. Nucl. Med. Biol. 2004, 31, 867–874. [Google Scholar] [CrossRef] [PubMed]
  217. Davoodpour, P.; Landström, M. 2-Methoxyestradiol-induced apoptosis in prostate cancer cells requires Smad7. J. Biol. Chem. 2005, 280, 14773–14779. [Google Scholar] [CrossRef] [PubMed]
  218. Ganapathy, M.; Ghosh, R.; Jianping, X.; Zhang, X.; Bedolla, R.; Schoolfield, J.; Yeh, I.T.; Troyer, D.A.; Olumi, A.F.; Kumar, A.P. Involvement of FLIP in 2-methoxyestradiol-induced tumor regression in transgenic adenocarcinoma of mouse prostate model. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2009, 15, 1601–1611. [Google Scholar] [CrossRef]
  219. Sato, F.; Fukuhara, H.; Basilion, J.P. Effects of hormone deprivation and 2-methoxyestradiol combination therapy on hormone-dependent prostate cancer in vivo. Neoplasia 2005, 7, 838–846. [Google Scholar] [CrossRef]
  220. Yang, F.; Song, L.; Wang, H.; Wang, J.; Xu, Z.; Xing, N. Combination of Quercetin and 2-Methoxyestradiol Enhances Inhibition of Human Prostate Cancer LNCaP and PC-3 Cells Xenograft Tumor Growth. PLoS ONE 2015, 10, e0128277. [Google Scholar] [CrossRef] [PubMed]
  221. Garcia, G.E.; Wisniewski, H.G.; Lucia, M.S.; Arevalo, N.; Slaga, T.J.; Kraft, S.L.; Strange, R.; Kumar, A.P. 2-Methoxyestradiol inhibits prostate tumor development in transgenic adenocarcinoma of mouse prostate: Role of tumor necrosis factor-alpha-stimulated gene 6. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2006, 12, 980–988. [Google Scholar] [CrossRef] [PubMed]
  222. Kuiper, G.G.; Shughrue, P.J.; Merchenthaler, I.; Gustafsson, J.A. The estrogen receptor beta subtype: A novel mediator of estrogen action in neuroendocrine systems. Front. Neuroendocr. 1998, 19, 253–286. [Google Scholar] [CrossRef] [PubMed]
  223. Nilsson, S.; Mäkelä, S.; Treuter, E.; Tujague, M.; Thomsen, J.; Andersson, G.; Enmark, E.; Pettersson, K.; Warner, M.; Gustafsson, J.A. Mechanisms of estrogen action. Physiol. Rev. 2001, 81, 1535–1565. [Google Scholar] [CrossRef] [PubMed]
  224. Kuiper, G.G.; Carlsson, B.; Grandien, K.; Enmark, E.; Häggblad, J.; Nilsson, S.; Gustafsson, J.A. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology 1997, 138, 863–870. [Google Scholar] [CrossRef] [PubMed]
  225. Dey, P.; Ström, A.; Gustafsson, J. Estrogen receptor β upregulates FOXO3a and causes induction of apoptosis through PUMA in prostate cancer. Oncogene 2014, 33, 4213–4225. [Google Scholar] [CrossRef] [PubMed]
  226. Dondi, D.; Piccolella, M.; Biserni, A.; Della Torre, S.; Ramachandran, B.; Locatelli, A.; Rusmini, P.; Sau, D.; Caruso, D.; Maggi, A.; et al. Estrogen receptor beta and the progression of prostate cancer: Role of 5alpha-androstane-3beta,17beta-diol. Endocr. -Relat. Cancer 2010, 17, 731–742. [Google Scholar] [CrossRef]
  227. Guerini, V.; Sau, D.; Scaccianoce, E.; Rusmini, P.; Ciana, P.; Maggi, A.; Martini, P.G.; Katzenellenbogen, B.S.; Martini, L.; Motta, M.; et al. The androgen derivative 5alpha-androstane-3beta,17beta-diol inhibits prostate cancer cell migration through activation of the estrogen receptor beta subtype. Cancer Res. 2005, 65, 5445–5453. [Google Scholar] [CrossRef] [PubMed]
  228. Christoforou, P.; Christopoulos, P.F.; Koutsilieris, M. The Role of Estrogen Receptor β in Prostate Cancer. Mol. Med. 2014, 20, 427–434. [Google Scholar] [CrossRef]
  229. Kowalska, K.; Piastowska-Ciesielska, A.W. Oestrogens and oestrogen receptors in prostate cancer. SpringerPlus 2016, 5, 522. [Google Scholar] [CrossRef]
  230. Lennartsson, J.; Rönnstrand, L. Stem Cell Factor Receptor/c-Kit: From Basic Science to Clinical Implications. Physiol. Rev. 2012, 92, 1619–1649. [Google Scholar] [CrossRef] [PubMed]
  231. Ulivi, P.; Zoli, W.; Medri, L.; Amadori, D.; Saragoni, L.; Barbanti, F.; Calistri, D.; Silvestrini, R. c-kit and SCF expression in normal and tumor breast tissue. Breast Cancer Res. Treat. 2004, 83, 33–42. [Google Scholar] [CrossRef] [PubMed]
  232. Di Lorenzo, G.; Autorino, R.; D’Armiento, F.P.; Mignogna, C.; De Laurentiis, M.; De Sio, M.; D’Armiento, M.; Damiano, R.; Vecchio, G.; De Placido, S. Expression of proto-oncogene c-kit in high risk prostate cancer. Eur. J. Surg. Oncol. (EJSO) 2004, 30, 987–992. [Google Scholar] [CrossRef] [PubMed]
  233. Gould, K.L.; Nurse, P. Tyrosine phosphorylation of the fission yeast cdc2+ protein kinase regulates entry into mitosis. Nature 1989, 342, 39–45. [Google Scholar] [CrossRef] [PubMed]
  234. Nurse, P. Universal control mechanism regulating onset of M-phase. Nature 1990, 344, 503–508. [Google Scholar] [CrossRef]
  235. Perez-Stable, C. 2-Methoxyestradiol and paclitaxel have similar effects on the cell cycle and induction of apoptosis in prostate cancer cells. Cancer Lett. 2006, 231, 49–64. [Google Scholar] [CrossRef] [PubMed]
  236. Chinnaiyan, A.M.; O’Rourke, K.; Tewari, M.; Dixit, V.M. FADD, a novel death domain-containing protein, interacts with the death domain of Fas and initiates apoptosis. Cell 1995, 81, 505–512. [Google Scholar] [CrossRef]
  237. Nagata, S. Apoptosis by death factor. Cell 1997, 88, 355–365. [Google Scholar] [CrossRef]
  238. Ashkenazi, A.; Dixit, V.M. Death receptors: Signaling and modulation. Science 1998, 281, 1305–1308. [Google Scholar] [CrossRef]
  239. Krueger, A.; Baumann, S.; Krammer, P.H.; Kirchhoff, S. FLICE-inhibitory proteins: Regulators of death receptor-mediated apoptosis. Mol. Cell. Biol. 2001, 21, 8247–8254. [Google Scholar] [CrossRef]
  240. Irmler, M.; Thome, M.; Hahne, M.; Schneider, P.; Hofmann, K.; Steiner, V.; Bodmer, J.L.; Schröter, M.; Burns, K.; Mattmann, C.; et al. Inhibition of death receptor signals by cellular FLIP. Nature 1997, 388, 190–195. [Google Scholar] [CrossRef] [PubMed]
  241. Heldin, C.H.; Miyazono, K.; ten Dijke, P. TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Nature 1997, 390, 465–471. [Google Scholar] [CrossRef] [PubMed]
  242. Moustakas, A.; Souchelnytskyi, S.; Heldin, C.H. Smad regulation in TGF-beta signal transduction. J. Cell Sci. 2001, 114, 4359–4369. [Google Scholar] [CrossRef] [PubMed]
  243. Shi, Y.; Massagué, J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 2003, 113, 685–700. [Google Scholar] [CrossRef]
  244. Edlund, S.; Bu, S.; Schuster, N.; Aspenström, P.; Heuchel, R.; Heldin, N.-E.; Dijke, P.T.; Heldin, C.-H.; Landström, M. Transforming Growth Factor-β1 (TGF-β)–induced Apoptosis of Prostate Cancer Cells Involves Smad7-dependent Activation of p38 by TGF-β-activated Kinase 1 and Mitogen-activated Protein Kinase Kinase 3. Mol. Biol. Cell 2003, 14, 529–544. [Google Scholar] [CrossRef] [PubMed]
  245. Mazars, A.; Lallemand, F.; Prunier, C.; Marais, J.; Ferrand, N.; Pessah, M.; Cherqui, G.; Atfi, A. Evidence for a role of the JNK cascade in Smad7-mediated apoptosis. J. Biol. Chem. 2001, 276, 36797–36803. [Google Scholar] [CrossRef] [PubMed]
  246. Chang, I.; Majid, S.; Saini, S.; Zaman, M.S.; Yamamura, S.; Chiyomaru, T.; Shahryari, V.; Fukuhara, S.; Deng, G.; Dahiya, R.; et al. Hrk mediates 2-methoxyestradiol-induced mitochondrial apoptotic signaling in prostate cancer cells. Mol. Cancer Ther. 2013, 12, 1049–1059. [Google Scholar] [CrossRef]
  247. Van Veldhuizen, P.J.; Ray, G.; Banerjee, S.; Dhar, G.; Kambhampati, S.; Dhar, A.; Banerjee, S.K. 2-Methoxyestradiol modulates beta-catenin in prostate cancer cells: A possible mediator of 2-methoxyestradiol-induced inhibition of cell growth. Int. J. Cancer 2008, 122, 567–571. [Google Scholar] [CrossRef] [PubMed]
  248. Luo, X.; Budihardjo, I.; Zou, H.; Slaughter, C.; Wang, X. Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell 1998, 94, 481–490. [Google Scholar] [CrossRef]
  249. Garrido, C.; Galluzzi, L.; Brunet, M.; Puig, P.E.; Didelot, C.; Kroemer, G. Mechanisms of cytochrome c release from mitochondria. Cell Death Differ. 2006, 13, 1423–1433. [Google Scholar] [CrossRef]
  250. Banerjee, P.P.; Banerjee, S.; Brown, T.R.; Zirkin, B.R. Androgen action in prostate function and disease. Am J. Clin. Exp. Urol. 2018, 6, 62–77. [Google Scholar] [PubMed]
  251. Harris, W.P.; Mostaghel, E.A.; Nelson, P.S.; Montgomery, B. Androgen deprivation therapy: Progress in understanding mechanisms of resistance and optimizing androgen depletion. Nat. Clin. Pract. Urol. 2009, 6, 76–85. [Google Scholar] [CrossRef] [PubMed]
  252. Gehrig, J.; Kaulfuß, S.; Jarry, H.; Bremmer, F.; Stettner, M.; Burfeind, P.; Thelen, P. Prospects of estrogen receptor β activation in the treatment of castration-resistant prostate cancer. Oncotarget 2017, 8, 34971–34979. [Google Scholar] [CrossRef] [PubMed]
  253. McPherson, S.J.; Hussain, S.; Balanathan, P.; Hedwards, S.L.; Niranjan, B.; Grant, M.; Chandrasiri, U.P.; Toivanen, R.; Wang, Y.; Taylor, R.A.; et al. Estrogen receptor-beta activated apoptosis in benign hyperplasia and cancer of the prostate is androgen independent and TNFalpha mediated. Proc. Natl. Acad. Sci. USA 2010, 107, 3123–3128. [Google Scholar] [CrossRef] [PubMed]
  254. Hussain, S.; Lawrence, M.G.; Taylor, R.A.; Lo, C.Y.; Frydenberg, M.; Ellem, S.J.; Furic, L.; Risbridger, G.P. Estrogen receptor β activation impairs prostatic regeneration by inducing apoptosis in murine and human stem/progenitor enriched cell populations. PLoS ONE 2012, 7, e40732. [Google Scholar] [CrossRef] [PubMed]
  255. Mobley, J.A.; L’Esperance, J.O.; Wu, M.; Friel, C.J.; Hanson, R.H.; Ho, S.M. The novel estrogen 17alpha-20Z-21-[(4-amino)phenyl]-19-norpregna-1,3,5(10),20-tetraene-3,17beta-diol induces apoptosis in prostate cancer cell lines at nanomolar concentrations in vitro. Mol. Cancer Ther. 2004, 3, 587–595. [Google Scholar] [CrossRef] [PubMed]
  256. Morris, G.Z.; Williams, R.L.; Elliott, M.S.; Beebe, S.J. Resveratrol induces apoptosis in LNCaP cells and requires hydroxyl groups to decrease viability in LNCaP and DU 145 cells. Prostate 2002, 52, 319–329. [Google Scholar] [CrossRef] [PubMed]
  257. Robertson, C.N.; Roberson, K.M.; Padilla, G.M.; O’Brien, E.T.; Cook, J.M.; Kim, C.S.; Fine, R.L. Induction of apoptosis by diethylstilbestrol in hormone-insensitive prostate cancer cells. J. Natl. Cancer Inst. 1996, 88, 908–917. [Google Scholar] [CrossRef] [PubMed]
  258. Koike, H.; Ito, K.; Takezawa, Y.; Oyama, T.; Yamanaka, H.; Suzuki, K. Insulin-like growth factor binding protein-6 inhibits prostate cancer cell proliferation: Implication for anticancer effect of diethylstilbestrol in hormone refractory prostate cancer. Br. J. Cancer 2005, 92, 1538–1544. [Google Scholar] [CrossRef]
  259. Montgomery, R.B.; Bonham, M.; Nelson, P.S.; Grim, J.; Makary, E.; Vessella, R.; Stahl, W.L. Estrogen effects on tubulin expression and taxane mediated cytotoxicity in prostate cancer cells. Prostate 2005, 65, 141–150. [Google Scholar] [CrossRef]
  260. Muronetz, V.I.; Wang, Z.X.; Keith, T.J.; Knull, H.R.; Srivastava, D.K. Binding constants and stoichiometries of glyceraldehyde 3-phosphate dehydrogenase-tubulin complexes. Arch. Biochem. Biophys. 1994, 313, 253–260. [Google Scholar] [CrossRef] [PubMed]
  261. Kawashima, H.; Tanaka, T.; Cheng, J.S.; Sugita, S.; Ezaki, K.; Kurisu, T.; Nakatani, T. Effect of anti-estrogens on the androgen receptor activity and cell proliferation in prostate cancer cells. Urol. Res. 2004, 32, 406–410. [Google Scholar] [CrossRef] [PubMed]
  262. Armiñán, A.; Mendes, L.; Carrola, J.; Movellan, J.; Vicent, M.J.; Duarte, I.F. HIF-1α inhibition by diethylstilbestrol and its polyacetal conjugate in hypoxic prostate tumour cells: Insights from NMR metabolomics. J. Drug Target. 2017, 25, 845–855. [Google Scholar] [CrossRef] [PubMed]
  263. Trauger, R.; Corey, E.; Bell, D.; White, S.; Garsd, A.; Stickney, D.; Reading, C.; Frincke, J. Inhibition of androstenediol-dependent LNCaP tumour growth by 17α-ethynyl-5α-androstane-3α, 17β-diol (HE3235). Br. J. Cancer 2009, 100, 1068–1072. [Google Scholar] [CrossRef] [PubMed]
  264. Koreckij, T.D.; Trauger, R.J.; Montgomery, R.B.; Pitts, T.E.; Coleman, I.; Nguyen, H.; Reading, C.L.; Nelson, P.S.; Vessella, R.L.; Corey, E. HE3235 inhibits growth of castration-resistant prostate cancer. Neoplasia 2009, 11, 1216–1225. [Google Scholar] [CrossRef] [PubMed]
  265. Moriyama-Gonda, N.; Shiina, H.; Terashima, M.; Satoh, K.; Igawa, M. Rationale and clinical implication of combined chemotherapy with cisplatin and oestrogen in prostate cancer: Primary evidence based on methylation analysis of oestrogen receptor-alpha. BJU Int. 2008, 101, 485–491. [Google Scholar] [CrossRef] [PubMed]
  266. Arvelo, F.; Sojo, F.; Cotte, C. Tumour progression and metastasis. Ecancermedicalscience 2016, 10, 617. [Google Scholar] [CrossRef] [PubMed]
  267. Lombardi, A.P.; Pisolato, R.; Vicente, C.M.; Lazari, M.F.; Lucas, T.F.; Porto, C.S. Estrogen receptor beta (ERβ) mediates expression of β-catenin and proliferation in prostate cancer cell line PC-3. Mol. Cell. Endocrinol. 2016, 430, 12–24. [Google Scholar] [CrossRef] [PubMed]
  268. Aberle, H.; Schwartz, H.; Kemler, R. Cadherin-catenin complex: Protein interactions and their implications for cadherin function. J. Cell. Biochem. 1996, 61, 514–523. [Google Scholar] [CrossRef]
  269. Cavallaro, U.; Christofori, G. Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat. Rev. Cancer 2004, 4, 118–132. [Google Scholar] [CrossRef]
  270. Silva, R.S.; Lombardi, A.P.G.; de Souza, D.S.; Vicente, C.M.; Porto, C.S. Activation of estrogen receptor beta (ERβ) regulates the expression of N-cadherin, E-cadherin and β-catenin in androgen-independent prostate cancer cells. Int. J. Biochem. Cell Biol. 2018, 96, 40–50. [Google Scholar] [CrossRef] [PubMed]
  271. Trudel, D.; Fradet, Y.; Meyer, F.; Harel, F.; Têtu, B. Significance of MMP-2 Expression in Prostate Cancer. an Immunohistochem. Study 2003, 63, 8511–8515. [Google Scholar]
  272. Trudel, D.; Fradet, Y.; Meyer, F.; Harel, F.; Têtu, B. Prostate cancer: MMP2, MMP9, MMP14, TIMP2 and disease-free survival. Cancer Res. 2004, 64, 264. [Google Scholar]
  273. van Moorselaar, R.J.; Voest, E.E. Angiogenesis in prostate cancer: Its role in disease progression and possible therapeutic approaches. Mol. Cell. Endocrinol. 2002, 197, 239–250. [Google Scholar] [CrossRef] [PubMed]
  274. Li, Y.; Cozzi, P.J. Angiogenesis as a strategic target for prostate cancer therapy. Med. Res. Rev. 2010, 30, 23–66. [Google Scholar] [CrossRef] [PubMed]
  275. Niu, Y.N.; Xia, S.J. Stroma-epithelium crosstalk in prostate cancer. Asian J. Androl. 2009, 11, 28–35. [Google Scholar] [CrossRef] [PubMed]
  276. Melincovici, C.S.; Boşca, A.B.; Şuşman, S.; Mărginean, M.; Mihu, C.; Istrate, M.; Moldovan, I.M.; Roman, A.L.; Mihu, C.M. Vascular endothelial growth factor (VEGF)—Key factor in normal and pathological angiogenesis. Rom. J. Morphol. Embryol. = Rev. Roum. De Morphol. Et Embryol. 2018, 59, 455–467. [Google Scholar]
  277. Siveen, K.S.; Prabhu, K.; Krishnankutty, R.; Kuttikrishnan, S.; Tsakou, M.; Alali, F.Q.; Dermime, S.; Mohammad, R.M.; Uddin, S. Vascular Endothelial Growth Factor (VEGF) Signaling in Tumour Vascularization: Potential and Challenges. Curr. Vasc. Pharmacol. 2017, 15, 339–351. [Google Scholar] [CrossRef] [PubMed]
  278. Wen, J.; Zhao, Y.; Li, J.; Weng, C.; Cai, J.; Yang, K.; Yuan, H.; Imperato-McGinley, J.; Zhu, Y.S. Suppression of DHT-induced paracrine stimulation of endothelial cell growth by estrogens via prostate cancer cells. Prostate 2013, 73, 1069–1081. [Google Scholar] [CrossRef]
  279. Yu, J.; Moon, A.; Kim, H.-R.C. Both Platelet-Derived Growth Factor Receptor (PDGFR)-α and PDGFR-β Promote Murine Fibroblast Cell Migration. Biochem. Biophys. Res. Commun. 2001, 282, 697–700. [Google Scholar] [CrossRef]
  280. Heldin, C.H. Targeting the PDGF signaling pathway in tumor treatment. Cell Commun. Signal. CCS 2013, 11, 97. [Google Scholar] [CrossRef] [PubMed]
  281. Adlercreutz, H. Phytoestrogens and breast cancer. J. Steroid Biochem. Mol. Biol. 2002, 83, 113–118. [Google Scholar] [CrossRef] [PubMed]
  282. Adlercreutz, H. Epidemiology of phytoestrogens. Bailliere’S Clin. Endocrinol. Metab. 1998, 12, 605–623. [Google Scholar] [CrossRef] [PubMed]
  283. Moon, Y.J.; Wang, X.; Morris, M.E. Dietary flavonoids: Effects on xenobiotic and carcinogen metabolism. Toxicol. Vitr. 2006, 20, 187–210. [Google Scholar] [CrossRef] [PubMed]
  284. Van Poppel, H.; Tombal, B. Chemoprevention of prostate cancer with nutrients and supplements. Cancer Manag. Res. 2011, 3, 91. [Google Scholar] [CrossRef] [PubMed]
  285. Shin, J.; Millstine, D.; Ruddy, B.; Wallace, M.; Fields, H. Effect of Plant- and Animal-Based Foods on Prostate Cancer Risk. J. Osteopath. Med. 2019, 119, 736–746. [Google Scholar] [CrossRef] [PubMed]
  286. Sirtori, C.R.; Arnoldi, A.; Johnson, S.K. Phytoestrogens: End of a tale? Ann. Med. 2005, 37, 423–438. [Google Scholar] [CrossRef] [PubMed]
  287. Kim, S.H.; Park, M.J. Effects of phytoestrogen on sexual development. Korean J. Pediatr. 2012, 55, 265–271. [Google Scholar] [CrossRef] [PubMed]
  288. Dixon, R.A. Phytoestrogens. Annu. Rev. Plant Biol. 2004, 55, 225–261. [Google Scholar] [CrossRef]
  289. Lephart, E.D. Modulation of Aromatase by Phytoestrogens. Enzym. Res. 2015, 2015, 594656. [Google Scholar] [CrossRef]
  290. Michel, T.; Halabalaki, M.; Skaltsounis, A.-L. New concepts, experimental approaches, and dereplication strategies for the discovery of novel phytoestrogens from natural sources. Planta Medica 2013, 79, 514–532. [Google Scholar] [PubMed]
  291. Hajirahimkhan, A.; Dietz, B.M.; Bolton, J.L. Botanical modulation of menopausal symptoms: Mechanisms of action? Planta Medica 2013, 79, 538–553. [Google Scholar] [CrossRef] [PubMed]
  292. Cassidy. Potential risks and benefits of phytoestrogen-rich diets. Int. J. Vitam. Nutr. Res. 2003, 73, 120–126. [Google Scholar] [CrossRef] [PubMed]
  293. Branca, F.; Lorenzetti, S. Health effects of phytoestrogens. In Diet Diversification and Health Promotion; Karger Publishers: Basel, Switzerland, 2005; Volume 57, pp. 100–111. [Google Scholar]
  294. Lichtenstein, A.H. Soy protein, isoflavones and cardiovascular disease risk. J. Nutr. 1998, 128, 1589–1592. [Google Scholar] [CrossRef] [PubMed]
  295. Chiang, S.-S.; Pan, T.-M. Beneficial effects of phytoestrogens and their metabolites produced by intestinal microflora on bone health. Appl. Microbiol. Biotechnol. 2013, 97, 1489–1500. [Google Scholar] [CrossRef] [PubMed]
  296. Vitale, D.C.; Piazza, C.; Melilli, B.; Drago, F.; Salomone, S. Isoflavones: Estrogenic activity, biological effect and bioavailability. Eur. J. Drug Metab. Pharmacokinet. 2013, 38, 15–25. [Google Scholar] [CrossRef] [PubMed]
  297. Rietjens, I.M.; Sotoca, A.M.; Vervoort, J.; Louisse, J. Mechanisms underlying the dualistic mode of action of major soy isoflavones in relation to cell proliferation and cancer risks. Mol. Nutr. Food Res. 2013, 57, 100–113. [Google Scholar] [CrossRef] [PubMed]
  298. Paterni, I.; Granchi, C.; Katzenellenbogen, J.A.; Minutolo, F. Estrogen receptors alpha (ERα) and beta (ERβ): Subtype-selective ligands and clinical potential. Steroids 2014, 90, 13–29. [Google Scholar] [CrossRef] [PubMed]
  299. Saczko, J.; Michel, O.; Chwiłkowska, A.; Sawicka, E.; Mączyńska, J.; Kulbacka, J. Estrogen receptors in cell membranes: Regulation and signaling. In Transport Across Natural and Modified Biological Membranes and Its Implications in Physiology and Therapy; Springer: Berlin/Heidelberg, Germany, 2017; pp. 93–105. [Google Scholar]
  300. Toyohira, Y.; Ueno, S.; Tsutsui, M.; Itoh, H.; Sakai, N.; Saito, N.; Takahashi, K.; Yanagihara, N. Stimulatory effects of the soy phytoestrogen genistein on noradrenaline transporter and serotonin transporter activity. Mol. Nutr. Food Res. 2010, 54, 516–524. [Google Scholar] [CrossRef]
  301. Woodside, J.V.; Campbell, M.J.; Denholm, E.E.; Newton, L.; Honour, J.W.; Morton, M.S.; Young, I.S.; Leathem, A.J. Short-term phytoestrogen supplementation alters insulin-like growth factor profile but not lipid or antioxidant status. J. Nutr. Biochem. 2006, 17, 211–215. [Google Scholar] [CrossRef]
  302. Teas, J.; Irhimeh, M.R.; Druker, S.; Hurley, T.G.; Hébert, J.R.; Savarese, T.M.; Kurzer, M.S. Serum IGF-1 concentrations change with soy and seaweed supplements in healthy postmenopausal American women. Nutr. Cancer 2011, 63, 743–748. [Google Scholar] [CrossRef] [PubMed]
  303. Bourque, M.; Dluzen, D.E.; Di Paolo, T. Signaling pathways mediating the neuroprotective effects of sex steroids and SERMs in Parkinson’s disease. Front. Neuroendocr. 2012, 33, 169–178. [Google Scholar] [CrossRef]
  304. McKay, D.L.; Blumberg, J.B. A review of the bioactivity of South African herbal teas: Rooibos (Aspalathus linearis) and honeybush (Cyclopia intermedia). Phytother. Res. Int. J. Devoted Pharmacol. Toxicol. Eval. Nat. Prod. Deriv. 2007, 21, 1–16. [Google Scholar] [CrossRef] [PubMed]
  305. Lim, U.; Song, M.-A. Dietary and lifestyle factors of DNA methylation. In Cancer Epigenetics; Springer: Berlin/Heidelberg, Germany, 2012; pp. 359–376. [Google Scholar]
  306. Ming, L.G.; Chen, K.M.; Xian, C.J. Functions and action mechanisms of flavonoids genistein and icariin in regulating bone remodeling. J. Cell. Physiol. 2013, 228, 513–521. [Google Scholar] [CrossRef] [PubMed]
  307. Molina, L.; Bustamante, F.A.; Bhoola, K.D.; Figueroa, C.D.; Ehrenfeld, P. Possible role of phytoestrogens in breast cancer via GPER-1/GPR30 signaling. Clin. Sci. 2018, 132, 2583–2598. [Google Scholar] [CrossRef] [PubMed]
  308. Mabry, T.; Markham, K.R.; Thomas, M.B. The Systematic Identification of Flavonoids; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2012. [Google Scholar]
  309. Sak, K. Cytotoxicity of dietary flavonoids on different human cancer types. Pharmacogn. Rev. 2014, 8, 122. [Google Scholar] [CrossRef] [PubMed]
  310. Mancini, M.C.S.; Ponte, L.G.S.; Silva, C.H.R.; Fagundes, I.; Pavan, I.C.B.; Romeiro, S.A.; da Silva, L.G.S.; Morelli, A.P.; Rostagno, M.A.; Simabuco, F.M.; et al. Beetroot and leaf extracts present protective effects against prostate cancer cells, inhibiting cell proliferation, migration, and growth signaling pathways. Phytother. Res. PTR 2021, 35, 5241–5258. [Google Scholar] [CrossRef] [PubMed]
  311. Chen, X.; Wu, Y.; Wang, X.; Xu, C.; Wang, L.; Jian, J.; Wu, D.; Wu, G. CDK6 is upregulated and may be a potential therapeutic target in enzalutamide-resistant castration-resistant prostate cancer. Eur. J. Med. Res. 2022, 27, 105. [Google Scholar] [CrossRef]
  312. Gupta, S.; Afaq, F.; Mukhtar, H. Selective growth-inhibitory, cell-cycle deregulatory and apoptotic response of apigenin in normal versus human prostate carcinoma cells. Biochem. Biophys. Res. Commun. 2001, 287, 914–920. [Google Scholar] [CrossRef]
  313. Kobayashi, T.; Nakata, T.; Kuzumaki, T. Effect of flavonoids on cell cycle progression in prostate cancer cells. Cancer Lett. 2002, 176, 17–23. [Google Scholar] [CrossRef]
  314. Pham, H.N.T.; Sakoff, J.A.; Vuong, Q.V.; Bowyer, M.C.; Scarlett, C.J. Phytochemical, antioxidant, anti-proliferative and antimicrobial properties of Catharanthus roseus root extract, saponin-enriched and aqueous fractions. Mol. Biol. Rep. 2019, 46, 3265–3273. [Google Scholar] [CrossRef] [PubMed]
  315. Hnit, S.S.T.; Yao, M.; Xie, C.; Bi, L.; Wong, M.; Liu, T.; De Souza, P.; Li, Z.; Dong, Q. Apigenin impedes cell cycle progression at G(2) phase in prostate cancer cells. Discover. Oncol. 2022, 13, 44. [Google Scholar] [CrossRef] [PubMed]
  316. Gupta, S.; Afaq, F.; Mukhtar, H. Involvement of nuclear factor-kappa B, Bax and Bcl-2 in induction of cell cycle arrest and apoptosis by apigenin in human prostate carcinoma cells. Oncogene 2002, 21, 3727–3738. [Google Scholar] [CrossRef] [PubMed]
  317. Shukla, S.; Gupta, S. Molecular mechanisms for apigenin-induced cell-cycle arrest and apoptosis of hormone refractory human prostate carcinoma DU145 cells. Mol. Carcinog. Publ. Coop. Univ. Tex. MD Anderson Cancer Cent. 2004, 39, 114–126. [Google Scholar] [CrossRef] [PubMed]
  318. Shukla, S.; Gupta, S. Molecular targets for apigenin-induced cell cycle arrest and apoptosis in prostate cancer cell xenograft. Mol. Cancer Ther. 2006, 5, 843–852. [Google Scholar] [CrossRef] [PubMed]
  319. Seo, Y.J.; Kim, B.S.; Chun, S.Y.; Park, Y.K.; Kang, K.S.; Kwon, T.G. Apoptotic effects of genistein, biochanin-A and apigenin on LNCaP and PC-3 cells by p21 through transcriptional inhibition of polo-like kinase-1. J. Korean Med. Sci. 2011, 26, 1489–1494. [Google Scholar] [CrossRef]
  320. Shukla, S.; Gupta, S. Suppression of constitutive and tumor necrosis factor-induced nuclear factor (NF)-B activation and induction of apoptosis by apigenin in human prostate carcinoma PC-3 cells: Correlation with down-regulation of NF-B-responsive genes. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2004, 10, 31693178. [Google Scholar] [CrossRef]
  321. Shukla, S.; Gupta, S. Apigenin-induced prostate cancer cell death is initiated by reactive oxygen species and p53 activation. Free Radic. Biol. Med. 2008, 44, 1833–1845. [Google Scholar] [CrossRef] [PubMed]
  322. Shukla, S.; Gupta, S. Apigenin-induced cell cycle arrest is mediated by modulation of MAPK, PI3K-Akt, and loss of cyclin D1 associated retinoblastoma dephosphorylation in human prostate cancer cells. Cell Cycle 2007, 6, 1102–1114. [Google Scholar] [CrossRef]
  323. Shukla, S.; MacLennan, G.T.; Fu, P.; Gupta, S. Apigenin attenuates insulin-like growth factor-I signaling in an autochthonous mouse prostate cancer model. Pharm. Res. 2012, 29, 1506–1517. [Google Scholar] [CrossRef]
  324. Shukla, S.; Gupta, S. Apigenin suppresses insulin-like growth factor I receptor signaling in human prostate cancer: An in vitro and in vivo study. Mol. Carcinog. Publ. Coop. Univ. Tex. MD Anderson Cancer Cent. 2009, 48, 243–252. [Google Scholar] [CrossRef] [PubMed]
  325. Babcook, M.A.; Gupta, S. Apigenin modulates insulin-like growth factor axis: Implications for prevention and therapy of prostate cancer. Curr. Drug Targets 2012. epub ahead of print. [Google Scholar]
  326. Palmieri, D.; Perego, P.; Palombo, D. Apigenin inhibits the TNFα-induced expression of eNOS and MMP-9 via modulating Akt signalling through oestrogen receptor engagement. Mol. Cell. Biochem. 2012, 371, 129–136. [Google Scholar] [CrossRef] [PubMed]
  327. Shukla, S.; Shankar, E.; Fu, P.; MacLennan, G.T.; Gupta, S. Suppression of NF-κB and NF-κB-regulated gene expression by apigenin through IκBα and IKK pathway in TRAMP mice. PLoS ONE 2015, 10, e0138710. [Google Scholar] [CrossRef] [PubMed]
  328. Erdogan, S.; Turkekul, K.; Dibirdik, I.; Doganlar, Z.B.; Doganlar, O.; Bilir, A. Midkine silencing enhances the anti-prostate cancer stem cell activity of the flavone apigenin: Cooperation on signaling pathways regulated by ERK, p38, PTEN, PARP, and NF-κB. Investig. New Drugs 2020, 38, 246–263. [Google Scholar] [CrossRef] [PubMed]
  329. Shukla, S.; Bhaskaran, N.; Babcook, M.A.; Fu, P.; MacLennan, G.T.; Gupta, S. Apigenin inhibits prostate cancer progression in TRAMP mice via targeting PI3K/Akt/FoxO pathway. Carcinogenesis 2014, 35, 452–460. [Google Scholar] [CrossRef]
  330. Kaur, P.; Shukla, S.; Gupta, S. Plant flavonoid apigenin inactivates Akt to trigger apoptosis in human prostate cancer: An in vitro and in vivo study. Carcinogenesis 2008, 29, 2210–2217. [Google Scholar] [CrossRef] [PubMed]
  331. Morrissey, C.; O’Neill, A.; Spengler, B.; Christoffel, V.; Fitzpatrick, J.M.; Watson, R.W.G. Apigenin drives the production of reactive oxygen species and initiates a mitochondrial mediated cell death pathway in prostate epithelial cells. Prostate 2005, 63, 131–142. [Google Scholar] [CrossRef] [PubMed]
  332. Horinaka, M.; Yoshida, T.; Shiraishi, T.; Nakata, S.; Wakada, M.; Sakai, T. The dietary flavonoid apigenin sensitizes malignant tumor cells to tumor necrosis factor–related apoptosis-inducing ligand. Mol. Cancer Ther. 2006, 5, 945–951. [Google Scholar] [CrossRef] [PubMed]
  333. Oishi, M.; Iizumi, Y.; Taniguchi, T.; Goi, W.; Miki, T.; Sakai, T. Apigenin sensitizes prostate cancer cells to Apo2L/TRAIL by targeting adenine nucleotide translocase-2. PLoS ONE 2013, 8, e55922. [Google Scholar] [CrossRef]
  334. Chien, M.-H.; Lin, Y.-W.; Wen, Y.-C.; Yang, Y.-C.; Hsiao, M.; Chang, J.-L.; Huang, H.-C.; Lee, W.-J. Targeting the SPOCK1-snail/slug axis-mediated epithelial-to-mesenchymal transition by apigenin contributes to repression of prostate cancer metastasis. J. Exp. Clin. Cancer Res. 2019, 38, 246. [Google Scholar] [CrossRef]
  335. Franzen, C.A.; Amargo, E.; Todorović, V.; Desai, B.V.; Huda, S.; Mirzoeva, S.; Chiu, K.; Grzybowski, B.A.; Chew, T.-L.; Green, K.J. The chemopreventive bioflavonoid apigenin inhibits prostate cancer cell motility through the focal adhesion kinase/Src signaling mechanism. Cancer Prev. Res. 2009, 2, 830–841. [Google Scholar] [CrossRef] [PubMed]
  336. Shukla, S.; MacLennan, G.T.; Flask, C.A.; Fu, P.; Mishra, A.; Resnick, M.I.; Gupta, S. Blockade of β-catenin signaling by plant flavonoid apigenin suppresses prostate carcinogenesis in TRAMP mice. Cancer Res. 2007, 67, 6925–6935. [Google Scholar] [CrossRef] [PubMed]
  337. Zhu, Y.; Wu, J.; Li, S.; Wang, X.; Liang, Z.; Xu, X.; Xu, X.; Hu, Z.; Lin, Y.; Chen, H. Apigenin inhibits migration and invasion via modulation of epithelial mesenchymal transition in prostate cancer. Mol. Med. Rep. 2015, 11, 1004–1008. [Google Scholar] [CrossRef] [PubMed]
  338. Mirzoeva, S.; Kim, N.D.; Chiu, K.; Franzen, C.A.; Bergan, R.C.; Pelling, J.C. Inhibition of HIF-1 alpha and VEGF expression by the chemopreventive bioflavonoid apigenin is accompanied by Akt inhibition in human prostate carcinoma PC3-M cells. Mol. Carcinog. Publ. Coop. Univ. Tex. MD Anderson Cancer Cent. 2008, 47, 686–700. [Google Scholar] [CrossRef] [PubMed]
  339. Mirzoeva, S.; Franzen, C.A.; Pelling, J.C. Apigenin inhibits TGF-β-induced VEGF expression in human prostate carcinoma cells via a Smad2/3-and Src-dependent mechanism. Mol. Carcinog. 2014, 53, 598–609. [Google Scholar] [CrossRef] [PubMed]
  340. Fang, J.; Zhou, Q.; Liu, L.-Z.; Xia, C.; Hu, X.; Shi, X.; Jiang, B.-H. Apigenin inhibits tumor angiogenesis through decreasing HIF-1α and VEGF expression. Carcinogenesis 2007, 28, 858–864. [Google Scholar] [CrossRef] [PubMed]
  341. Chen, C.; Pore, N.; Behrooz, A.; Ismail-Beigi, F.; Maity, A. Regulation of glut1 mRNA by hypoxia-inducible factor-1. Interaction between H-ras and hypoxia. J. Biol. Chem. 2001, 276, 9519–9525. [Google Scholar] [CrossRef] [PubMed]
  342. Gonzalez-Menendez, P.; Hevia, D.; Rodriguez-Garcia, A.; Mayo, J.C.; Sainz, R.M. Regulation of GLUT transporters by flavonoids in androgen-sensitive and-insensitive prostate cancer cells. Endocrinology 2014, 155, 3238–3250. [Google Scholar] [CrossRef]
  343. Melstrom, L.G.; Salabat, M.R.; Ding, X.Z.; Strouch, M.J.; Grippo, P.J.; Mirzoeva, S.; Pelling, J.C.; Bentrem, D.J. Apigenin down-regulates the hypoxia response genes: HIF-1α, GLUT-1, and VEGF in human pancreatic cancer cells. J. Surg. Res. 2011, 167, 173–181. [Google Scholar] [CrossRef]
  344. Brusselmans, K.; Vrolix, R.; Verhoeven, G.; Swinnen, J.V. Induction of cancer cell apoptosis by flavonoids is associated with their ability to inhibit fatty acid synthase activity. J. Biol. Chem. 2005, 280, 5636–5645. [Google Scholar] [CrossRef]
  345. Van de Sande, T.; Roskams, T.; Lerut, E.; Joniau, S.; Van Poppel, H.; Verhoeven, G.; Swinnen, J.V. High-level expression of fatty acid synthase in human prostate cancer tissues is linked to activation and nuclear localization of Akt/PKB. J. Pathol. 2005, 206, 214–219. [Google Scholar] [CrossRef] [PubMed]
  346. Tsai, C.-H.; Lin, F.-M.; Yang, Y.-C.; Lee, M.-T.; Cha, T.-L.; Wu, G.-J.; Hsieh, S.-C.; Hsiao, P.-W. Herbal extract of Wedelia chinensis attenuates androgen receptor activity and orthotopic growth of prostate cancer in nude mice. Clin. Cancer Res. 2009, 15, 5435–5444. [Google Scholar] [CrossRef] [PubMed]
  347. Tsai, C.-H.; Tzeng, S.-F.; Hsieh, S.-C.; Yang, Y.-C.; Hsiao, Y.-W.; Tsai, M.-H.; Hsiao, P.-W. A standardized herbal extract mitigates tumor inflammation and augments chemotherapy effect of docetaxel in prostate cancer. Sci. Rep. 2017, 7, 15624. [Google Scholar] [CrossRef] [PubMed]
  348. Genc, F.; Atabey, U.S.; Serttas, R.; Erdogan, S. Abiraterone Acetate, in Combination with Apigenin, Attenuates the Survival of Human Castration-Sensitive Prostate Cancer Cells. Anti-Cancer Agents Med. Chem. 2022, 22, 3148–3156. [Google Scholar] [CrossRef] [PubMed]
  349. Ayyildiz, A.; Koc, H.; Turkekul, K.; Erdogan, S. Co-administration of apigenin with doxorubicin enhances anti-migration and antiproliferative effects via PI3K/PTEN/AKT pathway in prostate cancer cells. Exp. Oncol. 2021, 43, 125–134. [Google Scholar] [CrossRef] [PubMed]
  350. Taghavi Bahreghani, M.; Geraily, G.H.; Alizadeh, S.H.; Najafi, M.; Shirazi, A. Apigenin Enhanced Radiation-Induced Apoptosis/Necrosis by Sensitization of LNCaP Prostate Cancer Cells to 6 MV Photon Beams. Cell J. 2021, 23, 730–735. [Google Scholar] [CrossRef] [PubMed]
  351. Parajuli, P.; Joshee, N.; Rimando, A.M.; Mittal, S.; Yadav, A.K. In vitro antitumor mechanisms of various Scutellaria extracts and constituent flavonoids. Planta Medica 2009, 75, 41–48. [Google Scholar] [CrossRef] [PubMed]
  352. Fu, B.; Xue, J.; Li, Z.; Shi, X.; Jiang, B.-H.; Fang, J. Chrysin inhibits expression of hypoxia-inducible factor-1α through reducing hypoxia-inducible factor-1α stability and inhibiting its protein synthesis. Mol. Cancer Ther. 2007, 6, 220–226. [Google Scholar] [CrossRef] [PubMed]
  353. Gresa-Arribas, N.; Serratosa, J.; Saura, J.; Solà, C. Inhibition of CCAAT/enhancer binding protein δ expression by chrysin in microglial cells results in anti-inflammatory and neuroprotective effects. J. Neurochem. 2010, 115, 526–536. [Google Scholar] [CrossRef]
  354. Torres-Piedra, M.; Ortiz-Andrade, R.; Villalobos-Molina, R.; Singh, N.; Medina-Franco, J.L.; Webster, S.P.; Binnie, M.; Navarrete-Vázquez, G.; Estrada-Soto, S. A comparative study of flavonoid analogues on streptozotocin–nicotinamide induced diabetic rats: Quercetin as a potential antidiabetic agent acting via 11β-hydroxysteroid dehydrogenase type 1 inhibition. Eur. J. Med. Chem. 2010, 45, 2606–2612. [Google Scholar] [CrossRef]
  355. Wang, J.; Qiu, J.; Dong, J.; Li, H.; Luo, M.; Dai, X.; Zhang, Y.; Leng, B.; Niu, X.; Zhao, S. Chrysin protects mice from Staphylococcus aureus pneumonia. J. Appl. Microbiol. 2011, 111, 1551–1558. [Google Scholar] [CrossRef] [PubMed]
  356. Du, Q.; Gu, X.; Cai, J.; Huang, M.; Su, M. Chrysin attenuates allergic airway inflammation by modulating the transcription factors T-bet and GATA-3 in mice. Mol. Med. Rep. 2012, 6, 100–104. [Google Scholar] [PubMed]
  357. Anand, K.V.; Mohamed Jaabir, M.S.; Thomas, P.A.; Geraldine, P. Protective role of chrysin against oxidative stress in d-galactose-induced aging in an experimental rat model. Geriatr. Gerontol. Int. 2012, 12, 741–750. [Google Scholar] [CrossRef] [PubMed]
  358. Gano, C.A.; Fatima, S.; Failes, T.W.; Arndt, G.M.; Sajinovic, M.; Mahns, D.; Saedisomeolia, A.; Coorssen, J.R.; Bucci, J.; de Souza, P.; et al. Anti-cancer potential of synergistic phytochemical combinations is influenced by the genetic profile of prostate cancer cell lines. Front. Nutr. 2023, 10, 1119274. [Google Scholar] [CrossRef] [PubMed]
  359. Han, D.H.; Denison, M.S.; Tachibana, H.; Yamada, K. Relationship between estrogen receptor-binding and estrogenic activities of environmental estrogens and suppression by flavonoids. Biosci. Biotechnol. Biochem. 2002, 66, 1479–1487. [Google Scholar] [CrossRef] [PubMed]
  360. Kim, K.M.; Jung, J. Upregulation of G Protein-Coupled Estrogen Receptor by Chrysin-Nanoparticles Inhibits Tumor Proliferation and Metastasis in Triple Negative Breast Cancer Xenograft Model. Front. Endocrinol. 2020, 11, 560605. [Google Scholar] [CrossRef] [PubMed]
  361. Lim, H.K.; Kwon, H.J.; Lee, G.S.; Moon, J.H.; Jung, J. Chrysin-Induced G Protein-Coupled Estrogen Receptor Activation Suppresses Pancreatic Cancer. Int. J. Mol. Sci. 2022, 23, 9673. [Google Scholar] [CrossRef] [PubMed]
  362. Ryu, S.; Lim, W.; Bazer, F.W.; Song, G. Chrysin induces death of prostate cancer cells by inducing ROS and ER stress. J. Cell. Physiol. 2017, 232, 3786–3797. [Google Scholar] [CrossRef]
  363. Samarghandian, S.; Afshari, J.T.; Davoodi, S. Chrysin reduces proliferation and induces apoptosis in the human prostate cancer cell line pc-3. Clinics 2011, 66, 1073–1079. [Google Scholar] [CrossRef]
  364. Kuiper, G.G.; Lemmen, J.G.; Carlsson, B.; Corton, J.C.; Safe, S.H.; van der Saag, P.T.; van der Burg, B.; Gustafsson, J.A. Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology 1998, 139, 4252–4263. [Google Scholar] [CrossRef]
  365. Han, H.; Lee, S.O.; Xu, Y.; Kim, J.E.; Lee, H.J. SPHK/HIF-1α Signaling Pathway Has a Critical Role in Chrysin-Induced Anticancer Activity in Hypoxia-Induced PC-3 Cells. Cells 2022, 11, 2787. [Google Scholar] [CrossRef] [PubMed]
  366. Lim, H.-K.; Kim, K.M.; Jeong, S.-Y.; Choi, E.K.; Jung, J. Chrysin increases the therapeutic efficacy of docetaxel and mitigates docetaxel-induced edema. Integr. Cancer Ther. 2017, 16, 496–504. [Google Scholar] [CrossRef] [PubMed]
  367. Medjakovic, S.; Jungbauer, A. Red clover isoflavones biochanin A and formononetin are potent ligands of the human aryl hydrocarbon receptor. J. Steroid Biochem. Mol. Biol. 2008, 108, 171–177. [Google Scholar] [CrossRef] [PubMed]
  368. Kumar, R.; Verma, V.; Jain, A.; Jain, R.K.; Maikhuri, J.P.; Gupta, G. Synergistic chemoprotective mechanisms of dietary phytoestrogens in a select combination against prostate cancer. J. Nutr. Biochem. 2011, 22, 723–731. [Google Scholar] [CrossRef] [PubMed]
  369. Felix, F.B.; Vago, J.P.; Fernandes, D.O.; Martins, D.G.; Moreira, I.Z.; Gonçalves, W.A.; Costa, W.C.; Araújo, J.M.D.; Queiroz-Junior, C.M.; Campolina-Silva, G.H.; et al. Biochanin A Regulates Key Steps of Inflammation Resolution in a Model of Antigen-Induced Arthritis via GPR30/PKA-Dependent Mechanism. Front. Pharmacol. 2021, 12, 662308. [Google Scholar] [CrossRef] [PubMed]
  370. Rice, L.; Samedi, V.G.; Medrano, T.A.; Sweeney, C.A.; Baker, H.V.; Stenstrom, A.; Furman, J.; Shiverick, K.T. Mechanisms of the growth inhibitory effects of the isoflavonoid biochanin A on LNCaP cells and xenografts. Prostate 2002, 52, 201–212. [Google Scholar] [CrossRef] [PubMed]
  371. Szliszka, E.; Czuba, Z.P.; Mertas, A.; Paradysz, A.; Krol, W. The dietary isoflavone biochanin-A sensitizes prostate cancer cells to TRAIL-induced apoptosis. Urol Oncol 2013, 31, 331–342. [Google Scholar] [CrossRef] [PubMed]
  372. Zand, R.S.R.; Jenkins, D.J.; Brown, T.J.; Diamandis, E.P. Flavonoids can block PSA production by breast and prostate cancer cell lines. Clin. Chim. Acta 2002, 317, 17–26. [Google Scholar] [CrossRef] [PubMed]
  373. Das, D.; Sarkar, S.; Bordoloi, J.; Wann, S.B.; Kalita, J.; Manna, P. Daidzein, its effects on impaired glucose and lipid metabolism and vascular inflammation associated with type 2 diabetes. Biofactors 2018, 44, 407–417. [Google Scholar] [CrossRef]
  374. Handayani, R.; Rice, L.; Cui, Y.; Medrano, T.A.; Samedi, V.G.; Baker, H.V.; Szabo, N.J.; Shiverick, K.T. Soy isoflavones alter expression of genes associated with cancer progression, including interleukin-8, in androgen-independent PC-3 human prostate cancer cells. J. Nutr. 2006, 136, 75–82. [Google Scholar] [CrossRef]
  375. Hsu, A.; Bray, T.M.; Helferich, W.G.; Doerge, D.R.; Ho, E. Differential effects of whole soy extract and soy isoflavones on apoptosis in prostate cancer cells. Exp. Biol. Med. 2010, 235, 90–97. [Google Scholar] [CrossRef] [PubMed]
  376. Onozawa, M.; Fukuda, K.; Ohtani, M.; Akaza, H.; Sugimura, T.; Wakabayashi, K. Effects of soybean isoflavones on cell growth and apoptosis of the human prostatic cancer cell line LNCaP. Jpn. J. Clin. Oncol. 1998, 28, 360–363. [Google Scholar] [CrossRef]
  377. Szliszka, E.; Krol, W. Soy isoflavones augment the effect of TRAIL-mediated apoptotic death in prostate cancer cells. Oncol. Rep. 2011, 26, 533–541. [Google Scholar] [PubMed]
  378. Hempstock, J.; Kavanagh, J.; George, N. Growth inhibition of prostate cell lines in vitro by phyto-oestrogens. Br. J. Urol. 1998, 82, 560–563. [Google Scholar] [CrossRef] [PubMed]
  379. Dong, X.; Xu, W.; Sikes, R.A.; Wu, C. Combination of low dose of genistein and daidzein has synergistic preventive effects on isogenic human prostate cancer cells when compared with individual soy isoflavone. Food Chem. 2013, 141, 1923–1933. [Google Scholar] [CrossRef] [PubMed]
  380. Alonso, V.; Pérez-Martínez, F.; Calahorra, F.; Esbrit, P. Phytoestrogen modulation of bone-related cytokines and its impact on cell viability in human prostate cancer cells. Life Sci. 2009, 85, 421–430. [Google Scholar] [CrossRef] [PubMed]
  381. Wang, S.; DeGroff, V.L.; Clinton, S.K. Tomato and soy polyphenols reduce insulin-like growth factor-I–stimulated rat prostate cancer cell proliferation and apoptotic resistance in vitro via inhibition of intracellular signaling pathways involving tyrosine kinase. J. Nutr. 2003, 133, 2367–2376. [Google Scholar] [CrossRef] [PubMed]
  382. Wang, B.F.; Wang, J.S.; Lu, J.F.; Kao, T.H.; Chen, B.H. Antiproliferation effect and mechanism of prostate cancer cell lines as affected by isoflavones from soybean cake. J. Agric. Food Chem. 2009, 57, 2221–2232. [Google Scholar] [CrossRef] [PubMed]
  383. Ajdžanović, V.; Mojić, M.; Maksimović-Ivanić, D.; Bulatović, M.; Mijatović, S.; Milošević, V.; Spasojević, I. Membrane fluidity, invasiveness and dynamic phenotype of metastatic prostate cancer cells after treatment with soy isoflavones. J. Membr. Biol. 2013, 246, 307–314. [Google Scholar] [CrossRef]
  384. Rabiau, N.; Kossaï, M.; Braud, M.; Chalabi, N.; Satih, S.; Bignon, Y.-J.; Bernard-Gallon, D.J. Genistein and daidzein act on a panel of genes implicated in cell cycle and angiogenesis by polymerase chain reaction arrays in human prostate cancer cell lines. Cancer Epidemiol. 2010, 34, 200–206. [Google Scholar] [CrossRef]
  385. Singh-Gupta, V.; Zhang, H.; Banerjee, S.; Kong, D.; Raffoul, J.J.; Sarkar, F.H.; Hillman, G.G. Radiation-induced HIF-1α cell survival pathway is inhibited by soy isoflavones in prostate cancer cells. Int. J. Cancer 2009, 124, 1675–1684. [Google Scholar] [CrossRef] [PubMed]
  386. Zhao, L.; Mao, Z.; Brinton, R.D. A select combination of clinically relevant phytoestrogens enhances estrogen receptor beta-binding selectivity and neuroprotective activities in vitro and in vivo. Endocrinology 2009, 150, 770–783. [Google Scholar] [CrossRef] [PubMed]
  387. Ranjithkumar, R.; Saravanan, K.; Balaji, B.; Hima, S.; Sreeja, S.; Timane, S.R.; Ram Pravin Kumar, M.; Kabilan, S.; Ramanathan, M. Novel daidzein molecules exhibited anti-prostate cancer activity through nuclear receptor ERβ modulation, in vitro and in vivo studies. J. Chemother. 2021, 33, 582–594. [Google Scholar] [CrossRef] [PubMed]
  388. Kajta, M.; Rzemieniec, J.; Litwa, E.; Lason, W.; Lenartowicz, M.; Krzeptowski, W.; Wojtowicz, A.K. The key involvement of estrogen receptor β and G-protein-coupled receptor 30 in the neuroprotective action of daidzein. Neuroscience 2013, 238, 345–360. [Google Scholar] [CrossRef] [PubMed]
  389. Zhou, C.; Li, P.; Han, M.; Gao, X. Daidzein stimulates fatty acid-induced fat deposition in C2C12 myoblast cells via the G protein-coupled receptor 30 pathway. Anim. Biotechnol. 2022, 33, 851–863. [Google Scholar] [CrossRef] [PubMed]
  390. Dalais, F.S.; Meliala, A.; Wattanapenpaiboon, N.; Frydenberg, M.; Suter, D.A.; Thomson, W.K.; Wahlqvist, M.L. Effects of a diet rich in phytoestrogens on prostate-specific antigen and sex hormones in men diagnosed with prostate cancer. Urology 2004, 64, 510–515. [Google Scholar] [CrossRef] [PubMed]
  391. Landström, M.; Zhang, J.X.; Hallmans, G.; Äman, P.; Bergh, A.; Damber, J.E.; Mazur, W.; Wähälä, K.; Adlercreutz, H. Inhibitory effects of soy and rye diets on the development of Dunning R3327 prostate adenocarcinoma in rats. Prostate 1998, 36, 151–161. [Google Scholar] [CrossRef]
  392. McCormick, D.L.; Johnson, W.D.; Bosland, M.C.; Lubet, R.A.; Steele, V.E. Chemoprevention of rat prostate carcinogenesis by soy isoflavones and by Bowman-Birk inhibitor. Nutr. Cancer 2007, 57, 184–193. [Google Scholar] [CrossRef] [PubMed]
  393. Kato, K.; Takahashi, S.; Cui, L.; Toda, T.; Suzuki, S.; Futakuchi, M.; Sugiura, S.; Shirai, T. Suppressive effects of dietary genistin and daidzin on rat prostate carcinogenesis. Jpn. J. Cancer Res. 2000, 91, 786–791. [Google Scholar] [CrossRef]
  394. Jarred, R.A.; Keikha, M.; Dowling, C.; McPherson, S.J.; Clare, A.M.; Husband, A.J.; Pedersen, J.S.; Frydenberg, M.; Risbridger, G.P. Induction of apoptosis in low to moderate-grade human prostate carcinoma by red clover-derived dietary isoflavones. Cancer Epidemiol. Prev. Biomark. 2002, 11, 1689–1696. [Google Scholar]
  395. Onozawa, M.; Kawamori, T.; Baba, M.; Fukuda, K.; Toda, T.; Sato, H.; Ohtani, M.; Akaza, H.; Sugimura, T.; Wakabayashi, K. Effects of a soybean isoflavone mixture on carcinogenesis in prostate and seminal vesicles of F344 rats. Jpn. J. Cancer Res. 1999, 90, 393–398. [Google Scholar] [CrossRef] [PubMed]
  396. Raffoul, J.J.; Banerjee, S.; Singh-Gupta, V.; Knoll, Z.E.; Fite, A.; Zhang, H.; Abrams, J.; Sarkar, F.H.; Hillman, G.G. Down-regulation of apurinic/apyrimidinic endonuclease 1/redox factor-1 expression by soy isoflavones enhances prostate cancer radiotherapy in vitro and in vivo. Cancer Res. 2007, 67, 2141–2149. [Google Scholar] [CrossRef] [PubMed]
  397. Pendleton, J.M.; Tan, W.W.; Anai, S.; Chang, M.; Hou, W.; Shiverick, K.T.; Rosser, C.J. Phase II trial of isoflavone in prostate-specific antigen recurrent prostate cancer after previous local therapy. BMC Cancer 2008, 8, 132. [Google Scholar] [CrossRef] [PubMed]
  398. Raffoul, J.J.; Banerjee, S.; Che, M.; Knoll, Z.E.; Doerge, D.R.; Abrams, J.; Kucuk, O.; Sarkar, F.H.; Hillman, G.G. Soy isoflavones enhance radiotherapy in a metastatic prostate cancer model. Int. J. Cancer 2007, 120, 2491–2498. [Google Scholar] [CrossRef] [PubMed]
  399. Pollard, M.; Luckert, P. Influence of Isoflavones in Soy Protein Isolates on Development of Induced Prostate-Related Cancers in L-W Rats; Taylor & Francis: Abingdon, UK, 1997. [Google Scholar]
  400. DeVere White, R.W.; Tsodikov, A.; Stapp, E.C.; Soares, S.E.; Fujii, H.; Hackman, R.M. Effects of a high dose, aglycone-rich soy extract on prostate-specific antigen and serum isoflavone concentrations in men with localized prostate cancer. Nutr. Cancer 2010, 62, 1036–1043. [Google Scholar] [CrossRef]
  401. Zhou, J.-R.; Gugger, E.T.; Tanaka, T.; Guo, Y.; Blackburn, G.L.; Clinton, S.K. Soybean phytochemicals inhibit the growth of transplantable human prostate carcinoma and tumor angiogenesis in mice. J. Nutr. 1999, 129, 1628–1635. [Google Scholar] [CrossRef]
  402. Singh-Gupta, V.; Zhang, H.; Yunker, C.K.; Ahmad, Z.; Zwier, D.; Sarkar, F.H.; Hillman, G.G. Daidzein effect on hormone refractory prostate cancer in vitro and in vivo compared to genistein and soy extract: Potentiation of radiotherapy. Pharm. Res. 2010, 27, 1115–1127. [Google Scholar] [CrossRef] [PubMed]
  403. Reger, M.K.; Zollinger, T.W.; Liu, Z.; Jones, J.F.; Zhang, J. Dietary intake of isoflavones and coumestrol and the risk of prostate cancer in the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial. Int. J. Cancer 2018, 142, 719–728. [Google Scholar] [CrossRef] [PubMed]
  404. Miltyk, W.; Craciunescu, C.N.; Fischer, L.; Jeffcoat, R.A.; Koch, M.A.; Lopaczynski, W.; Mahoney, C.; Crowell, J.; Paglieri, J.; Zeisel, S.H. Lack of significant genotoxicity of purified soy isoflavones (genistein, daidzein, and glycitein) in 20 patients with prostate cancer. Am. J. Clin. Nutr. 2003, 77, 875–882. [Google Scholar] [CrossRef]
  405. Wang, X.-L.; Hur, H.-G.; Lee, J.H.; Kim, K.T.; Kim, S.-I. Enantioselective synthesis of S-equol from dihydrodaidzein by a newly isolated anaerobic human intestinal bacterium. Appl. Environ. Microbiol. 2005, 71, 214–219. [Google Scholar] [CrossRef]
  406. Muthyala, R.S.; Ju, Y.H.; Sheng, S.; Williams, L.D.; Doerge, D.R.; Katzenellenbogen, B.S.; Helferich, W.G.; Katzenellenbogen, J.A. Equol, a natural estrogenic metabolite from soy isoflavones: Convenient preparation and resolution of R-and S-equols and their differing binding and biological activity through estrogen receptors alpha and beta. Bioorganic Med. Chem. 2004, 12, 1559–1567. [Google Scholar] [CrossRef] [PubMed]
  407. Setchell, K.D.; Brown, N.M.; Lydeking-Olsen, E. The clinical importance of the metabolite equol—A clue to the effectiveness of soy and its isoflavones. J. Nutr. 2002, 132, 3577–3584. [Google Scholar] [CrossRef]
  408. Decroos, K.; Vanhemmens, S.; Cattoir, S.; Boon, N.; Verstraete, W. Isolation and characterisation of an equol-producing mixed microbial culture from a human faecal sample and its activity under gastrointestinal conditions. Arch. Microbiol. 2005, 183, 45–55. [Google Scholar] [CrossRef] [PubMed]
  409. Setchell, K.D.; Clerici, C.; Lephart, E.D.; Cole, S.J.; Heenan, C.; Castellani, D.; Wolfe, B.E.; Nechemias-Zimmer, L.; Brown, N.M.; Lund, T.D. S-equol, a potent ligand for estrogen receptor β, is the exclusive enantiomeric form of the soy isoflavone metabolite produced by human intestinal bacterial flora. Am. J. Clin. Nutr. 2005, 81, 1072–1079. [Google Scholar] [CrossRef]
  410. Moriyama, M.; Hashimoto, A.; Satoh, H.; Kawabe, K.; Ogawa, M.; Takano, K.; Nakamura, Y. S-Equol, a Major Isoflavone from Soybean, Inhibits Nitric Oxide Production in Lipopolysaccharide-Stimulated Rat Astrocytes Partially via the GPR30-Mediated Pathway. Int. J. Inflamm. 2018, 2018, 8496973. [Google Scholar] [CrossRef]
  411. Rowlands, D.J.; Chapple, S.; Siow, R.C.; Mann, G.E. Equol-stimulated mitochondrial reactive oxygen species activate endothelial nitric oxide synthase and redox signaling in endothelial cells: Roles for F-actin and GPR30. Hypertension 2011, 57, 833–840. [Google Scholar] [CrossRef]
  412. Uehara, M.; Ishimi, Y.; Katsumata, S.-I.; Suzuki, K. Transformation of Daidzein to Equol and Its Bioactivity; ACS Publications: Washington, DC, USA, 2008. [Google Scholar]
  413. Hedlund, T.E.; Johannes, W.U.; Miller, G.J. Soy isoflavonoid equol modulates the growth of benign and malignant prostatic epithelial cells in vitro. Prostate 2003, 54, 68–78. [Google Scholar] [CrossRef] [PubMed]
  414. Hedlund, T.E.; Bokhoven, A.v.; Johannes, W.U.; Nordeen, S.K.; Ogden, L.G. Prostatic fluid concentrations of isoflavonoids in soy consumers are sufficient to inhibit growth of benign and malignant prostatic epithelial cells in vitro. Prostate 2006, 66, 557–566. [Google Scholar] [CrossRef]
  415. Itsumi, M.; Shiota, M.; Takeuchi, A.; Kashiwagi, E.; Inokuchi, J.; Tatsugami, K.; Kajioka, S.; Uchiumi, T.; Naito, S.; Eto, M. Equol inhibits prostate cancer growth through degradation of androgen receptor by S-phase kinase-associated protein 2. Cancer Sci. 2016, 107, 1022–1028. [Google Scholar] [CrossRef]
  416. Mitchell, J.H.; Duthie, S.J.; Collins, A.R. Effects of phytoestrogens on growth and DNA integrity in human prostate tumor cell lines: PC-3 and LNCaP. Nutr. Cancer 2000, 38, 223–228. [Google Scholar] [CrossRef]
  417. Zheng, W.; Zhang, Y.; Ma, D.; Shi, Y.; Liu, C.; Wang, P. (±) Equol inhibits invasion in prostate cancer DU145 cells possibly via down-regulation of matrix metalloproteinase-9, matrix metalloproteinase-2 and urokinase-type plasminogen activator by antioxidant activity. J. Clin. Biochem. Nutr. 2011, 51, 1203270148. [Google Scholar] [CrossRef]
  418. Kurahashi, N.; Iwasaki, M.; Inoue, M.; Sasazuki, S.; Tsugane, S. Plasma isoflavones and subsequent risk of prostate cancer in a nested case-control study: The Japan Public Health Center. J. Clin. Oncol. 2008, 26, 5923–5929. [Google Scholar] [CrossRef] [PubMed]
  419. Sugiyama, Y.; Masumori, N.; Fukuta, F.; Yoneta, A.; Hida, T.; Yamashita, T.; Minatoya, M.; Nagata, Y.; Mori, M.; Tsuji, H. Influence of isoflavone intake and equol-producing intestinal flora on prostate cancer risk. Asian Pac. J. Cancer Prev. 2013, 14, 1–4. [Google Scholar] [CrossRef] [PubMed]
  420. Lund, T.D.; Blake, C.; Bu, L.; Hamaker, A.N.; Lephart, E.D. Equol an isoflavonoid: Potential for improved prostate health, in vitro and in vivoevidence. Reprod. Biol. Endocrinol. 2011, 9, 4. [Google Scholar] [CrossRef]
  421. Liu, Y.; Wu, X.; Jiang, H. High dietary fat intake lowers serum equol concentration and promotes prostate carcinogenesis in a transgenic mouse prostate model. Nutr. Metab. 2019, 16, 24. [Google Scholar] [CrossRef] [PubMed]
  422. Miyanaga, N.; Akaza, H.; Hinotsu, S.; Fujioka, T.; Naito, S.; Namiki, M.; Takahashi, S.; Hirao, Y.; Horie, S.; Tsukamoto, T. Prostate cancer chemoprevention study: An investigative randomized control study using purified isoflavones in men with rising prostate-specific antigen. Cancer Sci. 2012, 103, 125–130. [Google Scholar] [CrossRef]
  423. Battaglia, A.; Devos, G.; Boeckx, G.; Goeman, L.; Tosco, L.; de Meerleer, G.; Joniau, S. Prostate-Specific Antigen Modulatory Effect of a Fermented Soy Supplement for Patients with an Elevated Risk of Prostate Cancer: A Non-Randomized, Retrospective Observational Registration. Curr. Urol. 2020, 14, 142–149. [Google Scholar] [CrossRef]
  424. Mu, H.; Bai, Y.-H.; Wang, S.-T.; Zhu, Z.-M.; Zhang, Y.-W. Research on antioxidant effects and estrogenic effect of formononetin from Trifolium pratense (red clover). Phytomedicine 2009, 16, 314–319. [Google Scholar] [CrossRef]
  425. Li, T.; Zhao, X.; Mo, Z.; Huang, W.; Yan, H.; Ling, Z.; Ye, Y. Formononetin promotes cell cycle arrest via downregulation of Akt/Cyclin D1/CDK4 in human prostate cancer cells. Cell. Physiol. Biochem. 2014, 34, 1351–1358. [Google Scholar] [CrossRef]
  426. Zhang, X.; Bi, L.; Ye, Y.; Chen, J. Formononetin induces apoptosis in PC-3 prostate cancer cells through enhancing the Bax/Bcl-2 ratios and regulating the p38/Akt pathway. Nutr. Cancer 2014, 66, 656–661. [Google Scholar] [CrossRef]
  427. Ye, Y.; Hou, R.; Chen, J.; Mo, L.; Zhang, J.; Huang, Y.; Mo, Z. Formononetin-induced apoptosis of human prostate cancer cells through ERK1/2 mitogen-activated protein kinase inactivation. Horm. Metab. Res. 2012, 44, 263–267. [Google Scholar] [CrossRef] [PubMed]
  428. Huang, W.-J.; Bi, L.-Y.; Li, Z.-Z.; Zhang, X.; Ye, Y. Formononetin induces the mitochondrial apoptosis pathway in prostate cancer cells via downregulation of the IGF-1/IGF-1R signaling pathway. Pharm. Biol. 2014, 52, 466–470. [Google Scholar] [CrossRef] [PubMed]
  429. Liu, X.-J.; Li, Y.-Q.; Chen, Q.-Y.; Xiao, S.-J.; Zeng, S.-E. Up-regulating of RASD1 and apoptosis of DU-145 human prostate cancer cells induced by formononetin in vitro. Asian Pac. J. Cancer Prev. 2014, 15, 2835–2839. [Google Scholar] [CrossRef] [PubMed]
  430. Dong, Z.; Wang, Y.; Guo, J.; Tian, C.; Pan, W.; Wang, H.; Yan, J. Prostate Cancer Therapy Using Docetaxel and Formononetin Combination: Hyaluronic Acid and Epidermal Growth Factor Receptor Targeted Peptide Dual Ligands Modified Binary Nanoparticles to Facilitate the in vivo Anti-Tumor Activity. Drug Des. Dev. Ther. 2022, 16, 2683–2693. [Google Scholar] [CrossRef]
  431. Yuan, W.; Chen, Y.; Zhou, Y.; Bao, K.; Yu, X.; Xu, Y.; Zhang, Y.; Zheng, J.; Jiang, G.; Hong, M. Formononetin attenuates atopic dermatitis by upregulating A20 expression via activation of G protein-coupled estrogen receptor. J. Ethnopharmacol. 2021, 266, 113397. [Google Scholar] [CrossRef] [PubMed]
  432. Chen, J.; Zhao, X.; Ye, Y.; Wang, Y.; Tian, J. Estrogen receptor beta-mediated proliferative inhibition and apoptosis in human breast cancer by calycosin and formononetin. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2013, 32, 1790–1797. [Google Scholar] [CrossRef] [PubMed]
  433. Perabo, F.; Von Löw, E.; Ellinger, J.; Von Rücker, A.; Müller, S.; Bastian, P. Soy isoflavone genistein in prevention and treatment of prostate cancer. Prostate Cancer Prostatic Dis. 2008, 11, 6–12. [Google Scholar] [CrossRef]
  434. Morito, K.; Aomori, T.; Hirose, T.; Kinjo, J.; Hasegawa, J.; Ogawa, S.; Inoue, S.; Muramatsu, M.; Masamune, Y. Interaction of phytoestrogens with estrogen receptors alpha and beta (II). Biol. Pharm. Bull. 2002, 25, 48–52. [Google Scholar] [CrossRef] [PubMed]
  435. Morito, K.; Hirose, T.; Kinjo, J.; Hirakawa, T.; Okawa, M.; Nohara, T.; Ogawa, S.; Inoue, S.; Muramatsu, M.; Masamune, Y. Interaction of phytoestrogens with estrogen receptors alpha and beta. Biol. Pharm. Bull. 2001, 24, 351–356. [Google Scholar] [CrossRef]
  436. Maggiolini, M.; Vivacqua, A.; Fasanella, G.; Recchia, A.G.; Sisci, D.; Pezzi, V.; Montanaro, D.; Musti, A.M.; Picard, D.; Andò, S. The G protein-coupled receptor GPR30 mediates c-fos up-regulation by 17beta-estradiol and phytoestrogens in breast cancer cells. J. Biol. Chem. 2004, 279, 27008–27016. [Google Scholar] [CrossRef]
  437. Vivacqua, A.; Bonofiglio, D.; Albanito, L.; Madeo, A.; Rago, V.; Carpino, A.; Musti, A.M.; Picard, D.; Andò, S.; Maggiolini, M. 17beta-estradiol, genistein, and 4-hydroxytamoxifen induce the proliferation of thyroid cancer cells through the g protein-coupled receptor GPR30. Mol. Pharmacol. 2006, 70, 1414–1423. [Google Scholar] [CrossRef] [PubMed]
  438. Raffoul, J.J.; Wang, Y.; Kucuk, O.; Forman, J.D.; Sarkar, F.H.; Hillman, G.G. Genistein inhibits radiation-induced activation of NF-κB in prostate cancer cells promoting apoptosis and G 2/M cell cycle arrest. BMC Cancer 2006, 6, 107. [Google Scholar] [CrossRef] [PubMed]
  439. Choi, Y.H.; Lee, W.H.; Park, K.Y.; Zhang, L. p53-independent induction of p21 (WAF1/CIP1), reduction of cyclin B1 and G2/M arrest by the isoflavone genistein in human prostate carcinoma cells. Jpn. J. Cancer Res. 2000, 91, 164–173. [Google Scholar] [CrossRef] [PubMed]
  440. Sakamoto, K. Synergistic effects of thearubigin and genistein on human prostate tumor cell (PC-3) growth via cell cycle arrest. Cancer Lett. 2000, 151, 103–109. [Google Scholar] [CrossRef] [PubMed]
  441. Zhao, R.; Xiang, N.; Domann, F.E.; Zhong, W. Effects of selenite and genistein on G2/M cell cycle arrest and apoptosis in human prostate cancer cells. Nutr. Cancer 2009, 61, 397–407. [Google Scholar] [CrossRef] [PubMed]
  442. Ouchi, H.; Ishiguro, H.; Ikeda, N.; Hori, M.; Kubota, Y.; Uemura, H. Genistein induces cell growth inhibition in prostate cancer through the suppression of telomerase activity. Int. J. Urol. 2005, 12, 73–80. [Google Scholar] [CrossRef] [PubMed]
  443. Zhang, L.; Li, L.; Jiao, M.; Wu, D.; Wu, K.; Li, X.; Zhu, G.; Yang, L.; Wang, X.; Hsieh, J.-T. Genistein inhibits the stemness properties of prostate cancer cells through targeting Hedgehog–Gli1 pathway. Cancer Lett. 2012, 323, 48–57. [Google Scholar] [CrossRef] [PubMed]
  444. Hirata, H.; Hinoda, Y.; Shahryari, V.; Deng, G.; Tanaka, Y.; Tabatabai, Z.L.; Dahiya, R. Genistein downregulates onco-miR-1260b and upregulates sFRP1 and Smad4 via demethylation and histone modification in prostate cancer cells. Br. J. Cancer 2014, 110, 1645–1654. [Google Scholar] [CrossRef]
  445. Davis, J.N.; Singh, B.; Bhuiyan, M.; Sarkar, F.H. Genistein-induced upregulation of p21 WAF1, downregulation of cyclin B, and induction of apoptosis in prostate cancer cells. Nutr. Cancer 1998, 32, 123–131. [Google Scholar] [CrossRef]
  446. Farhan, M.; El Oirdi, M.; Aatif, M.; Nahvi, I.; Muteeb, G.; Alam, M.W. Soy Isoflavones Induce Cell Death by Copper-Mediated Mechanism: Understanding Its Anticancer Properties. Molecules 2023, 28, 2925. [Google Scholar] [CrossRef]
  447. Khamesi, S.M.; Salehi Barough, M.; Zargan, J.; Shayesteh, M.; Banaee, N.; Haji Noormohammadi, A.; Keshavarz Alikhani, H.; Mousavi, M. Evaluation of Anticancer and Cytotoxic Effects of Genistein on PC3 Prostate Cell Line under Three-Dimensional Culture Medium. Iran. Biomed. J. 2022, 26, 380–388. [Google Scholar] [CrossRef] [PubMed]
  448. Shafiee, G.; Saidijam, M.; Tayebinia, H.; Khodadadi, I. Beneficial effects of genistein in suppression of proliferation, inhibition of metastasis, and induction of apoptosis in PC3 prostate cancer cells. Arch. Physiol. Biochem. 2022, 128, 694–702. [Google Scholar] [CrossRef] [PubMed]
  449. Tian, J.; Guo, F.; Chen, Y.; Li, Y.; Yu, B.; Li, Y. Nanoliposomal formulation encapsulating celecoxib and genistein inhibiting COX-2 pathway and Glut-1 receptors to prevent prostate cancer cell proliferation. Cancer Lett. 2019, 448, 1–10. [Google Scholar] [CrossRef] [PubMed]
  450. Yu, X.; Yan, J.; Li, Y.; Cheng, J.; Zheng, L.; Fu, T.; Zhu, Y. Inhibition of castration-resistant prostate cancer growth by genistein through suppression of AKR1C3. Food Nutr. Res. 2023, 67. [Google Scholar] [CrossRef] [PubMed]
  451. Li, M.; Zhang, Z.; Hill, D.L.; Chen, X.; Wang, H.; Zhang, R. Genistein, a dietary isoflavone, down-regulates the MDM2 oncogene at both transcriptional and posttranslational levels. Cancer Res. 2005, 65, 8200–8208. [Google Scholar] [CrossRef] [PubMed]
  452. Li, Y.; Sarkar, F.H. Inhibition of nuclear factor κB activation in PC3 cells by genistein is mediated via Akt signaling pathway. Clin. Cancer Res. 2002, 8, 2369–2377. [Google Scholar] [PubMed]
  453. Zhang, Z.; Wang, C.-Z.; Du, G.-J.; Qi, L.-W.; Calway, T.; He, T.-C.; Du, W.; Yuan, C.-S. Genistein induces G2/M cell cycle arrest and apoptosis via ATM/p53-dependent pathway in human colon cancer cells. Int. J. Oncol. 2013, 43, 289–296. [Google Scholar] [CrossRef] [PubMed]
  454. Miękus, K.; Madeja, Z. Genistein inhibits the contact-stimulated migration of prostate cancer cells. Cell. Mol. Biol. Lett. 2007, 12, 348–361. [Google Scholar] [CrossRef] [PubMed]
  455. Huang, X.; Chen, S.; Xu, L.; Liu, Y.; Deb, D.K.; Platanias, L.C.; Bergan, R.C. Genistein inhibits p38 map kinase activation, matrix metalloproteinase type 2, and cell invasion in human prostate epithelial cells. Cancer Res. 2005, 65, 3470–3478. [Google Scholar] [CrossRef]
  456. Kumi-Diaka, J.; Hassanhi, M.; Merchant, K.; Horman, V. Influence of genistein isoflavone on matrix metalloproteinase-2 expression in prostate cancer cells. J. Med. Food 2006, 9, 491–497. [Google Scholar] [CrossRef]
  457. Li, Y.; Che, M.; Bhagat, S.; Ellis, K.-L.; Kucuk, O.; Doerge, D.R.; Abrams, J.; Cher, M.L.; Sarkar, F.H. Regulation of gene expression and inhibition of experimental prostate cancer bone metastasis by dietary genistein. Neoplasia 2004, 6, 354. [Google Scholar] [CrossRef] [PubMed]
  458. Zhang, H.; Gordon, R.; Li, W.; Yang, X.; Pattanayak, A.; Fowler, G.; Zhang, L.; Catalona, W.J.; Ding, Y.; Xu, L. Genistein treatment duration effects biomarkers of cell motility in human prostate. PLoS ONE 2019, 14, e0214078. [Google Scholar] [CrossRef] [PubMed]
  459. Zhang, L.L.; Li, L.; Wu, D.P.; Fan, J.H.; Li, X.; Wu, K.J.; Wang, X.Y.; He, D.L. A novel anti-cancer effect of genistein: Reversal of epithelial mesenchymal transition in prostate cancer cells 1. Acta Pharmacol. Sin. 2008, 29, 1060–1068. [Google Scholar] [CrossRef] [PubMed]
  460. Xu, L.; Ding, Y.; Catalona, W.J.; Yang, X.J.; Anderson, W.F.; Jovanovic, B.; Wellman, K.; Killmer, J.; Huang, X.; Scheidt, K.A. MEK4 function, genistein treatment, and invasion of human prostate cancer cells. JNCI J. Natl. Cancer Inst. 2009, 101, 1141–1155. [Google Scholar] [CrossRef] [PubMed]
  461. Kyle, E.; Neckers, L.; Takimoto, C.; Curt, G.; Bergan, R. Genistein-induced apoptosis of prostate cancer cells is preceded by a specific decrease in focal adhesion kinase activity. Mol. Pharmacol. 1997, 51, 193–200. [Google Scholar] [CrossRef] [PubMed]
  462. Johnson, T.R.; Khandrika, L.; Kumar, B.; Venezia, S.; Koul, S.; Chandhoke, R.; Maroni, P.; Donohue, R.; Meacham, R.B.; Koul, H.K. Focal Adhesion Kinase Controls Aggressive Phenotype of Androgen-Independent Prostate Cancer. Mol. Cancer Res. 2008, 6, 1639–1648. [Google Scholar] [CrossRef] [PubMed]
  463. Guo, Y.; Wang, S.; Hoot, D.R.; Clinton, S.K. Suppression of VEGF-mediated autocrine and paracrine interactions between prostate cancer cells and vascular endothelial cells by soy isoflavones. J. Nutr. Biochem. 2007, 18, 408–417. [Google Scholar] [CrossRef] [PubMed]
  464. Li, Y.; Sarkar, F.H. Down-regulation of invasion and angiogenesis-related genes identified by cDNA microarray analysis of PC3 prostate cancer cells treated with genistein. Cancer Lett. 2002, 186, 157–164. [Google Scholar] [CrossRef] [PubMed]
  465. Davis, J.N.; Kucuk, O.; Sarkar, F.H. Expression of prostate-specific antigen is transcriptionally regulated by genistein in prostate cancer cells. Mol. Carcinog. Publ. Coop. Univ. Tex. MD Anderson Cancer Cent. 2002, 34, 91–101. [Google Scholar] [CrossRef]
  466. Davis, J.; Muqim, N.; Bhuiyan, M.; Kucuk, O.; Pienta, K.; Sarkar, F. Inhibition of prostate specific antigen expression by genistein in prostate cancer cells. Int. J. Oncol. 2000, 16, 1091–1098. [Google Scholar] [CrossRef]
  467. Carlos-Reyes, Á.; López-González, J.S.; Meneses-Flores, M.; Gallardo-Rincón, D.; Ruíz-García, E.; Marchat, L.A.; Astudillo-de la Vega, H.; Hernández de la Cruz, O.N.; López-Camarillo, C. Dietary Compounds as Epigenetic Modulating Agents in Cancer. Front. Genet. 2019, 10, 79. [Google Scholar] [CrossRef] [PubMed]
  468. Mahmoud, A.M.; Al-alem, U.; Ali, M.M.; Bosland, M.C. Genistein increases estrogen receptor beta expression in prostate cancer via reducing its promoter methylation. J. Steroid Biochem. Mol. Biol. 2015, 152, 62–75. [Google Scholar] [CrossRef] [PubMed]
  469. Fang, M.Z.; Chen, D.; Sun, Y.; Jin, Z.; Christman, J.K.; Yang, C.S. Reversal of hypermethylation and reactivation of p16INK4a, RARbeta, and MGMT genes by genistein and other isoflavones from soy. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2005, 11, 7033–7041. [Google Scholar] [CrossRef] [PubMed]
  470. Vardi, A.; Bosviel, R.; Rabiau, N.; Adjakly, M.; Satih, S.; Dechelotte, P.; Boiteux, J.P.; Fontana, L.; Bignon, Y.J.; Guy, L.; et al. Soy phytoestrogens modify DNA methylation of GSTP1, RASSF1A, EPH2 and BRCA1 promoter in prostate cancer cells. In Vivo 2010, 24, 393–400. [Google Scholar] [PubMed]
  471. Majid, S.; Dar, A.A.; Shahryari, V.; Hirata, H.; Ahmad, A.; Saini, S.; Tanaka, Y.; Dahiya, A.V.; Dahiya, R. Genistein reverses hypermethylation and induces active histone modifications in tumor suppressor gene B-Cell translocation gene 3 in prostate cancer. Cancer Interdiscip. Int. J. Am. Cancer Soc. 2010, 116, 66–76. [Google Scholar] [CrossRef] [PubMed]
  472. Chiyomaru, T.; Yamamura, S.; Zaman, M.S.; Majid, S.; Deng, G.; Shahryari, V.; Saini, S.; Hirata, H.; Ueno, K.; Chang, I.; et al. Genistein Suppresses Prostate Cancer Growth through Inhibition of Oncogenic MicroRNA-151. PLoS ONE 2012, 7, e43812. [Google Scholar] [CrossRef] [PubMed]
  473. Chiyomaru, T.; Yamamura, S.; Fukuhara, S.; Hidaka, H.; Majid, S.; Saini, S.; Arora, S.; Deng, G.; Shahryari, V.; Chang, I.; et al. Genistein Up-Regulates Tumor Suppressor MicroRNA-574-3p in Prostate Cancer. PLoS ONE 2013, 8, e58929. [Google Scholar] [CrossRef] [PubMed]
  474. Cai, L.Q.; Cai, J.; Wu, W.; Zhu, Y.S. 17α-Estradiol and genistein inhibit high fat diet induced prostate gene expression and prostate growth in the rat. J. Urol. 2011, 186, 1489–1496. [Google Scholar] [CrossRef] [PubMed]
  475. Wang, J.; Eltoum, I.E.; Carpenter, M.; Lamartiniere, C.A. Genistein mechanisms and timing of prostate cancer chemoprevention in lobund-wistar rats. Asian Pac. J. Cancer Prev. 2009, 10, 143–150. [Google Scholar]
  476. Harper, C.E.; Cook, L.M.; Patel, B.B.; Wang, J.; Eltoum, I.A.; Arabshahi, A.; Shirai, T.; Lamartiniere, C.A. Genistein and resveratrol, alone and in combination, suppress prostate cancer in SV-40 tag rats. Prostate 2009, 69, 1668–1682. [Google Scholar] [CrossRef]
  477. Wang, J.; Eltoum, I.-E.; Lamartiniere, C.A. Dietary genistein suppresses chemically induced prostate cancer in Lobund–Wistar rats. Cancer Lett. 2002, 186, 11–18. [Google Scholar] [CrossRef] [PubMed]
  478. Aronson, W.J.; Tymchuk, C.N.; Elashoff, R.M.; McBride, W.H.; McLean, C.; Wang, H.; Heber, D. Decreased growth of human prostate LNCaP tumors in SCID mice fed a low-fat, soy protein diet with isoflavones. Nutr. Cancer 1999, 35, 130–136. [Google Scholar] [CrossRef] [PubMed]
  479. El Touny, L.H.; Banerjee, P.P. Akt–GSK-3 pathway as a target in genistein-induced inhibition of TRAMP prostate cancer progression toward a poorly differentiated phenotype. Carcinogenesis 2007, 28, 1710–1717. [Google Scholar] [CrossRef] [PubMed]
  480. Mentor-Marcel, R.; Lamartiniere, C.A.; Eltoum, I.A.; Greenberg, N.M.; Elgavish, A. Dietary genistein improves survival and reduces expression of osteopontin in the prostate of transgenic mice with prostatic adenocarcinoma (TRAMP). J. Nutr. 2005, 135, 989–995. [Google Scholar] [CrossRef] [PubMed]
  481. Wang, J.; Eltoum, I.-E.; Lamartiniere, C.A. Genistein chemoprevention of prostate cancer in TRAMP mice. J. Carcinog. 2007, 6, 3. [Google Scholar] [CrossRef] [PubMed]
  482. Mentor-Marcel, R.; Lamartiniere, C.A.; Eltoum, I.-E.; Greenberg, N.M.; Elgavish, A. Genistein in the diet reduces the incidence of poorly differentiated prostatic adenocarcinoma in transgenic mice (TRAMP). Cancer Res. 2001, 61, 6777–6782. [Google Scholar] [PubMed]
  483. Fritz, W.A.; Wang, J.; Eltoum, I.-E.; Lamartiniere, C.A. Dietary genistein down-regulates androgen and estrogen receptor expression in the rat prostate. Mol. Cell. Endocrinol. 2002, 186, 89–99. [Google Scholar] [CrossRef] [PubMed]
  484. Bektic, J.; Berger, A.P.; Pfeil, K.; Dobler, G.; Bartsch, G.; Klocker, H. Androgen receptor regulation by physiological concentrations of the isoflavonoid genistein in androgen-dependent LNCaP cells is mediated by estrogen receptor β. Eur. Urol. 2004, 45, 245–251. [Google Scholar] [CrossRef] [PubMed]
  485. Chung, A.C.; Zhou, S.; Liao, L.; Tien, J.C.; Greenberg, N.M.; Xu, J. Genetic ablation of the amplified-in-breast cancer 1 inhibits spontaneous prostate cancer progression in mice. Cancer Res. 2007, 67, 5965–5975. [Google Scholar] [CrossRef]
  486. Wolk, A.; Mantzoros, C.S.; Andersson, S.O.; Bergström, R.; Signorello, L.B.; Lagiou, P.; Adami, H.O.; Trichopoulos, D. Insulin-like growth factor 1 and prostate cancer risk: A population-based, case-control study. J. Natl. Cancer Inst. 1998, 90, 911–915. [Google Scholar] [CrossRef]
  487. Wu, T.T.; Sikes, R.A.; Cui, Q.; Thalmann, G.N.; Kao, C.; Murphy, C.F.; Yang, H.; Zhau, H.E.; Balian, G.; Chung, L.W. Establishing human prostate cancer cell xenografts in bone: Induction of osteoblastic reaction by prostate-specific antigen-producing tumors in athymic and SCID/bg mice using LNCaP and lineage-derived metastatic sublines. Int. J. Cancer 1998, 77, 887–894. [Google Scholar] [CrossRef]
  488. Thalmann, G.N.; Sikes, R.A.; Devoll, R.E.; Kiefer, J.A.; Markwalder, R.; Klima, I.; Farach-Carson, C.M.; Studer, U.E.; Chung, L.W.K. Osteopontin: Possible Role in Prostate Cancer Progression. Clin. Cancer Res. 1999, 5, 2271–2277. [Google Scholar] [PubMed]
  489. Tuck, A.B.; O’Malley, F.P.; Singhal, H.; Tonkin, K.S.; Harris, J.F.; Bautista, D.; Chambers, A.F. Osteopontin and p53 expression are associated with tumor progression in a case of synchronous, bilateral, invasive mammary carcinomas. Arch. Pathol. Lab. Med. 1997, 121, 578–584. [Google Scholar] [PubMed]
  490. Oates, A.J.; Barraclough, R.; Rudland, P.S. The identification of osteopontin as a metastasis-related gene product in a rodent mammary tumour model. Oncogene 1996, 13, 97–104. [Google Scholar] [PubMed]
  491. Lazarevic, B.; Boezelijn, G.; Diep, L.M.; Kvernrod, K.; Ogren, O.; Ramberg, H.; Moen, A.; Wessel, N.; Berg, R.E.; Egge-Jacobsen, W. Efficacy and safety of short-term genistein intervention in patients with localized prostate cancer prior to radical prostatectomy: A randomized, placebo-controlled, double-blind Phase 2 clinical trial. Nutr. Cancer 2011, 63, 889–898. [Google Scholar] [CrossRef]
  492. Geller, J.; Sionit, L.; Partido, C.; Li, L.; Tan, X.; Youngkin, T.; Nachtsheim, D.; Hoffman, R.M. Genistein inhibits the growth of human-patient BPH and prostate cancer in histoculture. Prostate 1998, 34, 75–79. [Google Scholar] [CrossRef]
  493. Takimoto, C.H.; Glover, K.; Huang, X.; Hayes, S.A.; Gallot, L.; Quinn, M.; Jovanovic, B.D.; Shapiro, A.; Hernandez, L.; Goetz, A. Phase I pharmacokinetic and pharmacodynamic analysis of unconjugated soy isoflavones administered to individuals with cancer. Cancer Epidemiol. Prev. Biomark. 2003, 12, 1213–1221. [Google Scholar]
  494. Jarrard, D.; Konety, B.; Huang, W.; Downs, T.; Kolesar, J.; Kim, K.M.; Havighurst, T.; Slaton, J.; House, M.G.; Parnes, H.L. Phase IIa, randomized placebo-controlled trial of single high dose cholecalciferol (vitamin D3) and daily Genistein (G-2535) versus double placebo in men with early stage prostate cancer undergoing prostatectomy. Am. J. Clin. Exp. Urol. 2016, 4, 17. [Google Scholar] [PubMed]
  495. White, R.W.D.; Hackman, R.M.; Soares, S.E.; Beckett, L.A.; Li, Y.; Sun, B. Effects of a genistein-rich extract on PSA levels in men with a history of prostate cancer. Urology 2004, 63, 259–263. [Google Scholar] [CrossRef]
  496. Bosland, M.C.; Huang, J.; Schlicht, M.J.; Enk, E.; Xie, H.; Kato, I. Impact of 18-Month Soy Protein Supplementation on Steroid Hormones and Serum Biomarkers of Angiogenesis, Apoptosis, and the Growth Hormone/IGF-1 Axis: Results of a Randomized, Placebo-Controlled Trial in Males Following Prostatectomy. Nutr. Cancer 2022, 74, 110–121. [Google Scholar] [CrossRef]
  497. Bosland, M.C.; Schmoll, J.; Watanabe, H.; Randolph, C.; Kato, I. Randomized, Placebo-Controlled Six-Month Intervention Study of Soy Protein Isolate in Men with Biochemical Recurrence after Radical Prostatectomy: A Pilot Study. Nutr. Cancer 2022, 74, 555–564. [Google Scholar] [CrossRef] [PubMed]
  498. Burich, R.A.; Holland, W.S.; Vinall, R.L.; Tepper, C.; DeVere White, R.W.; Mack, P.C. Genistein combined polysaccharide enhances activity of docetaxel, bicalutamide and Src kinase inhibition in androgen-dependent and independent prostate cancer cell lines. BJU Int. 2008, 102, 1458–1466. [Google Scholar] [CrossRef] [PubMed]
  499. Zhang, S.; Wang, Y.; Chen, Z.; Kim, S.; Iqbal, S.; Chi, A.; Ritenour, C.; Wang, Y.A.; Kucuk, O.; Wu, D. Genistein enhances the efficacy of cabazitaxel chemotherapy in metastatic castration-resistant prostate cancer cells. Prostate 2013, 73, 1681–1689. [Google Scholar] [CrossRef] [PubMed]
  500. Shin, S.B.; Woo, S.U.; Yim, H. Cotargeting Plk1 and androgen receptor enhances the therapeutic sensitivity of paclitaxel-resistant prostate cancer. Ther. Adv. Med. Oncol. 2019, 11, 1758835919846375. [Google Scholar] [CrossRef] [PubMed]
  501. Hermann, R.M.; Wolff, H.A.; Jarry, H.; Thelen, P.; Gruendker, C.; Rave-Fraenk, M.; Schmidberger, H.; Christiansen, H. In vitro studies on the modification of low-dose hyper-radiosensitivity in prostate cancer cells by incubation with genistein and estradiol. Radiat. Oncol. 2008, 3, 19. [Google Scholar] [CrossRef]
  502. Hillman, G.G.; Wang, Y.; Kucuk, O.; Che, M.; Doerge, D.R.; Yudelev, M.; Joiner, M.C.; Marples, B.; Forman, J.D.; Sarkar, F.H. Genistein potentiates inhibition of tumor growth by radiation in a prostate cancer orthotopic model. Mol. Cancer Ther. 2004, 3, 1271–1279. [Google Scholar] [CrossRef]
  503. Hillman, G.G.; Forman, J.D.; Kucuk, O.; Yudelev, M.; Maughan, R.L.; Rubio, J.; Layer, A.; Tekyi-Mensah, S.; Abrams, J.; Sarkar, F.H. Genistein potentiates the radiation effect on prostate carcinoma cells. Clin. Cancer Res. 2001, 7, 382–390. [Google Scholar]
  504. Tang, Q.; Ma, J.; Sun, J.; Yang, L.; Yang, F.; Zhang, W.; Li, R.; Wang, L.; Wang, Y.; Wang, H. Genistein and AG1024 synergistically increase the radiosensitivity of prostate cancer cells. Oncol. Rep. 2018, 40, 579–588. [Google Scholar] [PubMed]
  505. Wang, Y.; Raffoul, J.J.; Che, M.; Doerge, D.R.; Joiner, M.C.; Kucuk, O.; Sarkar, F.H.; Hillman, G.G. Prostate cancer treatment is enhanced by genistein in vitro and in vivo in a syngeneic orthotopic tumor model. Radiat. Res. 2006, 166, 73–80. [Google Scholar] [CrossRef]
  506. Jackson, I.L.; Pavlovic, R.; Alexander, A.A.; Connors, C.Q.; Newman, D.; Mahmood, J.; Eley, J.; Harvey, A.J.; Kaytor, M.D.; Vujaskovic, Z. BIO 300, a nanosuspension of genistein, mitigates radiation-induced erectile dysfunction and sensitizes human prostate cancer xenografts to radiation therapy. Int. J. Radiat. Oncol. * Biol. * Phys. 2019, 105, 400–409. [Google Scholar] [CrossRef]
  507. Davis, D.A.; Sarkar, S.H.; Hussain, M.; Li, Y.; Sarkar, F.H. Increased therapeutic potential of an experimental anti-mitotic inhibitor SB715992 by genistein in PC-3 human prostate cancer cell line. BMC Cancer 2006, 6, 22. [Google Scholar] [CrossRef] [PubMed]
  508. Aditya, N.; Shim, M.; Lee, I.; Lee, Y.; Im, M.-H.; Ko, S. Curcumin and genistein coloaded nanostructured lipid carriers: In vitro digestion and antiprostate cancer activity. J. Agric. Food Chem. 2013, 61, 1878–1883. [Google Scholar] [CrossRef] [PubMed]
  509. Song, Y.Y.; Yuan, Y.; Shi, X.; Che, Y.Y. Improved drug delivery and anti-tumor efficacy of combinatorial liposomal formulation of genistein and plumbagin by targeting Glut1 and Akt3 proteins in mice bearing prostate tumor. Colloids Surf. B Biointerfaces 2020, 190, 110966. [Google Scholar] [CrossRef] [PubMed]
  510. Tian, J.Y.; Chi, C.L.; Bian, G.; Xing, D.; Guo, F.J.; Wang, X.Q. PSMA conjugated combinatorial liposomal formulation encapsulating genistein and plumbagin to induce apoptosis in prostate cancer cells. Colloids Surf. B Biointerfaces 2021, 203, 111723. [Google Scholar] [CrossRef] [PubMed]
  511. Wang, G.; Zhang, D.; Yang, S.; Wang, Y.; Tang, Z.; Fu, X. Co-administration of genistein with doxorubicin-loaded polypeptide nanoparticles weakens the metastasis of malignant prostate cancer by amplifying oxidative damage. Biomater. Sci. 2018, 6, 827–835. [Google Scholar] [CrossRef] [PubMed]
  512. Vodnik, V.V.; Mojić, M.; Stamenović, U.; Otoničar, M.; Ajdžanović, V.; Maksimović-Ivanić, D.; Mijatović, S.; Marković, M.M.; Barudžija, T.; Filipović, B.; et al. Development of genistein-loaded gold nanoparticles and their antitumor potential against prostate cancer cell lines. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 124, 112078. [Google Scholar] [CrossRef] [PubMed]
  513. Nakamura, H.; Wang, Y.; Kurita, T.; Adomat, H.; Cunha, G.R.; Wang, Y. Genistein increases epidermal growth factor receptor signaling and promotes tumor progression in advanced human prostate cancer. PLoS ONE 2011, 6, e20034. [Google Scholar] [CrossRef] [PubMed]
  514. El Touny, L.H.; Banerjee, P.P. Genistein induces the metastasis suppressor kangai-1 which mediates its anti-invasive effects in TRAMP cancer cells. Biochem. Biophys. Res. Commun. 2007, 361, 169–175. [Google Scholar] [CrossRef] [PubMed]
  515. Terzioglu-Usak, S.; Yildiz, M.T.; Goncu, B.; Ozten-Kandas, N. Achieving the balance: Biphasic effects of genistein on PC-3 cells. J. Food Biochem. 2019, 43, e12951. [Google Scholar] [CrossRef]
  516. El Touny, L.H.; Banerjee, P.P. Identification of a biphasic role for genistein in the regulation of prostate cancer growth and metastasis. Cancer Res. 2009, 69, 3695–3703. [Google Scholar] [CrossRef]
  517. Mahmoud, A.M.; Zhu, T.; Parray, A.; Siddique, H.R.; Yang, W.; Saleem, M.; Bosland, M.C. Differential effects of genistein on prostate cancer cells depend on mutational status of the androgen receptor. PLoS ONE 2013, 8, e78479. [Google Scholar] [CrossRef] [PubMed]
  518. Totta, P.; Acconcia, F.; Leone, S.; Cardillo, I.; Marino, M. Mechanisms of Naringenin-induced Apoptotic Cascade in Cancer Cells: Involvement of Estrogen Receptor a and ß Signalling. IUBMB Life 2004, 56, 491–499. [Google Scholar] [CrossRef] [PubMed]
  519. Ruh, M.F.; Zacharewski, T.; Connor, K.; Howell, J.; Chen, I.; Safe, S. Naringenin: A weakly estrogenic bioflavonoid that exhibits antiestrogenic activity. Biochem. Pharmacol. 1995, 50, 1485–1493. [Google Scholar] [CrossRef] [PubMed]
  520. Lim, W.; Park, S.; Bazer, F.W.; Song, G. Naringenin-Induced Apoptotic Cell Death in Prostate Cancer Cells Is Mediated via the PI3K/AKT and MAPK Signaling Pathways. J. Cell. Biochem. 2017, 118, 1118–1131. [Google Scholar] [CrossRef] [PubMed]
  521. Torricelli, P.; Elia, A.C.; Magara, G.; Feriotto, G.; Forni, C.; Borromeo, I.; De Martino, A.; Tabolacci, C.; Mischiati, C.; Beninati, S. Reduction of oxidative stress and ornithine decarboxylase expression in a human prostate cancer cell line PC-3 by a combined treatment with α-tocopherol and naringenin. Amino Acids 2021, 53, 63–72. [Google Scholar] [CrossRef] [PubMed]
  522. Aktas, H.G.; Akgun, T. Naringenin inhibits prostate cancer metastasis by blocking voltage-gated sodium channels. Biomed. Pharmacother. 2018, 106, 770–775. [Google Scholar] [CrossRef] [PubMed]
  523. Han, K.-Y.; Chen, P.-N.; Hong, M.-C.; Hseu, Y.-C.; Chen, K.-M.; Hsu, L.-S.; Chen, W.-J. Naringenin Attenuated Prostate Cancer Invasion via Reversal of Epithelial–to–Mesenchymal Transition and Inhibited uPA Activity. Anticancer Res. 2018, 38, 6753–6758. [Google Scholar] [CrossRef] [PubMed]
  524. Gao, K.; Henning, S.M.; Niu, Y.; Youssefian, A.A.; Seeram, N.P.; Xu, A.; Heber, D. The citrus flavonoid naringenin stimulates DNA repair in prostate cancer cells. J. Nutr. Biochem. 2006, 17, 89–95. [Google Scholar] [CrossRef]
  525. Xu, Z.; Huang, B.; Liu, J.; Wu, X.; Luo, N.; Wang, X.; Zheng, X.; Pan, X. Combinatorial anti-proliferative effects of tamoxifen and naringenin: The role of four estrogen receptor subtypes. Toxicology 2018, 410, 231–246. [Google Scholar] [CrossRef]
  526. Calderon-Montano, J.M.; Burgos-Morón, E.; Pérez-Guerrero, C.; López-Lázaro, M. A review on the dietary flavonoid kaempferol. Mini Rev. Med. Chem. 2011, 11, 298–344. [Google Scholar] [CrossRef]
  527. Geybels, M.S.; Verhage, B.A.J.; Arts, I.C.W.; van Schooten, F.J.; Goldbohm, R.A.; van den Brandt, P.A. Dietary Flavonoid Intake, Black Tea Consumption, and Risk of Overall and Advanced Stage Prostate Cancer. Am. J. Epidemiol. 2013, 177, 1388–1398. [Google Scholar] [CrossRef] [PubMed]
  528. Boam, T. Anti-androgenic effects of flavonols in prostate cancer. Ecancermedicalscience 2015, 9, 585. [Google Scholar] [CrossRef] [PubMed]
  529. Da, J.; Xu, M.; Wang, Y.; Li, W.; Lu, M.; Wang, Z. Kaempferol Promotes Apoptosis While Inhibiting Cell Proliferation via Androgen-Dependent Pathway and Suppressing Vasculogenic Mimicry and Invasion in Prostate Cancer. Anal. Cell. Pathol. 2019, 2019, 1907698. [Google Scholar] [CrossRef] [PubMed]
  530. Huang, S.P.; Ho, T.M.; Yang, C.W.; Chang, Y.J.; Chen, J.F.; Shaw, N.S.; Horng, J.C.; Hsu, S.L.; Liao, M.Y.; Wu, L.C.; et al. Chemopreventive Potential of Ethanolic Extracts of Luobuma Leaves (Apocynum venetum L.) in Androgen Insensitive Prostate Cancer. Nutrients 2017, 9, 948. [Google Scholar] [CrossRef] [PubMed]
  531. Mamouni, K.; Zhang, S.; Li, X.; Chen, Y.; Yang, Y.; Kim, J.; Bartlett, M.G.; Coleman, I.M.; Nelson, P.S.; Kucuk, O. A novel flavonoid composition targets androgen receptor signaling and inhibits prostate cancer growth in preclinical models. Neoplasia 2018, 20, 789–799. [Google Scholar] [CrossRef] [PubMed]
  532. Gasmi, J.; Sanderson, J.T. Growth inhibitory, antiandrogenic, and pro-apoptotic effects of punicic acid in LNCaP human prostate cancer cells. J. Agric. Food Chem. 2010, 58, 12149–12156. [Google Scholar] [CrossRef] [PubMed]
  533. Knowles, L.M.; Zigrossi, D.A.; Tauber, R.A.; Hightower, C.; Milner, J.A. Flavonoids suppress androgen-independent human prostate tumor proliferation. Nutr. Cancer 2000, 38, 116–122. [Google Scholar] [CrossRef] [PubMed]
  534. Wang, X.; Zhu, J.; Yan, H.; Shi, M.; Zheng, Q.; Wang, Y.; Zhu, Y.; Miao, L.; Gao, X. Kaempferol inhibits benign prostatic hyperplasia by resisting the action of androgen. Eur. J. Pharmacol. 2021, 907, 174251. [Google Scholar] [CrossRef] [PubMed]
  535. Zhang, Y.; Chen, J.; Fang, W.; Liang, K.; Li, X.; Zhang, F.; Pang, Y.; Fang, G.; Wang, X. Kaempferol suppresses androgen-dependent and androgen-independent prostate cancer by regulating Ki67 expression. Mol. Biol. Rep. 2022, 49, 4607–4617. [Google Scholar] [CrossRef]
  536. Szliszka, E.; Zydowicz, G.; Janoszka, B.; Dobosz, C.; Kowalczyk-Ziomek, G.; Krol, W. Ethanolic extract of Brazilian green propolis sensitizes prostate cancer cells to TRAIL-induced apoptosis. Int. J. Oncol. 2011, 38, 941–953. [Google Scholar]
  537. Halimah, E.; Diantini, A.; Destiani, D.P.; Pradipta, I.S.; Sastramihardja, H.S.; Lestari, K.; Subarnas, A.; Abdulah, R.; Koyama, H. Induction of caspase cascade pathway by kaempferol-3-O-rhamnoside in LNCaP prostate cancer cell lines. Biomed. Rep. 2015, 3, 115–117. [Google Scholar] [CrossRef] [PubMed]
  538. Wang, H.; Gao, M.; Wang, J. Kaempferol inhibits cancer cell growth by antagonizing estrogen-related receptor α and γ activities. Cell Biol. Int. 2013, 37, 1190–1196. [Google Scholar] [CrossRef] [PubMed]
  539. Pang, X.; Fu, W.; Wang, J.; Kang, D.; Xu, L.; Zhao, Y.; Liu, A.L.; Du, G.H. Identification of Estrogen Receptor α Antagonists from Natural Products via In Vitro and In Silico Approaches. Oxid. Med. Cell. Longev. 2018, 2018, 6040149. [Google Scholar] [CrossRef] [PubMed]
  540. Ross, J.A.; Kasum, C.M. Dietary flavonoids: Bioavailability, metabolic effects, and safety. Annu. Rev. Nutr. 2002, 22, 19–34. [Google Scholar] [CrossRef] [PubMed]
  541. Basli, A.; Soulet, S.; Chaher, N.; Mérillon, J.-M.; Chibane, M.; Monti, J.-P.; Richard, T. Wine polyphenols: Potential agents in neuroprotection. Oxidative Med. Cell. Longev. 2012, 2012, 805762. [Google Scholar] [CrossRef] [PubMed]
  542. Ong, K.C.; Khoo, H.-E. Biological effects of myricetin. Gen. Pharmacol. Vasc. Syst. 1997, 29, 121–126. [Google Scholar] [CrossRef] [PubMed]
  543. Xu, R.; Zhang, Y.; Ye, X.; Xue, S.; Shi, J.; Pan, J.; Chen, Q. Inhibition effects and induction of apoptosis of flavonoids on the prostate cancer cell line PC-3 in vitro. Food Chem. 2013, 138, 48–53. [Google Scholar] [CrossRef] [PubMed]
  544. Ye, C.; Zhang, C.; Huang, H.; Yang, B.; Xiao, G.; Kong, D.; Tian, Q.; Song, Q.; Song, Y.; Tan, H. The natural compound myricetin effectively represses the malignant progression of prostate cancer by inhibiting PIM1 and disrupting the PIM1/CXCR4 interaction. Cell. Physiol. Biochem. 2018, 48, 1230–1244. [Google Scholar] [CrossRef] [PubMed]
  545. Nawijn, M.C.; Alendar, A.; Berns, A. For better or for worse: The role of Pim oncogenes in tumorigenesis. Nat. Rev. Cancer 2011, 11, 23–34. [Google Scholar] [CrossRef] [PubMed]
  546. Dhanasekaran, S.M.; Barrette, T.R.; Ghosh, D.; Shah, R.; Varambally, S.; Kurachi, K.; Pienta, K.J.; Rubin, M.A.; Chinnaiyan, A.M. Delineation of prognostic biomarkers in prostate cancer. Nature 2001, 412, 822–826. [Google Scholar] [CrossRef]
  547. Santio, N.M.; Eerola, S.K.; Paatero, I.; Yli-Kauhaluoma, J.; Anizon, F.; Moreau, P.; Tuomela, J.; Härkönen, P.; Koskinen, P.J. Pim Kinases Promote Migration and Metastatic Growth of Prostate Cancer Xenografts. PLoS ONE 2015, 10, e0130340. [Google Scholar] [CrossRef] [PubMed]
  548. Linn, D.E.; Yang, X.; Xie, Y.; Alfano, A.; Deshmukh, D.; Wang, X.; Shimelis, H.; Chen, H.; Li, W.; Xu, K.; et al. Differential regulation of androgen receptor by PIM-1 kinases via phosphorylation-dependent recruitment of distinct ubiquitin E3 ligases. J. Biol. Chem. 2012, 287, 22959–22968. [Google Scholar] [CrossRef] [PubMed]
  549. Santio, N.M.; Salmela, M.; Arola, H.; Eerola, S.K.; Heino, J.; Rainio, E.M.; Koskinen, P.J. The PIM1 kinase promotes prostate cancer cell migration and adhesion via multiple signalling pathways. Exp. Cell Res. 2016, 342, 113–124. [Google Scholar] [CrossRef] [PubMed]
  550. Grundler, R.; Brault, L.; Gasser, C.; Bullock, A.N.; Dechow, T.; Woetzel, S.; Pogacic, V.; Villa, A.; Ehret, S.; Berridge, G.; et al. Dissection of PIM serine/threonine kinases in FLT3-ITD-induced leukemogenesis reveals PIM1 as regulator of CXCL12-CXCR4-mediated homing and migration. J. Exp. Med. 2009, 206, 1957–1970. [Google Scholar] [CrossRef]
  551. Liu, J.S.; Fang, W.K.; Yang, S.M.; Wu, M.C.; Chen, T.J.; Chen, C.M.; Lin, T.Y.; Liu, K.L.; Wu, C.M.; Chen, Y.C.; et al. Natural product myricetin is a pan-KDM4 inhibitor which with poly lactic-co-glycolic acid formulation effectively targets castration-resistant prostate cancer. J. Biomed. Sci. 2022, 29, 29. [Google Scholar] [CrossRef] [PubMed]
  552. Maruthanila, V.L.; Elancheran, R.; Roy, N.K.; Bhattacharya, A.; Kunnumakkara, A.B.; Kabilan, S.; Kotoky, J. In silico Molecular Modelling of Selected Natural Ligands and their Binding Features with Estrogen Receptor Alpha. Curr. Comput. -Aided Drug Des. 2019, 15, 89–96. [Google Scholar] [CrossRef] [PubMed]
  553. Maggiolini, M.; Recchia, A.G.; Bonofiglio, D.; Catalano, S.; Vivacqua, A.; Carpino, A.; Rago, V.; Rossi, R.; Andò, S. The red wine phenolics piceatannol and myricetin act as agonists for estrogen receptor alpha in human breast cancer cells. J. Mol. Endocrinol. 2005, 35, 269–281. [Google Scholar] [CrossRef] [PubMed]
  554. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef]
  555. Lamson, D.W.; Brignall, M. Antioxidants and cancer, part 3: Quercetin. Altern. Med. Rev. A J. Clin. Ther. 2000, 5, 196–208. [Google Scholar]
  556. Bischoff, S.C. Quercetin: Potentials in the prevention and therapy of disease. Curr. Opin. Clin. Nutr. Metab. Care 2008, 11, 733–740. [Google Scholar] [CrossRef]
  557. Murakami, A.; Ashida, H.; Terao, J. Multitargeted cancer prevention by quercetin. Cancer Lett. 2008, 269, 315–325. [Google Scholar] [CrossRef] [PubMed]
  558. Son, S.Y.; Choi, J.H.; Kim, E.B.; Yin, J.; Seonu, S.Y.; Jin, S.Y.; Oh, J.Y.; Lee, M.W. Chemopreventive Activity of Ellagitannins from Acer pseudosieboldianum (Pax) Komarov Leaves on Prostate Cancer Cells. Plants 2023, 12, 1047. [Google Scholar] [CrossRef] [PubMed]
  559. Liu, K.C.; Yen, C.Y.; Wu, R.S.C.; Yang, J.S.; Lu, H.F.; Lu, K.W.; Lo, C.; Chen, H.Y.; Tang, N.Y.; Wu, C.C. The roles of endoplasmic reticulum stress and mitochondrial apoptotic signaling pathway in quercetin-mediated cell death of human prostate cancer PC-3 cells. Environ. Toxicol. 2014, 29, 428–439. [Google Scholar] [CrossRef] [PubMed]
  560. Gopalakrishnan, A.; Xu, C.-J.; Nair, S.S.; Chen, C.; Hebbar, V.; Kong, A.-N.T. Modulation of activator protein-1 (AP-1) and MAPK pathway by flavonoids in human prostate cancer PC3 cells. Arch. Pharmacal Res. 2006, 29, 633. [Google Scholar] [CrossRef] [PubMed]
  561. Morris, J.D.; Pramanik, R.; Zhang, X.; Carey, A.-M.; Ragavan, N.; Martin, F.L.; Muir, G.H. Selenium-or quercetin-induced retardation of DNA synthesis in primary prostate cells occurs in the presence of a concomitant reduction in androgen-receptor activity. Cancer Lett. 2006, 239, 111–122. [Google Scholar] [CrossRef] [PubMed]
  562. Wang, P.; Phan, T.; Gordon, D.; Chung, S.; Henning, S.M.; Vadgama, J.V. Arctigenin in combination with quercetin synergistically enhances the antiproliferative effect in prostate cancer cells. Mol. Nutr. Food Res. 2015, 59, 250–261. [Google Scholar] [CrossRef]
  563. Kampa, M.; Hatzoglou, A.; Notas, G.; Damianaki, A.; Bakogeorgou, E.; Gemetzi, C.; Kouroumalis, E.; Martin, P.-M.; Castanas, E. Wine antioxidant polyphenols inhibit the proliferation of human prostate cancer cell lines. Nutr. Cancer 2000, 37, 223–233. [Google Scholar] [CrossRef] [PubMed]
  564. Wang, P.; Heber, D.; Henning, S.M. Quercetin increased the antiproliferative activity of green tea polyphenol (-)-epigallocatechin gallate in prostate cancer cells. Nutr. Cancer 2012, 64, 580–587. [Google Scholar] [CrossRef] [PubMed]
  565. Huynh, H.; Nguyen, T.T.T.; Chan, E.; Tran, E. Inhibition of ErbB-2 and ErbB-3 expression by quercetin prevents transforming growth factor alpha (TGF-α)-and epidermal growth factor (EGF)-induced human PC-3 prostate cancer cell proliferation. Int. J. Oncol. 2003, 23, 821–829. [Google Scholar] [CrossRef]
  566. Firdous, A.; Sharmila, G.; Balakrishnan, S.; RajaSingh, P.; Suganya, S.; Srinivasan, N.; Arunakaran, J. Quercetin, a natural dietary flavonoid, acts as a chemopreventive agent against prostate cancer in an in vivo model by inhibiting the EGFR signaling pathway. Food Funct. 2014, 5, 2632–2645. [Google Scholar] [CrossRef]
  567. Vijayababu, M.R.; Arunkumar, A.; Kanagaraj, P.; Arunakaran, J. Effects of quercetin on insulin-like growth factors (IGFs) and their binding protein-3 (IGFBP-3) secretion and induction of apoptosis in human prostate cancer cells. J. Carcinog. 2006, 5, 10. [Google Scholar] [CrossRef] [PubMed]
  568. Vijayababu, M.; Kanagaraj, P.; Arunkumar, A.; Ilangovan, R.; Aruldhas, M.; Arunakaran, J. Quercetin-induced growth inhibition and cell death in prostatic carcinoma cells (PC-3) are associated with increase in p21 and hypophosphorylated retinoblastoma proteins expression. J. Cancer Res. Clin. Oncol. 2005, 131, 765–771. [Google Scholar] [CrossRef] [PubMed]
  569. Song, J.; Bai, J.; Wang, S.; Liu, L.; Zhao, Z. Effects of Quercetin on Autophagy and Phosphatidylinositol 3-kinase/Protein Kinase B/Mammalian Target of Rapamycin Signaling Pathway in Human Prostate Cancer PC-3 Cells. Zhongguo Yi Xue Ke Xue Yuan Xue Bao. Acta Acad. Med. Sin. 2020, 42, 578–584. [Google Scholar] [CrossRef]
  570. Yuan, H.; Young, C.Y.; Tian, Y.; Liu, Z.; Zhang, M.; Lou, H. Suppression of the androgen receptor function by quercetin through protein–protein interactions of Sp1, c-Jun, and the androgen receptor in human prostate cancer cells. Mol. Cell. Biochem. 2010, 339, 253–262. [Google Scholar] [CrossRef] [PubMed]
  571. Pang, X.G.; Cong, Y.; Bao, N.R.; Li, Y.G.; Zhao, J.N. Quercetin Stimulates Bone Marrow Mesenchymal Stem Cell Differentiation through an Estrogen Receptor-Mediated Pathway. BioMed Res. Int. 2018, 2018, 4178021. [Google Scholar] [CrossRef] [PubMed]
  572. Jung, Y.-H.; Heo, J.; Lee, Y.J.; Kwon, T.K.; Kim, Y.-H. Quercetin enhances TRAIL-induced apoptosis in prostate cancer cells via increased protein stability of death receptor 5. Life Sci. 2010, 86, 351–357. [Google Scholar] [CrossRef] [PubMed]
  573. Kim, Y.-H.; Lee, D.-H.; Jeong, J.-H.; Guo, Z.S.; Lee, Y.J. Quercetin augments TRAIL-induced apoptotic death: Involvement of the ERK signal transduction pathway. Biochem. Pharmacol. 2008, 75, 1946–1958. [Google Scholar] [CrossRef]
  574. Aalinkeel, R.; Bindukumar, B.; Reynolds, J.L.; Sykes, D.E.; Mahajan, S.D.; Chadha, K.C.; Schwartz, S.A. The dietary bioflavonoid, quercetin, selectively induces apoptosis of prostate cancer cells by down-regulating the expression of heat shock protein 90. Prostate 2008, 68, 1773–1789. [Google Scholar] [CrossRef] [PubMed]
  575. Senthilkumar, K.; Elumalai, P.; Arunkumar, R.; Banudevi, S.; Gunadharini, N.D.; Sharmila, G.; Selvakumar, K.; Arunakaran, J. Quercetin regulates insulin like growth factor signaling and induces intrinsic and extrinsic pathway mediated apoptosis in androgen independent prostate cancer cells (PC-3). Mol. Cell. Biochem. 2010, 344, 173–184. [Google Scholar] [CrossRef]
  576. Vijayababu, M.R.; Kanagaraj, P.; Arunkumar, A.; Ilangovan, R.; Dharmarajan, A.; Arunakaran, J. Quercetin induces p53-independent apoptosis in human prostate cancer cells by modulating Bcl-2-related proteins: A possible mediation by IGFBP-3. Oncol. Res. Featur. Preclin. Clin. Cancer Ther. 2006, 16, 67–74. [Google Scholar] [CrossRef]
  577. Wang, G.; Song, L.; Wang, H.; Xing, N. Quercetin synergizes with 2-methoxyestradiol inhibiting cell growth and inducing apoptosis in human prostate cancer cells. Oncol. Rep. 2013, 30, 357–363. [Google Scholar] [CrossRef] [PubMed]
  578. Lee, D.-H.; Szczepanski, M.; Lee, Y.J. Role of Bax in quercetin-induced apoptosis in human prostate cancer cells. Biochem. Pharmacol. 2008, 75, 2345–2355. [Google Scholar] [CrossRef]
  579. Ward, A.B.; Mir, H.; Kapur, N.; Gales, D.N.; Carriere, P.P.; Singh, S. Quercetin inhibits prostate cancer by attenuating cell survival and inhibiting anti-apoptotic pathways. World J. Surg. Oncol. 2018, 16, 108. [Google Scholar] [CrossRef] [PubMed]
  580. Senthilkumar, K.; Arunkumar, R.; Elumalai, P.; Sharmila, G.; Gunadharini, D.N.; Banudevi, S.; Krishnamoorthy, G.; Benson, C.S.; Arunakaran, J. Quercetin inhibits invasion, migration and signalling molecules involved in cell survival and proliferation of prostate cancer cell line (PC-3). Cell Biochem. Funct. 2011, 29, 87–95. [Google Scholar] [CrossRef] [PubMed]
  581. Lu, X.; Chen, D.; Yang, F.; Xing, N. Quercetin Inhibits Epithelial-to-Mesenchymal Transition (EMT) Process and Promotes Apoptosis in Prostate Cancer via Downregulating lncRNA MALAT1. Cancer Manag. Res. 2020, 12, 1741–1750. [Google Scholar] [CrossRef] [PubMed]
  582. Tang, S.-N.; Singh, C.; Nall, D.; Meeker, D.; Shankar, S.; Srivastava, R.K. The dietary bioflavonoid quercetin synergizes with epigallocathechin gallate (EGCG) to inhibit prostate cancer stem cell characteristics, invasion, migration and epithelial-mesenchymal transition. J. Mol. Signal. 2010, 5, 14. [Google Scholar] [CrossRef] [PubMed]
  583. Tsai, P.-H.; Cheng, C.-H.; Lin, C.-Y.; Huang, Y.-T.; Lee, L.-T.; Kandaswami, C.C.; Lin, Y.-C.; Lee, K.P.-H.; Hung, C.-C.; Hwang, J.-J. Dietary flavonoids luteolin and quercetin suppressed cancer stem cell properties and metastatic potential of isolated prostate cancer cells. Anticancer Res. 2016, 36, 6367–6380. [Google Scholar] [CrossRef]
  584. Zhang, Q.; Cheng, G.; Qiu, H.; Wang, Y.; Wang, J.; Xu, H.; Zhang, T.; Liu, L.; Tao, Y.; Ren, Z. Expression of prostate stem cell antigen is downregulated during flavonoid-induced cytotoxicity in prostate cancer cells. Exp. Ther. Med. 2017, 14, 1795–1801. [Google Scholar] [CrossRef] [PubMed]
  585. Baruah, M.M.; Khandwekar, A.P.; Sharma, N. Quercetin modulates Wnt signaling components in prostate cancer cell line by inhibiting cell viability, migration, and metastases. Tumor Biol. 2016, 37, 14025–14034. [Google Scholar] [CrossRef]
  586. Bhat, F.A.; Sharmila, G.; Balakrishnan, S.; Arunkumar, R.; Elumalai, P.; Suganya, S.; Singh, P.R.; Srinivasan, N.; Arunakaran, J. Quercetin reverses EGF-induced epithelial to mesenchymal transition and invasiveness in prostate cancer (PC-3) cell line via EGFR/PI3K/Akt pathway. J. Nutr. Biochem. 2014, 25, 1132–1139. [Google Scholar] [CrossRef]
  587. Vijayababu, M.; Arunkumar, A.; Kanagaraj, P.; Venkataraman, P.; Krishnamoorthy, G.; Arunakaran, J. Quercetin downregulates matrix metalloproteinases 2 and 9 proteins expression in prostate cancer cells (PC-3). Mol. Cell. Biochem. 2006, 287, 109–116. [Google Scholar] [CrossRef] [PubMed]
  588. Pratheeshkumar, P.; Budhraja, A.; Son, Y.-O.; Wang, X.; Zhang, Z.; Ding, S.; Wang, L.; Hitron, A.; Lee, J.-C.; Xu, M. Quercetin inhibits angiogenesis mediated human prostate tumor growth by targeting VEGFR-2 regulated AKT/mTOR/P70S6K signaling pathways. PLoS ONE 2012, 7, e47516. [Google Scholar] [CrossRef] [PubMed]
  589. Bandyopadhyay, S.; Romero, J.R.; Chattopadhyay, N. Kaempferol and quercetin stimulate granulocyte-macrophage colony-stimulating factor secretion in human prostate cancer cells. Mol. Cell. Endocrinol. 2008, 287, 57–64. [Google Scholar] [CrossRef] [PubMed]
  590. Zhang, X.; Huang, J.; Yu, C.; Xiang, L.; Li, L.; Shi, D.; Lin, F. Quercetin Enhanced Paclitaxel Therapeutic Effects Towards PC-3 Prostate Cancer Through ER Stress Induction and ROS Production. OncoTargets Ther. 2020, 13, 513–523. [Google Scholar] [CrossRef] [PubMed]
  591. Sharma, S.; Cwiklinski, K.; Mahajan, S.D.; Schwartz, S.A.; Aalinkeel, R. Combination Modality Using Quercetin to Enhance the Efficacy of Docetaxel in Prostate Cancer Cells. Cancers 2023, 15, 902. [Google Scholar] [CrossRef] [PubMed]
  592. Omoboyede, V.; Ibrahim, O.; Umar, H.I.; Oke, G.A.; Onile, O.S.; Chukwuemeka, P.O. Computer-aided analysis of quercetin mechanism of overcoming docetaxel resistance in docetaxel-resistant prostate cancer. J. Genet. Eng. Biotechnol. 2023, 21, 47. [Google Scholar] [CrossRef] [PubMed]
  593. Antonarakis, E.S.; Lu, C.; Wang, H.; Luber, B.; Nakazawa, M.; Roeser, J.C.; Chen, Y.; Mohammad, T.A.; Chen, Y.; Fedor, H.L.; et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N. Engl. J. Med. 2014, 371, 1028–1038. [Google Scholar] [CrossRef] [PubMed]
  594. Verma, S.; Prajapati, K.S.; Kushwaha, P.P.; Shuaib, M.; Kumar Singh, A.; Kumar, S.; Gupta, S. Resistance to second generation antiandrogens in prostate cancer: Pathways and mechanisms. Cancer Drug Resist. 2020, 3, 742–761. [Google Scholar] [CrossRef] [PubMed]
  595. Takayama, K.I. Splicing Factors Have an Essential Role in Prostate Cancer Progression and Androgen Receptor Signaling. Biomolecules 2019, 9, 131. [Google Scholar] [CrossRef]
  596. Lu, X.; Yang, F.; Chen, D.; Zhao, Q.; Chen, D.; Ping, H.; Xing, N. Quercetin reverses docetaxel resistance in prostate cancer via androgen receptor and PI3K/Akt signaling pathways. Int. J. Biol. Sci. 2020, 16, 1121–1134. [Google Scholar] [CrossRef]
  597. Chen, D.; Chou, F.J.; Chen, Y.; Huang, C.P.; Tian, H.; Wang, Y.; Niu, Y.; You, B.; Yeh, S.; Xing, N.; et al. Targeting the radiation-induced ARv7-mediated circNHS/miR-512-5p/XRCC5 signaling with Quercetin increases prostate cancer radiosensitivity. J. Exp. Clin. Cancer Res. CR 2022, 41, 235. [Google Scholar] [CrossRef] [PubMed]
  598. Sharmila, G.; Bhat, F.A.; Arunkumar, R.; Elumalai, P.; Raja Singh, P.; Senthilkumar, K.; Arunakaran, J. Chemopreventive effect of quercetin, a natural dietary flavonoid on prostate cancer in in vivo model. Clin. Nutr. 2014, 33, 718–726. [Google Scholar] [CrossRef] [PubMed]
  599. Ma, Z.; Hung Nguyen, T.; Hoa Huynh, T.; Tien Do, P.; Huynh, H. Reduction of rat prostate weight by combined quercetin-finasteride treatment is associated with cell cycle deregulation. J. Endocrinol. 2004, 181, 493–507. [Google Scholar] [CrossRef] [PubMed]
  600. Hemati, M.; Haghiralsadat, F.; Yazdian, F.; Jafari, F.; Moradi, A.; Malekpour-Dehkordi, Z. Development and characterization of a novel cationic PEGylated niosome-encapsulated forms of doxorubicin, quercetin and siRNA for the treatment of cancer by using combination therapy. Artif. Cells Nanomed. Biotechnol. 2019, 47, 1295–1311. [Google Scholar] [CrossRef] [PubMed]
  601. Zhao, J.; Liu, J.; Wei, T.; Ma, X.; Cheng, Q.; Huo, S.; Zhang, C.; Zhang, Y.; Duan, X.; Liang, X.-J. Quercetin-loaded nanomicelles to circumvent human castration-resistant prostate cancer in vitro and in vivo. Nanoscale 2016, 8, 5126–5138. [Google Scholar] [CrossRef] [PubMed]
  602. Shitole, A.A.; Sharma, N.; Giram, P.; Khandwekar, A.; Baruah, M.; Garnaik, B.; Koratkar, S. LHRH-conjugated, PEGylated, poly-lactide-co-glycolide nanocapsules for targeted delivery of combinational chemotherapeutic drugs Docetaxel and Quercetin for prostate cancer. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 114, 111035. [Google Scholar] [CrossRef] [PubMed]
  603. Yaghoubi, F.; Hosseini Motlagh, N.S.; Moradi, A.; Haghiralsadat, F. Carboxylated Graphene Oxide as a Nanocarrier for Drug Delivery of Quercetin as an Effective Anticancer Agent. Iran. Biomed. J. 2022, 26, 324–329. [Google Scholar] [CrossRef] [PubMed]
  604. Essa, D.; Kondiah, P.P.D.; Kumar, P.; Choonara, Y.E. Design of Chitosan-Coated, Quercetin-Loaded PLGA Nanoparticles for Enhanced PSMA-Specific Activity on LnCap Prostate Cancer Cells. Biomedicines 2023, 11, 1201. [Google Scholar] [CrossRef] [PubMed]
  605. Mousavi, N.; Rahimi, S.; Emami, H.; Kazemi, A.H.; Mohammad Taghi Kashi, R.; Heidarian, R. The Effect of Quercetin Nanosuspension on Prostate Cancer Cell Line LNCaP via Hedgehog Signaling Pathway. Rep. Biochem. Mol. Biol. 2021, 10, 69–75. [Google Scholar] [CrossRef]
  606. Castro, C.C.; Pagnussat, A.S.; Orlandi, L.; Worm, P.; Moura, N.; Etgen, A.M.; Netto, C.A. Coumestrol has neuroprotective effects before and after global cerebral ischemia in female rats. Brain Res. 2012, 1474, 82–90. [Google Scholar] [CrossRef]
  607. Wu, X.T.; Wang, B.; Wei, J.N. Coumestrol promotes proliferation and osteoblastic differentiation in rat bone marrow stromal cells. J. Biomed. Mater. Res. Part B Appl. Biomater. Off. J. Soc. Biomater. Jpn. Soc. Biomater. Aust. Soc. Biomater. Korean Soc. Biomater. 2009, 90, 621–628. [Google Scholar] [CrossRef] [PubMed]
  608. Liu, S.; Hsieh, D.; Yang, Y.-L.; Xu, Z.; Peto, C.; Jablons, D.M.; You, L. Coumestrol from the national cancer Institute’s natural product library is a novel inhibitor of protein kinase CK2. BMC Pharmacol. Toxicol. 2013, 14, 36. [Google Scholar] [CrossRef] [PubMed]
  609. Zhao, E.; Mu, Q. Phytoestrogen biological actions on mammalian reproductive system and cancer growth. Sci. Pharm. 2011, 79, 1–20. [Google Scholar] [CrossRef] [PubMed]
  610. Lim, W.; Jeong, M.; Bazer, F.W.; Song, G. Coumestrol inhibits proliferation and migration of prostate cancer cells by regulating AKT, ERK1/2, and JNK MAPK cell signaling cascades. J. Cell. Physiol. 2017, 232, 862–871. [Google Scholar] [CrossRef] [PubMed]
  611. Cho, S.-Y.; Cho, S.; Park, E.; Kim, B.; Sohn, E.J.; Oh, B.; Lee, E.-O.; Lee, H.-J.; Kim, S.-H. Coumestrol suppresses hypoxia inducible factor 1α by inhibiting ROS mediated sphingosine kinase 1 in hypoxic PC-3 prostate cancer cells. Bioorganic Med. Chem. Lett. 2014, 24, 2560–2564. [Google Scholar] [CrossRef] [PubMed]
  612. Spiegel, S.; Milstien, S. Sphingosine-1-phosphate: An enigmatic signalling lipid. Nat. Rev. Mol. Cell Biol. 2003, 4, 397–407. [Google Scholar] [CrossRef] [PubMed]
  613. Hla, T.; Lee, M.J.; Ancellin, N.; Liu, C.H.; Thangada, S.; Thompson, B.D.; Kluk, M. Sphingosine-1-phosphate: Extracellular mediator or intracellular second messenger. Biochem. Pharmacol. 1999, 58, 201–207. [Google Scholar] [CrossRef] [PubMed]
  614. Spiegel, S.; Cuvillier, O.; Edsall, L.C.; Kohama, T.; Menzeleev, R.; Olah, Z.; Olivera, A.; Pirianov, G.; Thomas, D.M.; Tu, Z.; et al. Sphingosine-1-Phosphate in Cell Growth and Cell Deatha. Ann. N. Y. Acad. Sci. 1998, 845, 11–18. [Google Scholar] [CrossRef] [PubMed]
  615. Oskouian, B.; Saba, J. Sphingosine-1-Phosphate Metabolism and Intestinal Tumorigenesis: Lipid Signaling Strikes Again. Cell Cycle 2007, 6, 522–527. [Google Scholar] [CrossRef]
  616. Strom, S.S.; Yamamura, Y.; Duphorne, C.M.; Spitz, M.R.; Babaian, R.J.; Pillow, P.C.; Hursting, S.D. Phytoestrogen intake and prostate cancer: A case-control study using a new database. Nutr. Cancer 1999, 33, 20–25. [Google Scholar] [CrossRef]
  617. Mueller, S.O.; Simon, S.; Chae, K.; Metzler, M.; Korach, K.S. Phytoestrogens and their human metabolites show distinct agonistic and antagonistic properties on estrogen receptor alpha (ERalpha) and ERbeta in human cells. Toxicol. Sci. Off. J. Soc. Toxicol. 2004, 80, 14–25. [Google Scholar] [CrossRef] [PubMed]
  618. Hedelin, M.; Bälter, K.A.; Chang, E.T.; Bellocco, R.; Klint, A.; Johansson, J.E.; Wiklund, F.; Thellenberg-Karlsson, C.; Adami, H.O.; Grönberg, H. Dietary intake of phytoestrogens, estrogen receptor-beta polymorphisms and the risk of prostate cancer. Prostate 2006, 66, 1512–1520. [Google Scholar] [CrossRef] [PubMed]
  619. Athar, M.; Back, J.H.; Tang, X.; Kim, K.H.; Kopelovich, L.; Bickers, D.R.; Kim, A.L. Resveratrol: A review of preclinical studies for human cancer prevention. Toxicol. Appl. Pharmacol. 2007, 224, 274–283. [Google Scholar] [CrossRef] [PubMed]
  620. Baur, J.A.; Sinclair, D.A. Therapeutic potential of resveratrol: The in vivo evidence. Nat. Rev. Drug Discov. 2006, 5, 493–506. [Google Scholar] [CrossRef] [PubMed]
  621. Chakraborty, S.; Levenson, A.S.; Biswas, P.K. Structural insights into Resveratrol’s antagonist and partial agonist actions on estrogen receptor alpha. BMC Struct. Biol. 2013, 13, 27. [Google Scholar] [CrossRef] [PubMed]
  622. Dong, W.H.; Chen, J.C.; He, Y.L.; Xu, J.J.; Mei, Y.A. Resveratrol inhibits K(v)2.2 currents through the estrogen receptor GPR30-mediated PKC pathway. Am. J. Physiol. Cell Physiol. 2013, 305, C547–C557. [Google Scholar] [CrossRef] [PubMed]
  623. Song, T.; Chen, J.; Yang, S.; Liu, B.; Zhang, L.; Zhang, Q.; Cheng, J.C.; Fang, L. Resveratrol stimulates StAR expression and progesterone production by GPER-mediated downregulation of Snail expression in human granulosa cells. J. Food Drug Anal. 2023, 31, 315–325. [Google Scholar] [CrossRef] [PubMed]
  624. Fonseca, J.; Moradi, F.; Maddalena, L.A.; Ferreira-Tollstadius, B.; Selim, S.; Stuart, J.A. Resveratrol integrates metabolic and growth effects in PC3 prostate cancer cells-involvement of prolyl hydroxylase and hypoxia inducible factor-1. Oncol. Lett. 2019, 17, 697–705. [Google Scholar] [CrossRef] [PubMed]
  625. Jang, Y.G.; Go, R.E.; Hwang, K.A.; Choi, K.C. Resveratrol inhibits DHT-induced progression of prostate cancer cell line through interfering with the AR and CXCR4 pathway. J Steroid Biochem. Mol. Biol. 2019, 192, 105406. [Google Scholar] [CrossRef]
  626. Li, J.; Wang, Z.; Li, H.; Cao, J.; Nan, N.; Zhai, X.; Liu, Y.; Chong, T. Resveratrol inhibits TRAF6/PTCH/SMO signal and regulates prostate cancer progression. Cytotechnology 2022, 74, 549–558. [Google Scholar] [CrossRef]
  627. Khusbu, F.Y.; Zhou, X.; Roy, M.; Chen, F.-Z.; Cao, Q.; Chen, H.-C. Resveratrol induces depletion of TRAF6 and suppresses prostate cancer cell proliferation and migration. Int. J. Biochem. Cell Biol. 2020, 118, 105644. [Google Scholar] [CrossRef] [PubMed]
  628. Cheng, T.-M.; Chin, Y.-T.; Ho, Y.; Chen, Y.-R.; Yang, Y.-N.; Yang, Y.-C.; Shih, Y.-J.; Lin, T.-I.; Lin, H.-Y.; Davis, P.J. Resveratrol induces sumoylated COX-2-dependent anti-proliferation in human prostate cancer LNCaP cells. Food Chem. Toxicol. 2018, 112, 67–75. [Google Scholar] [CrossRef] [PubMed]
  629. Hsieh, T.-C. Antiproliferative effects of resveratrol and the mediating role of resveratrol targeting protein NQO2 in androgen receptor-positive, hormone-non-responsive CWR22Rv1 cells. Anticancer Res. 2009, 29, 3011–3017. [Google Scholar] [PubMed]
  630. Hsieh, T.-C.; Wu, J.M. Differential effects on growth, cell cycle arrest, and induction of apoptosis by resveratrol in human prostate cancer cell lines. Exp. Cell Res. 1999, 249, 109–115. [Google Scholar] [CrossRef] [PubMed]
  631. Rashid, A.; Liu, C.; Sanli, T.; Tsiani, E.; Singh, G.; Bristow, R.G.; Dayes, I.; Lukka, H.; Wright, J.; Tsakiridis, T. Resveratrol enhances prostate cancer cell response to ionizing radiation. Modulation of the AMPK, Akt and mTOR pathways. Radiat. Oncol. 2011, 6, 144. [Google Scholar] [CrossRef] [PubMed]
  632. Empl, M.T.; Albers, M.; Wang, S.; Steinberg, P. The Resveratrol Tetramer r-Viniferin Induces a Cell Cycle Arrest Followed by Apoptosis in the Prostate Cancer Cell Line LNCaP. Phytother. Res. 2015, 29, 1640–1645. [Google Scholar] [CrossRef] [PubMed]
  633. Kumar Singh, S.; Banerjee, S.; Acosta, E.P.; Lillard, J.W.; Singh, R. Resveratrol induces cell cycle arrest and apoptosis with docetaxel in prostate cancer cells via a p53/ p21 WAF1/CIP1 and p27 KIP1 pathway. Oncotarget 2017, 8, 17216. [Google Scholar] [CrossRef] [PubMed]
  634. Benitez, D.; Pozo-Guisado, E.; Clementi, M.; Castellón, E.; Fernandez-Salguero, P. Non-genomic action of resveratrol on androgen and oestrogen receptors in prostate cancer: Modulation of the phosphoinositide 3-kinase pathway. Br. J. Cancer 2007, 96, 1595–1604. [Google Scholar] [CrossRef] [PubMed]
  635. Harper, C.E.; Patel, B.B.; Wang, J.; Arabshahi, A.; Eltoum, I.A.; Lamartiniere, C.A. Resveratrol suppresses prostate cancer progression in transgenic mice. Carcinogenesis 2007, 28, 1946–1953. [Google Scholar] [CrossRef] [PubMed]
  636. Benitez, D.A.; Hermoso, M.A.; Pozo-Guisado, E.; Fernández-Salguero, P.M.; Castellón, E.A. Regulation of cell survival by resveratrol involves inhibition of NFκB-regulated gene expression in prostate cancer cells. Prostate 2009, 69, 1045–1054. [Google Scholar] [CrossRef] [PubMed]
  637. Taniguchi, T.; Iizumi, Y.; Watanabe, M.; Masuda, M.; Morita, M.; Aono, Y.; Toriyama, S.; Oishi, M.; Goi, W.; Sakai, T. Resveratrol directly targets DDX5 resulting in suppression of the mTORC1 pathway in prostate cancer. Cell Death Dis. 2016, 7, e2211. [Google Scholar] [CrossRef] [PubMed]
  638. Kumar, S.; Eroglu, E.; Stokes, J.A., III; Scissum-Gunn, K.; Saldanha, S.N.; Singh, U.P.; Manne, U.; Ponnazhagan, S.; Mishra, M.K. Resveratrol induces mitochondria-mediated, caspase-independent apoptosis in murine prostate cancer cells. Oncotarget 2017, 8, 20895. [Google Scholar] [CrossRef] [PubMed]
  639. Wang, D.; Gao, Z.; Zhang, X. Resveratrol induces apoptosis in murine prostate cancer cells via hypoxia-inducible factor 1-alpha (HIF-1α)/reactive oxygen species (ROS)/P53 signaling. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2018, 24, 8970. [Google Scholar] [CrossRef] [PubMed]
  640. Aziz, M.H.; Nihal, M.; Fu, V.X.; Jarrard, D.F.; Ahmad, N. Resveratrol-caused apoptosis of human prostate carcinoma LNCaP cells is mediated via modulation of phosphatidylinositol 3′-kinase/Akt pathway and Bcl-2 family proteins. Mol. Cancer Ther. 2006, 5, 1335–1341. [Google Scholar] [CrossRef] [PubMed]
  641. Benitez, D.A.; Pozo-Guisado, E.; Alvarez-Barrientos, A.; Fernandez-Salguero, P.M.; Castellón, E.A. Mechanisms Involved in Resveratrol-Induced Apoptosis and Cell Cycle Arrest in Prostate Cancer—Derived Cell Lines. J. Androl. 2007, 28, 282–293. [Google Scholar] [CrossRef] [PubMed]
  642. Ferruelo, A.; Romero, I.; Cabrera, P.; Arance, I.; Andrés, G.; Angulo, J. Effects of resveratrol and other wine polyphenols on the proliferation, apoptosis and androgen receptor expression in LNCaP cells. Actas Urológicas Españolas (Engl. Ed.) 2014, 38, 397–404. [Google Scholar] [CrossRef]
  643. Ganapathy, S.; Chen, Q.; Singh, K.P.; Shankar, S.; Srivastava, R.K. Resveratrol enhances antitumor activity of TRAIL in prostate cancer xenografts through activation of FOXO transcription factor. PLoS ONE 2010, 5, e15627. [Google Scholar] [CrossRef] [PubMed]
  644. Shankar, S.; Chen, Q.; Siddiqui, I.; Sarva, K.; Srivastava, R.K. Sensitization of TRAIL-resistant LNCaP cells by resveratrol (3, 4′, 5 tri-hydroxystilbene): Molecular mechanisms and therapeutic potential. J. Mol. Signal. 2007, 2, 7. [Google Scholar] [CrossRef] [PubMed]
  645. Selvaraj, S.; Sun, Y.; Sukumaran, P.; Singh, B.B. Resveratrol activates autophagic cell death in prostate cancer cells via downregulation of STIM1 and the mTOR pathway. Mol. Carcinog. 2016, 55, 818–831. [Google Scholar] [CrossRef] [PubMed]
  646. Sheth, S.; Jajoo, S.; Kaur, T.; Mukherjea, D.; Sheehan, K.; Rybak, L.P.; Ramkumar, V. Resveratrol reduces prostate cancer growth and metastasis by inhibiting the Akt/MicroRNA-21 pathway. PLoS ONE 2012, 7, e51655. [Google Scholar] [CrossRef]
  647. Fraser, S.P.; Peters, A.; Fleming-Jones, S.; Mukhey, D.; Djamgoz, M.B.A. Resveratrol: Inhibitory effects on metastatic cell behaviors and voltage-gated Na+ channel activity in rat prostate cancer in vitro. Nutr. Cancer 2014, 66, 1047–1058. [Google Scholar] [CrossRef] [PubMed]
  648. Wang, K.; Chen, Z.; Shi, J.; Feng, Y.; Yu, M.; Sun, Y.; Zhuang, Q.; Liang, B.; Luo, G.; Xu, X.; et al. Resveratrol inhibits the tumor migration and invasion by upregulating TET1 and reducing TIMP2/3 methylation in prostate carcinoma cells. Prostate 2020, 80, 977–985. [Google Scholar] [CrossRef] [PubMed]
  649. Hsieh, T.C.; Wu, J.M. Resveratrol Suppresses Prostate Cancer Epithelial Cell Scatter/Invasion by Targeting Inhibition of Hepatocyte Growth Factor (HGF) Secretion by Prostate Stromal Cells and Upregulation of E-cadherin by Prostate Cancer Epithelial Cells. Int. J. Mol. Sci. 2020, 21, 1760. [Google Scholar] [CrossRef] [PubMed]
  650. Mitani, T.; Harada, N.; Tanimori, S.; NAkANO, Y.; Inui, H.; Yamaji, R. Resveratrol inhibits hypoxia-inducible factor-1α-mediated androgen receptor signaling and represses tumor progression in castration-resistant prostate cancer. J. Nutr. Sci. Vitaminol. 2014, 60, 276–282. [Google Scholar] [CrossRef] [PubMed]
  651. Lodi, A.; Saha, A.; Lu, X.; Wang, B.; Sentandreu, E.; Collins, M.; Kolonin, M.G.; DiGiovanni, J.; Tiziani, S. Combinatorial treatment with natural compounds in prostate cancer inhibits prostate tumor growth and leads to key modulations of cancer cell metabolism. NPJ Precis. Oncol. 2017, 1, 30. [Google Scholar] [CrossRef] [PubMed]
  652. Saralkar, P.; Dash, A.K. Alginate nanoparticles containing curcumin and resveratrol: Preparation, characterization, and in vitro evaluation against DU145 prostate cancer cell line. AAPS PharmSciTech 2017, 18, 2814–2823. [Google Scholar] [CrossRef] [PubMed]
  653. Sanna, V.; Siddiqui, I.A.; Sechi, M.; Mukhtar, H. Resveratrol-Loaded Nanoparticles Based on Poly (epsilon-caprolactone) and Poly (d, l-lactic-co-glycolic acid)–Poly (ethylene glycol) Blend for Prostate Cancer Treatment. Mol. Pharm. 2013, 10, 3871–3881. [Google Scholar] [CrossRef] [PubMed]
  654. Singh, S.K.; Lillard, J.W., Jr.; Singh, R. Reversal of drug resistance by planetary ball milled (PBM) nanoparticle loaded with resveratrol and docetaxel in prostate cancer. Cancer Lett. 2018, 427, 49–62. [Google Scholar] [CrossRef]
  655. Chaudhary, Z.; Subramaniam, S.; Khan, G.M.; Abeer, M.M.; Qu, Z.; Janjua, T.; Kumeria, T.; Batra, J.; Popat, A. Encapsulation and controlled release of resveratrol within functionalized mesoporous silica nanoparticles for prostate cancer therapy. Front. Bioeng. Biotechnol. 2019, 7, 225. [Google Scholar] [CrossRef]
  656. Thipe, V.C.; Amiri, K.P.; Bloebaum, P.; Karikachery, A.R.; Khoobchandani, M.; Katti, K.K.; Jurisson, S.S.; Katti, K.V. Development of resveratrol-conjugated gold nanoparticles: Interrelationship of increased resveratrol corona on anti-tumor efficacy against breast, pancreatic and prostate cancers. Int. J. Nanomed. 2019, 14, 4413. [Google Scholar] [CrossRef]
  657. Nassir, A.M.; Shahzad, N.; Ibrahim, I.A.; Ahmad, I.; Md, S.; Ain, M.R. Resveratrol-loaded PLGA nanoparticles mediated programmed cell death in prostate cancer cells. Saudi Pharm. J. 2018, 26, 876–885. [Google Scholar] [CrossRef] [PubMed]
  658. Eroglu, E. A Resveratrol-Loaded Poly(2-hydroxyethyl methacrylate)-Chitosan Based Nanotherapeutic: Characterization and In Vitro Cytotoxicity Against Prostate Cancer. J. Nanosci. Nanotechnol. 2021, 21, 2090–2098. [Google Scholar] [CrossRef] [PubMed]
  659. Zhang, L.; Lin, Z.; Chen, Y.; Gao, D.; Wang, P.; Lin, Y.; Wang, Y.; Wang, F.; Han, Y.; Yuan, H. Co-delivery of Docetaxel and Resveratrol by liposomes synergistically boosts antitumor efficiency against prostate cancer. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. 2022, 174, 106199. [Google Scholar] [CrossRef] [PubMed]
  660. Sharma, A.N.; Upadhyay, P.K.; Dewangan, H.K. Development, evaluation, pharmacokinetic and biodistribution estimation of resveratrol-loaded solid lipid nanoparticles for prostate cancer targeting. J. Microencapsul. 2022, 39, 563–574. [Google Scholar] [CrossRef] [PubMed]
  661. Martínez-Martínez, D.; Soto, A.; Gil-Araujo, B.; Gallego, B.; Chiloeches, A.; Lasa, M. Resveratrol promotes apoptosis through the induction of dual specificity phosphatase 1 and sensitizes prostate cancer cells to cisplatin. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2019, 124, 273–279. [Google Scholar] [CrossRef] [PubMed]
  662. Lee, S.H.; Lee, Y.J. Synergistic anticancer activity of resveratrol in combination with docetaxel in prostate carcinoma cells. Nutr. Res. Pract. 2021, 15, 12–25. [Google Scholar] [CrossRef] [PubMed]
  663. Xu, X.; Liu, Q.; Li, J.; Xiao, M.; Gao, T.; Zhang, X.; Lu, G.; Wang, J.; Guo, Y.; Wen, P.; et al. Co-Treatment With Resveratrol and FGF1 Protects Against Acute Liver Toxicity After Doxorubicin Treatment via the AMPK/NRF2 Pathway. Front. Pharmacol. 2022, 13, 940406. [Google Scholar] [CrossRef] [PubMed]
  664. Scarlatti, F.; Sala, G.; Ricci, C.; Maioli, C.; Milani, F.; Minella, M.; Botturi, M.; Ghidoni, R. Resveratrol sensitization of DU145 prostate cancer cells to ionizing radiation is associated to ceramide increase. Cancer Lett. 2007, 253, 124–130. [Google Scholar] [CrossRef] [PubMed]
  665. Fang, Y.; DeMarco, V.G.; Nicholl, M.B. Resveratrol enhances radiation sensitivity in prostate cancer by inhibiting cell proliferation and promoting cell senescence and apoptosis. Cancer Sci. 2012, 103, 1090–1098. [Google Scholar] [CrossRef]
  666. Fang, Y.; Herrick, E.J.; Nicholl, M.B. A Possible Role for Perforin and Granzyme B in Resveratrol-Enhanced Radiosensitivity of Prostate Cancer. J. Androl. 2012, 33, 752–760. [Google Scholar] [CrossRef]
  667. Chen, Y.-A.; Lien, H.-M.; Kao, M.-C.; Lo, U.-G.; Lin, L.-C.; Lin, C.-J.; Chang, S.-J.; Chen, C.-C.; Hsieh, J.-T.; Lin, H. Sensitization of radioresistant prostate cancer cells by resveratrol isolated from arachis hypogaea stems. PLoS ONE 2017, 12, e0169204. [Google Scholar] [CrossRef] [PubMed]
  668. El-Benhawy, S.A.; Morsi, M.I.; Fahmy, E.I.; Soula, M.A.; Khalil, F.; Arab, A.R. Role of Resveratrol as Radiosensitizer by Targeting Cancer Stem Cells in Radioresistant Prostate Cancer Cells (PC-3). Asian Pac. J. Cancer Prev. 2021, 22, 3823–3837. [Google Scholar] [CrossRef] [PubMed]
  669. Paller, C.J.; Heath, E.I.; Taplin, M.-E.; Stein, M.N.; Bubley, G.J.; Pili, R.; Mayer, T.M.; Zhou, X.C.; Hudson, T.; Abbas, M.; et al. A phase II study of muscadine grape skin extract in men with biochemically recurrent prostate cancer. J. Clin. Oncol. 2017, 35, 248. [Google Scholar] [CrossRef]
  670. Sautour, M.; Mitaine-Offer, A.-C.; Lacaille-Dubois, M.-A. The Dioscorea genus: A review of bioactive steroid saponins. J. Nat. Med. 2007, 61, 91–101. [Google Scholar] [CrossRef]
  671. Djerassi, C.; Rosenkranz, G.; Pataki, J.; Kaufmann, S. Steroids, XXVII. Synthesis of allopregnane-3beta, 11beta, 17alpha-, 20beta, 21-pentol from cortisone and diosgenin. J. Biol. Chem. 1952, 194, 115–118. [Google Scholar] [CrossRef] [PubMed]
  672. Shah, H.J.L.S. Extraction of Diosgenin, a Bioactive Compound from Natural Source Dioscorea alata Var purpurae. J. Anal. Bioanal. Tech. 2012, 3, 141. [Google Scholar] [CrossRef]
  673. Król-Kogus, B.; Lamine, K.M.; Migas, P.; Boudjeniba, M.; Krauze-Baranowska, M. HPTLC determination of diosgenin in fenugreek seeds. Acta Pharm. 2018, 68, 97–107. [Google Scholar] [CrossRef]
  674. Chaudhary, S.A.; Chaudhary, P.S.; Syed, B.A.; Misra, R.; Bagali, P.G.; Vitalini, S.; Iriti, M. Validation of a method for diosgenin extraction from fenugreek (Trigonella foenum-graecum L.). Acta Sci. Pol. Technol. Aliment. 2018, 17, 377–385. [Google Scholar] [CrossRef]
  675. Selim, S.; Al Jaouni, S. Anticancer and apoptotic effects on cell proliferation of diosgenin isolated from Costus speciosus (Koen.) Sm. BMC Complement. Altern. Med. 2015, 15, 301. [Google Scholar] [CrossRef]
  676. Obidiegwu, J.E.; Lyons, J.B.; Chilaka, C.A. The Dioscorea Genus (Yam)-An Appraisal of Nutritional and Therapeutic Potentials. Foods 2020, 9, 1304. [Google Scholar] [CrossRef]
  677. Liagre, B.; Bertrand, J.; Leger, D.Y.; Beneytout, J.L. Diosgenin, a plant steroid, induces apoptosis in COX-2 deficient K562 cells with activation of the p38 MAP kinase signalling and inhibition of NF-kappaB binding. Int. J. Mol. Med. 2005, 16, 1095–1101. [Google Scholar] [CrossRef] [PubMed]
  678. Leger, D.Y.; Liagre, B.; Corbière, C.; Cook-Moreau, J.; Beneytout, J.L. Diosgenin induces cell cycle arrest and apoptosis in HEL cells with increase in intracellular calcium level, activation of cPLA2 and COX-2 overexpression. Int. J. Oncol. 2004, 25, 555–562. [Google Scholar] [CrossRef] [PubMed]
  679. Liu, M.J.; Wang, Z.; Ju, Y.; Wong, R.N.; Wu, Q.Y. Diosgenin induces cell cycle arrest and apoptosis in human leukemia K562 cells with the disruption of Ca2+ homeostasis. Cancer Chemother. Pharmacol. 2005, 55, 79–90. [Google Scholar] [CrossRef] [PubMed]
  680. Jiang, S.; Fan, J.; Wang, Q.; Ju, D.; Feng, M.; Li, J.; Guan, Z.B.; An, D.; Wang, X.; Ye, L. Diosgenin induces ROS-dependent autophagy and cytotoxicity via mTOR signaling pathway in chronic myeloid leukemia cells. Phytomedicine 2016, 23, 243–252. [Google Scholar] [CrossRef] [PubMed]
  681. Hu, M.; Xu, L.; Yin, L.; Qi, Y.; Li, H.; Xu, Y.; Han, X.; Peng, J.; Wan, X. Cytotoxicity of dioscin in human gastric carcinoma cells through death receptor and mitochondrial pathways. J. Appl. Toxicol. JAT 2013, 33, 712–722. [Google Scholar] [CrossRef] [PubMed]
  682. Zhao, X.; Xu, L.; Zheng, L.; Yin, L.; Qi, Y.; Han, X.; Xu, Y.; Peng, J. Potent effects of dioscin against gastric cancer in vitro and in vivo. Phytomedicine 2016, 23, 274–282. [Google Scholar] [CrossRef] [PubMed]
  683. Rahmati-Yamchi, M.; Ghareghomi, S.; Haddadchi, G.; Milani, M.; Aghazadeh, M.; Daroushnejad, H. Fenugreek extract diosgenin and pure diosgenin inhibit the hTERT gene expression in A549 lung cancer cell line. Mol. Biol. Rep. 2014, 41, 6247–6252. [Google Scholar] [CrossRef] [PubMed]
  684. Xu, L.; Xu, D.; Li, Z.; Gao, Y.; Chen, H. Synthesis and potent cytotoxic activity of a novel diosgenin derivative and its phytosomes against lung cancer cells. Beilstein J. Nanotechnol. 2019, 10, 1933–1942. [Google Scholar] [CrossRef]
  685. Mohammad, R.Y.; Somayyeh, G.; Gholamreza, H.; Majid, M.; Yousef, R. Diosgenin inhibits hTERT gene expression in the A549 lung cancer cell line. Asian Pac. J. Cancer Prev. 2013, 14, 6945–6948. [Google Scholar] [CrossRef]
  686. Srinivasan, S.; Koduru, S.; Kumar, R.; Venguswamy, G.; Kyprianou, N.; Damodaran, C. Diosgenin targets Akt-mediated prosurvival signaling in human breast cancer cells. Int. J. Cancer 2009, 125, 961–967. [Google Scholar] [CrossRef]
  687. He, Z.; Chen, H.; Li, G.; Zhu, H.; Gao, Y.; Zhang, L.; Sun, J. Diosgenin inhibits the migration of human breast cancer MDA-MB-231 cells by suppressing Vav2 activity. Phytomedicine 2014, 21, 871–876. [Google Scholar] [CrossRef] [PubMed]
  688. Li, F.; Fernandez, P.P.; Rajendran, P.; Hui, K.M.; Sethi, G. Diosgenin, a steroidal saponin, inhibits STAT3 signaling pathway leading to suppression of proliferation and chemosensitization of human hepatocellular carcinoma cells. Cancer Lett. 2010, 292, 197–207. [Google Scholar] [CrossRef] [PubMed]
  689. Chen, Z.; Xu, J.; Wu, Y.; Lei, S.; Liu, H.; Meng, Q.; Xia, Z. Diosgenin inhibited the expression of TAZ in hepatocellular carcinoma. Biochem. Biophys. Res. Commun. 2018, 503, 1181–1185. [Google Scholar] [CrossRef] [PubMed]
  690. Yu, H.; Liu, Y.; Niu, C.; Cheng, Y. Diosgenin increased DDX3 expression in hepatocellular carcinoma. Am. J. Transl. Res. 2018, 10, 3590–3599. [Google Scholar] [PubMed]
  691. Sethi, G.; Shanmugam, M.K.; Warrier, S.; Merarchi, M.; Arfuso, F.; Kumar, A.P.; Bishayee, A. Pro-Apoptotic and Anti-Cancer Properties of Diosgenin: A Comprehensive and Critical Review. Nutrients 2018, 10, 645. [Google Scholar] [CrossRef] [PubMed]
  692. Malisetty, V.S.; Patlolla, J.M.R.; Raju, J.; Marcus, L.A.; Choi, C.-I.; Rao, C.V. Chemoprevention of colon cancer by diosgenin, a steroidal saponin constituent of fenugreek. Cancer Res. 2005, 65, 580. [Google Scholar]
  693. Lepage, C.; Léger, D.Y.; Bertrand, J.; Martin, F.; Beneytout, J.L.; Liagre, B. Diosgenin induces death receptor-5 through activation of p38 pathway and promotes TRAIL-induced apoptosis in colon cancer cells. Cancer Lett. 2011, 301, 193–202. [Google Scholar] [CrossRef] [PubMed]
  694. Aggarwal, B.; Prasad, S.; Sung, B.; Krishnan, S.; Guha, S. Prevention and Treatment of Colorectal Cancer by Natural Agents From Mother Nature. Curr. Color. Cancer Rep. 2013, 9, 37–56. [Google Scholar] [CrossRef] [PubMed]
  695. Guo, W.; Chen, Y.; Gao, J.; Zhong, K.; Wei, H.; Li, K.; Tang, M.; Zhao, X.; Liu, X.; Nie, C.; et al. Diosgenin exhibits tumor suppressive function via down-regulation of EZH2 in pancreatic cancer cells. Cell Cycle 2019, 18, 1745–1758. [Google Scholar] [CrossRef]
  696. Long, C.; Chen, J.; Zhou, H.; Jiang, T.; Fang, X.; Hou, D.; Liu, P.; Duan, H. Diosgenin exerts its tumor suppressive function via inhibition of Cdc20 in osteosarcoma cells. Cell Cycle 2019, 18, 346–358. [Google Scholar] [CrossRef]
  697. Hernández-Vázquez, J.M.V.; López-Muñoz, H.; Escobar-Sánchez, M.L.; Flores-Guzmán, F.; Weiss-Steider, B.; Hilario-Martínez, J.C.; Sandoval-Ramírez, J.; Fernández-Herrera, M.A.; Sánchez Sánchez, L. Apoptotic, necrotic, and antiproliferative activity of diosgenin and diosgenin glycosides on cervical cancer cells. Eur. J. Pharmacol. 2020, 871, 172942. [Google Scholar] [CrossRef] [PubMed]
  698. Corbiere, C.; Liagre, B.; Terro, F.; Beneytout, J.L. Induction of antiproliferative effect by diosgenin through activation of p53, release of apoptosis-inducing factor (AIF) and modulation of caspase-3 activity in different human cancer cells. Cell Res. 2004, 14, 188–196. [Google Scholar] [CrossRef] [PubMed]
  699. Das, S.; Dey, K.K.; Dey, G.; Pal, I.; Majumder, A.; MaitiChoudhury, S.; Kundu, S.C.; Mandal, M. Antineoplastic and apoptotic potential of traditional medicines thymoquinone and diosgenin in squamous cell carcinoma. PLoS ONE 2012, 7, e46641. [Google Scholar] [CrossRef] [PubMed]
  700. Pons-Fuster López, E.; Wang, Q.T.; Wei, W.; López Jornet, P. Potential chemotherapeutic effects of diosgenin, zoledronic acid and epigallocatechin-3-gallate on PE/CA-PJ15 oral squamous cancer cell line. Arch. Oral Biol. 2017, 82, 141–146. [Google Scholar] [CrossRef] [PubMed]
  701. Chen, P.S.; Shih, Y.W.; Huang, H.C.; Cheng, H.W. Diosgenin, a steroidal saponin, inhibits migration and invasion of human prostate cancer PC-3 cells by reducing matrix metalloproteinases expression. PLoS ONE 2011, 6, e20164. [Google Scholar] [CrossRef] [PubMed]
  702. Nie, C.; Zhou, J.; Qin, X.; Shi, X.; Zeng, Q.; Liu, J.; Yan, S.; Zhang, L. Diosgenin-induced autophagy and apoptosis in a human prostate cancer cell line. Mol. Med. Rep. 2016, 14, 4349–4359. [Google Scholar] [CrossRef] [PubMed]
  703. Sun, G.C.; Jan, C.R.; Liang, W.Z. Exploring the impact of a naturally occurring sapogenin diosgenin on underlying mechanisms of Ca(2+) movement and cytotoxicity in human prostate cancer cells. Environ. Toxicol. 2020, 35, 395–403. [Google Scholar] [CrossRef] [PubMed]
  704. Zhang, J.; Xie, J.J.; Zhou, S.J.; Chen, J.; Hu, Q.; Pu, J.X.; Lu, J.L. Diosgenin inhibits the expression of NEDD4 in prostate cancer cells. Am. J. Transl. Res. 2019, 11, 3461–3471. [Google Scholar] [PubMed]
  705. Shabbeer, S.; Sobolewski, M.; Anchoori, R.K.; Kachhap, S.; Hidalgo, M.; Jimeno, A.; Davidson, N.; Carducci, M.A.; Khan, S.R. Fenugreek: A naturally occurring edible spice as an anticancer agent. Cancer Biol. Ther. 2009, 8, 272–278. [Google Scholar] [CrossRef]
  706. Lorin, S.; Hamaï, A.; Mehrpour, M.; Codogno, P. Autophagy regulation and its role in cancer. Semin. Cancer Biol. 2013, 23, 361–379. [Google Scholar] [CrossRef]
  707. Thorburn, A.; Thamm, D.H.; Gustafson, D.L. Autophagy and cancer therapy. Mol. Pharmacol. 2014, 85, 830–838. [Google Scholar] [CrossRef] [PubMed]
  708. Polewska, J. Autophagy--molecular mechanism, apoptosis and cancer. Postep. Hig. I Med. Dosw. 2012, 66, 921–936. [Google Scholar] [CrossRef] [PubMed]
  709. Chang, H.Y.; Kao, M.C.; Way, T.D.; Ho, C.T.; Fu, E. Diosgenin suppresses hepatocyte growth factor (HGF)-induced epithelial-mesenchymal transition by down-regulation of Mdm2 and vimentin. J. Agric. Food Chem. 2011, 59, 5357–5363. [Google Scholar] [CrossRef] [PubMed]
  710. Chen, J.; Zhang, H.F.; Xiong, C.M.; Ruan, J.L. Inhibitory effect of diosgenin on experimentally induced benign prostatic hyperplasia in rats. J. Huazhong Univ. Sci. Technology. Med. Sci. = Hua Zhong Ke Ji Da Xue Xue Bao. Yi Xue Ying De Wen Ban = Huazhong Keji Daxue Xuebao. Yixue Yingdewen Ban 2016, 36, 806–810. [Google Scholar] [CrossRef] [PubMed]
  711. Peng, Y.; Tang, R.; Ding, L.; Zheng, R.; Liu, Y.; Yin, L.; Fu, Y.; Deng, T.; Li, X. Diosgenin inhibits prostate cancer progression by inducing UHRF1 protein degradation. Eur. J. Pharmacol. 2023, 942, 175522. [Google Scholar] [CrossRef] [PubMed]
  712. Sikka, S.; Shanmugam, M.K.; Siveen, K.S.; Ong, T.H.; Yang, M.H.; Lee, J.H.; Rajendran, P.; Chinnathambi, A.; Alharbi, S.A.; Alahmadi, T.A.; et al. Diosgenin attenuates tumor growth and metastasis in transgenic prostate cancer mouse model by negatively regulating both NF-κB/STAT3 signaling cascades. Eur. J. Pharmacol. 2021, 906, 174274. [Google Scholar] [CrossRef] [PubMed]
  713. Liu, C.; Xie, F.; Zhao, G.D.; Wang, D.F.; Lou, H.X.; Liu, Z.P. Synthetic studies towards 1α-hydroxysolasodine from diosgenin and the unexpected tetrahydrofuran ring opening in the Birch reduction process. Steroids 2015, 104, 214–219. [Google Scholar] [CrossRef] [PubMed]
  714. Hamid, A.A.; Kaushal, T.; Ashraf, R.; Singh, A.; Chand Gupta, A.; Prakash, O.; Sarkar, J.; Chanda, D.; Bawankule, D.U.; Khan, F.; et al. (22β,25R)-3β-Hydroxy-spirost-5-en-7-iminoxy-heptanoic acid exhibits anti-prostate cancer activity through caspase pathway. Steroids 2017, 119, 43–52. [Google Scholar] [CrossRef] [PubMed]
  715. Mironov, M.E.; Oleshko, O.S.; Pokrovskii, M.A.; Rybalova, T.V.; Pechurov, V.K.; Pokrovskii, A.G.; Cheresis, S.V.; Mishinov, S.V.; Stupak, V.V.; Shults, E.E. 6-(4′-Aryl-1′,2′,3′-triazolyl)-spirostan-3,5-diols and 6-(4′-Aryl-1′,2′,3′-triazolyl)-7-hydroxyspirosta-1,4-dien-3-ones: Synthesis and analysis of their cytotoxicity. Steroids 2019, 151, 108460. [Google Scholar] [CrossRef]
  716. Michalak, O.; Krzeczyński, P.; Cieślak, M.; Cmoch, P.; Cybulski, M.; Królewska-Golińska, K.; Kaźmierczak-Barańska, J.; Trzaskowski, B.; Ostrowska, K. Synthesis and anti-tumour, immunomodulating activity of diosgenin and tigogenin conjugates. J Steroid Biochem. Mol. Biol. 2020, 198, 105573. [Google Scholar] [CrossRef]
  717. Parama, D.; Boruah, M.; Yachna, K.; Rana, V.; Banik, K.; Harsha, C.; Thakur, K.K.; Dutta, U.; Arya, A.; Mao, X.; et al. Diosgenin, a steroidal saponin, and its analogs: Effective therapies against different chronic diseases. Life Sci. 2020, 260, 118182. [Google Scholar] [CrossRef] [PubMed]
  718. Smith, M.R.; Kaufman, D.; George, D.; Oh, W.K.; Kazanis, M.; Manola, J.; Kantoff, P.W. Selective aromatase inhibition for patients with androgen-independent prostate carcinoma. Cancer 2002, 95, 1864–1868. [Google Scholar] [CrossRef] [PubMed]
  719. Santen, R.J.; Petroni, G.R.; Fisch, M.J.; Myers, C.E.; Theodorescu, D.; Cohen, R.B. Use of the aromatase inhibitor anastrozole in the treatment of patients with advanced prostate carcinoma. Cancer 2001, 92, 2095–2101. [Google Scholar] [CrossRef] [PubMed]
  720. Lutz, S.Z.; Hennenlotter, J.; Scharpf, M.; Todenhofer, T.; Heni, M.; Guirguis, A.; Peter, A.; Staiger, H.; Fritsche, A.; Stenzl, A.; et al. The classical nuclear estrogen receptors ERα and ERβ, and the G-protein-coupled receptor-30 are differently expressed in prostate cancer depending on diabetes status. In Proceedings of the EASD Virtual Meeting, Munich, Germany, 12–16 September 2016. [Google Scholar]
  721. Gross, M.; Ramirez, C.; Luthringer, D.; Nepomuceno, E.; Vollmer, R.; Burchette, J.; Freedland, S.J. Expression of androgen and estrogen related proteins in normal weight and obese prostate cancer patients. Prostate 2009, 69, 520–527. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Estrogen biosynthesis pathway. Cholesterol is the precursor of steroid hormones. It originates pregnenolone through the cholesterol side-chain cleavage enzyme (CYP11A1), which is converted to progesterone by 3β-hydroxysteroid dehydrogenase (HSD3β). This enzyme is also responsible for the conversion of 17-hydroxypregnenolone to 17-hydroxyprogesterone, dehydroepiandrosterone (DHEA) to androstenedione, and androstenediol to testosterone. Estrogens are produced through the activity of aromatase (CYP19A1), which converts androstenedione and testosterone to estrone (E1) and 17β-estradiol (E2), respectively. E1 is reversibly converted to E2 by the enzyme 17β-hydroxysteroid dehydrogenase (HSD17β1/2), and both can originate E3 through the cytochrome P450 3A4/5 (CYP3A4/5).
Figure 1. Estrogen biosynthesis pathway. Cholesterol is the precursor of steroid hormones. It originates pregnenolone through the cholesterol side-chain cleavage enzyme (CYP11A1), which is converted to progesterone by 3β-hydroxysteroid dehydrogenase (HSD3β). This enzyme is also responsible for the conversion of 17-hydroxypregnenolone to 17-hydroxyprogesterone, dehydroepiandrosterone (DHEA) to androstenedione, and androstenediol to testosterone. Estrogens are produced through the activity of aromatase (CYP19A1), which converts androstenedione and testosterone to estrone (E1) and 17β-estradiol (E2), respectively. E1 is reversibly converted to E2 by the enzyme 17β-hydroxysteroid dehydrogenase (HSD17β1/2), and both can originate E3 through the cytochrome P450 3A4/5 (CYP3A4/5).
Biomedicines 12 01636 g001
Figure 2. Structure of synthetic estrogens with reported effects in PCa. Legend: DES—diethylstilbestrol; HE3235—17α-ethynyl-5α-androstane-3α, 17β-diol; 8β-VE2—8β-vinylestra-1,3,5(10)-triene-3β,17β-diol; APVE2—17α-20Ζ-21-[(4-amino)phenyl]-19-norpregna-1,3,5(10),20-tetraene-3,17β-diolthe; 2F-E2—2-fluoroestradiol.
Figure 2. Structure of synthetic estrogens with reported effects in PCa. Legend: DES—diethylstilbestrol; HE3235—17α-ethynyl-5α-androstane-3α, 17β-diol; 8β-VE2—8β-vinylestra-1,3,5(10)-triene-3β,17β-diol; APVE2—17α-20Ζ-21-[(4-amino)phenyl]-19-norpregna-1,3,5(10),20-tetraene-3,17β-diolthe; 2F-E2—2-fluoroestradiol.
Biomedicines 12 01636 g002
Figure 3. Antitumorigenic effects and signaling pathways activated by estrogens in controlling prostate cell survival, proliferation, apoptosis, invasion, migration, and angiogenesis. Summary of effects triggered by endogenous (E2, αE2, 2-ME2, 3α-diol, 3β-Adiol) and synthetic (8β-VE2, APVE2, DES) estrogenic compounds according to the information provided in the text and Supplementary Table S1). JNK—c-Jun N-terminal kinase; MAPK—mitogen-activated protein kinase; ERK—extra-cellular signal regulated kinase; NF-κβ—factor nuclear kappa B; FOXO3a—Forkhead box class O 3a; FOXO1—Forkhead box protein O1; KLF5—Kruppel-like transcription factor 5; CDC25C—cell division cycle 25C; PUMA—p53 upregulated modulator of apoptosis; FLIP—FLICE-inhibitory protein; XIAP—X-linked inhibitor of apoptosis; Fas—Fas receptor; Fas L—Fas ligand; MMP-2/9—matrix metalloproteinase-2 and 9; IGF-1R—insulin-like growth factor 1 receptor; IGFBP-3/4—insulin-like growth factors binding protein-3/4; TGF-β1—transforming growth factor beta-1; TGF-β RI—transforming growth factor beta receptor 1; TGF-β RII—transforming growth factor beta receptor 2; VEGF—vascular endothelial growth factor; PDGFA—platelet derived growth factor subunit A.
Figure 3. Antitumorigenic effects and signaling pathways activated by estrogens in controlling prostate cell survival, proliferation, apoptosis, invasion, migration, and angiogenesis. Summary of effects triggered by endogenous (E2, αE2, 2-ME2, 3α-diol, 3β-Adiol) and synthetic (8β-VE2, APVE2, DES) estrogenic compounds according to the information provided in the text and Supplementary Table S1). JNK—c-Jun N-terminal kinase; MAPK—mitogen-activated protein kinase; ERK—extra-cellular signal regulated kinase; NF-κβ—factor nuclear kappa B; FOXO3a—Forkhead box class O 3a; FOXO1—Forkhead box protein O1; KLF5—Kruppel-like transcription factor 5; CDC25C—cell division cycle 25C; PUMA—p53 upregulated modulator of apoptosis; FLIP—FLICE-inhibitory protein; XIAP—X-linked inhibitor of apoptosis; Fas—Fas receptor; Fas L—Fas ligand; MMP-2/9—matrix metalloproteinase-2 and 9; IGF-1R—insulin-like growth factor 1 receptor; IGFBP-3/4—insulin-like growth factors binding protein-3/4; TGF-β1—transforming growth factor beta-1; TGF-β RI—transforming growth factor beta receptor 1; TGF-β RII—transforming growth factor beta receptor 2; VEGF—vascular endothelial growth factor; PDGFA—platelet derived growth factor subunit A.
Biomedicines 12 01636 g003
Figure 4. Phytoestrogens with a role in controlling PCa cell survival, proliferation, apoptosis, invasion, migration, and angiogenesis and activated signaling pathways. JNK—c-Jun N-terminal kinase; JAK2—Janus kinase 2; STAT3—signal transducer and activator of transcription; MAPK—mitogen-activated protein kinase; ERK—extra-cellular signal regulated kinase; NF-κβ—factor nuclear kappa B; PI3K—phosphatidylinositol 3′-kinase; AMPK—AMP-activated protein kinase; FOXO—Forkhead box; pRb—retinoblastoma protein; CDKs 1/2/4/6—cyclin-dependent kinases 1/2/4/6; PUMA—p53 upregulated modulator of apoptosis; XIAP—X-linked inhibitor of apoptosis; Fas—Fas receptor; Fas L—Fas ligand; TRAIL—TNF-related apoptosis-inducing ligand; MMP-1/2/7/9/13—matrix metalloproteinase 1/2/7/9/13; IGF-1—insulin-like growth factor 1; IGF-1R—insulin-like growth factor 1 receptor; TGF-β—transforming growth factor beta; HIFα—hypoxia-inducible factor α; COX-2—cyclooxygenase-2; IL6—interleukin 6; VEGF—vascular endothelial growth factor; EGFR—epidermal growth factor receptor; GLUT1—glucose transporter type 1; FASN—fatty acid synthase.
Figure 4. Phytoestrogens with a role in controlling PCa cell survival, proliferation, apoptosis, invasion, migration, and angiogenesis and activated signaling pathways. JNK—c-Jun N-terminal kinase; JAK2—Janus kinase 2; STAT3—signal transducer and activator of transcription; MAPK—mitogen-activated protein kinase; ERK—extra-cellular signal regulated kinase; NF-κβ—factor nuclear kappa B; PI3K—phosphatidylinositol 3′-kinase; AMPK—AMP-activated protein kinase; FOXO—Forkhead box; pRb—retinoblastoma protein; CDKs 1/2/4/6—cyclin-dependent kinases 1/2/4/6; PUMA—p53 upregulated modulator of apoptosis; XIAP—X-linked inhibitor of apoptosis; Fas—Fas receptor; Fas L—Fas ligand; TRAIL—TNF-related apoptosis-inducing ligand; MMP-1/2/7/9/13—matrix metalloproteinase 1/2/7/9/13; IGF-1—insulin-like growth factor 1; IGF-1R—insulin-like growth factor 1 receptor; TGF-β—transforming growth factor beta; HIFα—hypoxia-inducible factor α; COX-2—cyclooxygenase-2; IL6—interleukin 6; VEGF—vascular endothelial growth factor; EGFR—epidermal growth factor receptor; GLUT1—glucose transporter type 1; FASN—fatty acid synthase.
Biomedicines 12 01636 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Figueira, M.I.; Carvalho, T.M.A.; Macário-Monteiro, J.; Cardoso, H.J.; Correia, S.; Vaz, C.V.; Duarte, A.P.; Socorro, S. The Pros and Cons of Estrogens in Prostate Cancer: An Update with a Focus on Phytoestrogens. Biomedicines 2024, 12, 1636. https://doi.org/10.3390/biomedicines12081636

AMA Style

Figueira MI, Carvalho TMA, Macário-Monteiro J, Cardoso HJ, Correia S, Vaz CV, Duarte AP, Socorro S. The Pros and Cons of Estrogens in Prostate Cancer: An Update with a Focus on Phytoestrogens. Biomedicines. 2024; 12(8):1636. https://doi.org/10.3390/biomedicines12081636

Chicago/Turabian Style

Figueira, Marília I., Tiago M. A. Carvalho, Joana Macário-Monteiro, Henrique J. Cardoso, Sara Correia, Cátia V. Vaz, Ana P. Duarte, and Sílvia Socorro. 2024. "The Pros and Cons of Estrogens in Prostate Cancer: An Update with a Focus on Phytoestrogens" Biomedicines 12, no. 8: 1636. https://doi.org/10.3390/biomedicines12081636

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