Next Article in Journal
Combining Cortical Voltage Imaging and Hippocampal Electrophysiology for Investigating Global, Multi-Timescale Activity Interactions in the Brain
Next Article in Special Issue
Low Doses of PFOA Promote Prostate and Breast Cancer Cells Growth through Different Pathways
Previous Article in Journal
The Therapeutic Potential of Milk Extracellular Vesicles on Colorectal Cancer
Previous Article in Special Issue
Impact of Chemical Endocrine Disruptors and Hormone Modulators on the Endocrine System
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Breast Cancer Stem Cells Using Naturally Occurring Phytoestrogens

1
MD/PhD Program, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
2
Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
3
Department of Cancer Biology, Cardinal Bernardin Cancer Center, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(12), 6813; https://doi.org/10.3390/ijms23126813
Submission received: 8 March 2022 / Revised: 31 May 2022 / Accepted: 9 June 2022 / Published: 18 June 2022
(This article belongs to the Special Issue Advances in Endocrine Disruptors 2.0)

Abstract

:
Breast cancer therapies have made significant strides in improving survival for patients over the past decades. However, recurrence and drug resistance continue to challenge long-term recurrence-free and overall survival rates. Mounting evidence supports the cancer stem cell model in which the existence of a small population of breast cancer stem cells (BCSCs) within the tumor enables these cells to evade conventional therapies and repopulate the tumor, giving rise to more aggressive, recurrent tumors. Thus, successful breast cancer therapy would need to target these BCSCs, as well the tumor bulk cells. Since the Women’s Health Initiative study reported an increased risk of breast cancer with the use of conventional hormone replacement therapy in postmenopausal women, many have turned their attention to phytoestrogens as a natural alternative. Phytoestrogens are plant compounds that share structural similarities with human estrogens and can bind to the estrogen receptors to alter the endocrine responses. Recent studies have found that phytoestrogens can also target BCSCs and have the potential to complement conventional therapy eradicating BCSCs. This review summarized the latest findings of different phytoestrogens and their effect on BCSCs, along with their mechanisms of action, including selective estrogen receptor binding and inhibition of molecular pathways used by BCSCs. The latest results of phytoestrogens in clinical trials are also discussed to further evaluate the use of phytoestrogen in the treatment and prevention of breast cancer.

1. Introduction

Breast cancer is the most common cancer in the United States, representing 14.8% of all new cancer cases in 2021 [1]. Due to early detection and improved treatments, the death rate for breast cancer has been falling continuously since 1992, and survival rate is very high for most patients [1]. However, challenges remain, as many patients may still relapse and die from long-term recurrence, metastasis and drug resistance [2,3]. Breast cancer is difficult to treat due to its inherent heterogeneity across the molecular, phenotypic and functional features within a patient’s tumor and across different patients [4]. Tumor heterogeneity across patients may be explained by intrinsic molecular subtypes of breast cancer, while inter-tumor heterogeneity can be explained with the cancer stem cell hypothesis [5]. In fact, increasing evidence points to the existence of a subpopulation of breast cancer stem cells (BCSCs) within a tumor. These cells can resist conventional therapies and repopulate the tumor, leading to relapse, recurrence and metastasis of more aggressive tumors [6]. Thus, to totally eradicate breast cancer and prevent relapse, strategies must be devised to address these BCSCs, as well as the bulk breast cancer cells. This review will briefly summarize the characteristics of BCSCs and then discuss the use of naturally occurring, dietary phytoestrogens as a potential BCSC therapy.

2. Characteristics of Breast Cancer Stem Cells

2.1. Mammary Stem Cells and Origin of BCSCs

Breast tissues contain a large pool of long-lived, self-renewing and multipotent mammary stem cells (MaSCs) that can give rise to both the luminal and basal epithelial lineages [4,7,8]. These MaSCs reside within a stem cell niche and are wired to respond to environmental cues, such as estrogen, Wnt, Hedgehog, Notch, TGF-β and growth factor signaling pathways [7,9,10]. Proper communication and response by MaSCs lead to the dynamic changes in the mammary glands seen during puberty, menstruation and pregnancy [7]. However, MaSCs are also at risk of acquiring mutations leading to transformation and tumorigenesis due to their inherent stem cell characteristics [9,11]. These MaSCs or progenitors give rise to a tumor subpopulation displaying dysregulated mammary stem cell characteristics called BCSCs [6,12,13,14,15]. Recent data support the existence of this small population of undifferentiated BCSCs with self-renewal and full differentiation capacity within the tumor [9,16]. In accordance with the cancer stem cell hypothesis observed in leukemia and in other types of cancer, this subpopulation of cancer stem cells is able to repopulate and reproduce the full heterogeneity of the original tumors [6,17].

2.2. Identification, Phenotypes and Roles of BCSCs in Breast Cancer

BCSCs, also known as tumor-initiating cells, comprise 0.1–1.0% of the tumor bulk according to various estimates across different breast cancer subtypes [5,15,18,19,20]. These BCSCs are identified as cancer cells expressing aldehyde dehydrogenase 1 (ALDH1+) and/or CD44+/CD24−/low [21]. In 2003, Al-Haji et al. were the first to demonstrate that only a subpopulation of cells expressing the CD44+/CD24/Lineage phenotype in human breast tumors was able to form tumors in immunodeficient mice [22]. Moreover, as few as 100 cells with this phenotype were capable of forming tumors in mice with similar heterogeneity to that of the original tumor, while tens of thousands of cells with other phenotypes failed to form tumors [22]. In vitro, these CD44+/CD24 BCSCs displayed stemness properties, capable of self-renewal, extensive proliferation, clonal nonadherent spherical clusters (mammosphere formation), differentiation along mammary epithelial lineages and chemotherapy resistance [23,24]. Due to these stem-like characteristics, BCSCs were associated with poor prognosis and played critical roles in tumorigenesis, metastasis, recurrence and resistance to conventional therapy [5,15,19,25,26,27,28,29,30].

2.3. Signaling Pathways Critical for BCSCs

Studies have identified several key pathways, including Notch, Wnt/β-catenin, Hedgehog, Hippo, NF-κB, Stat3, HIF, TGF-β, PI3K/Akt, HER2 and BRCA1 as crucial in regulating the behavior and survival of BCSCs [2,9,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62]. Specifically, embryonic developmental pathways, such as Notch, Wnt/β-catenin, Hedgehog and Hippo, normally regulate self-renewal and differentiation of normal mammary stem cells. Dysregulation or aberrant activation of these pathways transformed normal stem cells into BCSCs and led to tumorigenesis [9,60]. Inflammation-related pathways (NF-κB and Stat3), BCSC microenvironment and tumor hypoxia (TGF-β and HIF), proliferative pathways (HER2 and PI3K/Akt) and the loss of tumor suppressor (BRCA1) also contributed to the stem cell properties in BCSCs [9,60]. In addition to these pathways, recent studies showed that several microRNAs and long non-coding RNAs could modulate genes associated with these stem cell pathways to regulate BCSCs [2,9,15,63,64,65,66,67].

2.4. BCSCs in Resistance and Stem Cell Targeted Therapy

Current data identified BCSCs as key drivers responsible for resistance to first-line treatment, including conventional chemotherapy and radiotherapy, leading to treatment failure and cancer relapse [9,68]. BCSCs displayed inherent resistance to conventional therapy due to their high expression of drug transporters, plasticity and quiescence state, oxidative stress resistance, active DNA damage response and anti-apoptotic effects [2,28,29,69,70]. Thus, new targeted approaches are needed to eradicate BCSCs. One rational approach is to target the self-renewal pathways that BCSCs depend on, using Notch inhibitors (γ-secretase inhibitors), Wnt monoclonal antibodies, Hedgehog small molecule inhibitors, Hippo/mevalonate pathway inhibitors, HER-2 inhibitors and PI3K/Akt inhibitors [2,9]. Other strategies target the BCSC microenvironment via TGF-β inhibitors and CD44v6 monoclonal antibody, the DNA damage response, ABC drug transporters, miRNAs and the ubiquitin–proteasome system (UPS) [2]. CDK inhibitors, which could induce senescence phenotypes, or agents inducing stem cell differentiation are also being investigated [2,9]. Lastly, immunotherapy targeting PD-L1 and other novel BCSC antigens are being explored as another potential treatment venue [9]. A number of these therapies targeting stemness pathways are being tested in clinical trials with encouraging results, but none have yet been approved [9]. Toxicities and off-target effects on normal stem cells remain major concerns. Overall, these highlight the need for more effective and safer therapy against BCSCs.

2.5. Phytoestrogens and Natural Products Targeting BSCSs

Natural products, especially plant-derived products, have long been used in traditional medicine around the world [71]. Many well-known drugs used to treat breast cancer actually originated from plants, such as taxol, vincristine and vinblastine [72]. The ability to suppress BCSCs by inhibiting the stemness pathways was recently reported for several naturally occurring compounds, including salinomycin, curcumin, green tea EGCG, sulforaphane and piperine [73,74,75,76,77,78,79,80,81,82]. In this regard, one class of natural products targeting BCSCs rose to prominence: dietary phytoestrogens. These plant compounds with estrogenic activities have gained public attention since the publication of the Women’s Health Initiative in 2002. This study raised concerns about the use of estrogen and progestin in healthy postmenopausal women, as it was associated with significant adverse effects and increased risk of breast cancer [83]. Since then, hormone therapy usage sharply decreased, while herbal/dietary supplements usage increased [84]. As dietary supplements, phytoestrogens may be beneficial for menopause in older women or those unsuitable for hormone therapy [85]. Phytoestrogens have been shown to inhibit breast cancer growth and, more recently, they have been reported to inhibit BCSCs specifically. Hence, the rest of this review will dive deep into the use of several phytoestrogens as potential BCSC therapy.

3. Phytoestrogens Targeting BCSCs and Their Mechanisms of Action

Phytoestrogens were accidentally discovered in the 1940s due to a case of sheep grazing on red clover containing phytoestrogens, which resulted in their infertility [86,87]. These natural compounds are produced by various plants in response to environment stressors, serving as antioxidants, antifungals and antibiotics for the defense of plants [88,89]. Humans are also exposed to phytoestrogens via our regular diet. Incidentally, phytoestrogens are nonsteroidal polyphenols that share structural similarities with estradiol (17-β-estradiol) and bind both estrogen receptors with varying affinities, resulting in estrogenic and/or antiestrogenic effects [88]. Based on their chemical structures, phytoestrogens can be classified into four main classes: flavonoids, stilbenes, lignans and isoflavonoids [90]. Many studies have examined the use of various phytoestrogens in relieving menopausal symptoms, decreasing risks of hormonal cancers, osteoporosis and cardiovascular diseases [91]. This review solely focused on phytoestrogens that affect BCSCs, including the flavonoids (genistein, S-equol and naringenin) and the stilbenes (resveratrol and pterostilbene). A summary of these phytoestrogens and their mechanisms targeting BCSCs is depicted in Figure 1.

3.1. Flavonoids: Genistein, S-Equol and Naringenin

3.1.1. Structures and Sources

One major group of dietary phytoestrogens is flavonoids. Flavonoids are composed of two aromatic rings bearing at least one hydroxyl group [88]. An important subclass of naturally occurring flavonoids is isoflavones, which are found mainly in soy and soy products. Major soy isoflavones include genistein and daidzein [88]. Genistein can be produced from the flavanone naringenin in soy plants [92]. Daidzein, upon consumption, can be further metabolized by microbes in the human gut to form the isoflavone S-equol. S-equol shows greater affinity for estrogen receptors than its precursor daidzein and is considered the more bioactive form [93,94]. However, studies estimated that only about 30% of the people in Western countries and 60% in Asian countries are able to produce S-equol following soy consumption, which might explain the different effects of soy observed in different studied populations [88,95].

3.1.2. Estrogen Receptor (ER) Affinity

Since genistein shares structural similarities with 17-β-estradiol, it is known to activate both ERα and ERβ through a classical mechanism [96]. Unlike endogenous estrogens, which bind both ERs equally, genistein and S-equol both prefer ERβ over ERα [94,96]. A later study further showed that genistein (along with daidzein and equol) was more selective at enhancing ERβ-regulated genes at multiple levels, from higher ERβ affinity to higher efficiency at coactivator recruitment and chromatin binding [97]. While ERα is associated with stimulating proliferation of breast cancer cells, ERβ activation opposes ERα actions [98,99]. Thus, genistein’s preference for ERβ suggests that it could have dose-dependent effects on breast cancer cells, based on the expression of ERα compared to ERβ [88,100,101].

3.1.3. Epidemiology

Soy isoflavones have gathered lots of interest in recent decades as potential chemopreventives. In fact, epidemiological studies associated higher intake of soy isoflavones in Asian countries (up to 47 mg/day) compared to Western countries (0.1–1.2 mg/day) with lower incidence of breast cancer [88,102,103]. Some studies further suggested that early soy/genistein exposure during childhood and adolescence might be needed to reduce breast cancer risk later in life [104,105,106,107,108]. In women with breast cancer, multiple Asian cohort studies found that soy food intake post-diagnosis was associated with lower mortality and cancer recurrence [109,110]. These observations suggest that soy isoflavones could inhibit breast cancer growth and recurrence, necessitating further studies to investigate causation and mechanisms.

3.1.4. Genistein and Growth of Breast Cancer Bulk Cells

Genistein at low doses (≤10 μmol/L) was found to be estrogenic and promoted the growth of hormone-dependent breast cancer cell lines and tumors in mouse models [111,112,113,114]. This pro-tumorigenic effect is likely due to genistein acting as a weak estrogen, activating the ERα pathway. At a higher dose, genistein had been shown to inhibit breast cancer by inducing apoptosis, promoting cell cycle arrest and inhibiting angiogenesis [111,115,116,117,118]. These anticancer and anti-angiogenic effects of genistein are attributed to estrogen-independent pathways, including caspase activation, inhibition of VEGF signaling, PTK tyrosine kinase and MAPK inhibition and epigenetic modifications.

3.1.5. Genistein and Its Role in BCSCs

Multiple studies showed that genistein inhibited BCSCs through direct inhibition of the pathways involved in stem cell growth and differentiation as well as paracrine signaling from surrounding cells. Montales et al. was the first study to report that both sera of adult mice fed with genistein and genistein itself were able to inhibit the basal stem-like CD44+/CD24/ESA+ and the luminal progenitor CD24+ subpopulations from MDA-MB-231 and MCF-7 cells [119]. Furthermore, this inhibition of breast cancer mammosphere formation is associated with AKT inhibition and upregulation of PTEN [119]. In vivo, genistein given by intraperitoneal injection to nude mice-bearing MCF-7 xenografts was able to reduce BCSCs in the tumor by downregulating the Hedgehog–Gli1 self-renewal pathway [120]. Aside from direct effects, BCSCs may also be affected by the surrounding cells within the stem cell niche through paracrine signaling. Genistein inhibited mammary adipogenesis in vitro and in vivo, through the activation of ERβ and inhibition of PPARγ and FASN expression. Subsequently, the conditioned medium from these genistein-treated adipose cells inhibited mammophere formation of ER+ breast cancer cells (MCF-7), suggesting a paracrine effect [121]. Genistein at physiological concentrations (40 nM–2 μM) in co-cultures also stimulated ER+ breast cancer cells (MCF-7) to release amphiregulin. Released amphiregulin then induced the neighboring ER-negative BCSCs to differentiate into epithelial-like cells, correlating with the activation of PI3K/Akt and MEK/ERK signaling pathways [122]. In contrast to previous studies, Lauricella et al. demonstrated that treatment of MCF-7 cells with 25 μM genistein or 17-β estradiol actually increased the number and sizes of the mammosphere formed. Treatment with genistein increased the expression of ERα36 in tertiary mammospheres, enhancing their proliferation [123].

3.1.6. S-Equol and Growth of Breast Cancer Bulk Cells and BCSCs

S-equol, which is the bioactive form of daidzein, has preference for ERβ over ERα, similar to that of genistein [94]. In fact, Yuan et al. showed that S-equol specifically induced ERβ tyrosine phosphorylation and inhibited breast cancer growth in vitro and in vivo [124]. However, S-equol also has dose-dependent effects on breast cancer cells, with higher S-equol concentrations (50–350 μM) inhibiting cancer growth and invasion [125,126,127,128,129,130]. At lower physiological concentrations (~1 μM), S-equol acted as weak estrogen to promote ER+ breast cancer proliferation in vitro but had no effect on tumor growth in mice [128,131,132,133]. In particular, the Dharmawardhane group showed that dietary daidzein increased breast tumor growth and metastasis in an ER-negative breast tumor mouse xenograft model [134]. This pro-tumorigenic effect can be recapitulated in vitro by the daidzein metabolite, S-equol (25–50 μM), increasing breast cancer proliferation of ER-negative breast cancer cells via upregulation of c-Myc, eIF4GI and enhanced protein synthesis [135]. A subsequent study showed that S-equol also increased the size and number of mammospheres in ER-negative MDA-MB-435 cells through the upregulation of c-Myc [136]. This suggests that S-equol in certain situations could enhance BCSCs in ER-negative breast cancer. However, data on S-equol in BCSCs are severely lacking and would merit further study to delineate the mechanism of action by S-equol.

3.1.7. Naringenin and Growth of Breast Cancer Bulk Cells and BCSCs

Naringenin is the precursor of genistein [92]. Naringenin (4′,5,7-trihydroxy flavanone) and naringin (4,5,7-trihydroxy flavonone 7-rhamnoglucoside) are the major flavanones found in citrus fruits, such as grapefruits, oranges, cherries and tomatoes [137,138,139]. In humans, the glycoside naringin is metabolized by gut bacteria into the aglycone naringenin [137,139,140,141]. Naringenin is considered the more bioactive form due to the lack of steric hindrance of the sugar moiety [142]. Similar to other phytoestrogens, naringenin weakly binds both ERα and ERβ, with a preference for ERβ (relative binding ERβ/ERα = 0.5%/0.08%, E2 = 100%) [143]. Naringenin was found to inhibit the growth and migration of both ER-negative (MDA-MB-453, MDA-MB-231) and ER-positive (MCF-7) breast cancer cells in vitro and in vivo [142,144,145,146,147,148,149,150,151,152,153]. Furthermore, combining tamoxifen with naringenin more effectively inhibited the proliferation of ER-positive breast cancer cells via inhibition of both PI3K and MAPK pathways and modulation of several ER-target genes [152,154]. Naringenin is also effective at targeting BCSCs, as reported in recent studies. Naringenin inhibited BCSCs both in vitro and in vivo in a DAXX-dependent manner [155]. Naringenin’s upregulation of DAXX and subsequent inhibition of BCSCs were more selective toward ERβ than ERα [155]. Similarly, Hermawan et al. found that naringenin inhibited BCSC mammosphere formation, potentially through the modulation of p53 and ERα mRNA [82]. Naringenin was also identified as the active compound in the Xihuang pill, a traditional medicine used to treat breast cancer in China [156]. In vitro, both the Xihuang pill and naringenin inhibited mammosphere formation in triple negative breast cancer cell lines by upregulating NR3C2 expression [156].

3.2. Stilbenes: Resveratrol and Pterostilbene

3.2.1. Structures and Sources

Stilbene is a class of phytoestrogens bearing two benzene rings joined by an ethanol or ethylene [157]. Arguably, the most well-known stilbene is resveratrol, which is a metabolized stilbene naturally produced by several plants in response to pathogen attacks [158,159]. Trans-resveratrol is the more dominant, estrogenic form due to its configuration, and it can be found in peanuts, blueberries, pines, grapevine and especially red wine [90,160,161,162]. Pterostilbene is a natural methoxylated derivative of resveratrol found in similar plants [163,164]. Pterostilbene is considered the more bioavailable and bioactive molecule compared to resveratrol [165,166].

3.2.2. Estrogen Receptor (ER) Affinity

Trans-resveratrol shares structural similarity with diethylstilbestrol (a synthetic human estrogen) and binds both ERα and ERβ equally, unlike other phytoestrogens, which preferentially bind ERβ [167]. However, resveratrol binds ERs with 7000-fold lower affinity than estradiol and can act as a weak estrogen in the absence of 17β-estradiol [167,168]. Resveratrol competes with 17β-estradiol for ERα on specific EREs but not ERβ [167]. These results indicate that resveratrol could differentially alter the activity of ERα and ERβ as an agonist or antagonist [167,169]. Although no direct ER binding data on pterostilbene were found, recent studies reported that the anticancer and antioxidant effects of pterostilbene required ERα and/or ERβ [170,171,172,173]. This suggests that pterostilbene could also alter ER activity due its structural similarity with resveratrol.

3.2.3. Epidemiology

The Mediterranean diet, which includes significant plant polyphenols and wine consumption, has been associated with lower incidence of breast cancer [174,175,176,177,178]. Resveratrol has been identified as one of the protective bioactive molecules in this diet [179]. Additionally, resveratrol is thought to be the ingredient in wine responsible for the “French Paradox” effect, in which moderate wine consumption in French people is associated with low incidence of heart disease despite their high-fat diet [90,160,180,181]. When resveratrol concentration was examined, studies reported some conflicting results. In the Swiss Canton of Vaud case study, Levi et al. found a significant inverse association between resveratrol intake from grape consumption and breast cancer risk [182]. Meanwhile, the Chianti cohort study found no association between urinary resveratrol and cancer-related mortality in older adults [183]. Overall, resveratrol still gathers significant interest as a chemopreventive agent with many benefits.

3.2.4. Resveratrol and Growth of Bulk Breast Cancer Cells

Resveratrol exhibits a dose-dependent, biphasic effect on ER+ breast cancer cells in vitro [184]. Various studies reported that resveratrol stimulated proliferation of ERα+ breast cancer cells at low doses (<10–22 μM) but inhibited proliferation and induced cell death at higher doses (>10–22 μM) [185,186,187,188,189]. For ER-negative breast cancer cells, resveratrol inhibited proliferation at both low and high doses [189,190,191,192,193]. Resveratrol inhibited breast cancer cell migration and invasion induced by nearby cancer-associated fibroblasts [194]. In vivo data from mouse and rat models showed that resveratrol treatment decreased breast tumor initiation and onset, tumor growth and angiogenesis, likely through the inhibition of oxidative DNA damage, tumor-promoting enzymes and/or NF-κB signaling [168,195,196,197,198,199].

3.2.5. Resveratrol and BCSCs

Recent studies found that resveratrol targeted BCSCs via multiple signaling pathways [165]. Fu et al. showed that resveratrol significantly inhibited survival of BCSCs and reduced the number and size of mammosphere formed in vitro mediated by induction of autophagy and suppression of Wnt/β-catenin signaling [200]. Injection of resveratrol in vivo likewise inhibited xenograft tumor growth and reduced the BCSC population [200]. In another study, resveratrol reduced BCSCs growth as assessed by mammosphere formation and inhibited tumor growth in xenograft mice models by suppressing lipogenesis and inducing Fas-mediated apoptosis [201]. Singh et al. reported that resveratrol inhibited mammosphere formation and breast carcinogenesis in the presence of estradiol in rats by inducing the NRF2 pathways [202]. A recent study by Peiffer et al. identified that resveratrol inhibited survival of BCSCs from ER+ breast cancer cells in a DAXX-dependent manner [155]. Additionally, resveratrol suppressed self-renewal activity of BCSCs mediated by cancer-associated fibroblasts through inhibition of Bmi-1 and Sox2 expression [194]. Alternatively, resveratrol has been shown to promote Argonaute2 (a central RNAi component) activity and enhanced tumor-suppressive miRNAs to inhibit BCSCs [203].

3.2.6. Pterostilbene and Growth of Bulk Cells and BCSCs

As a natural analog of resveratrol, pterostilbene has antioxidative activity by inhibiting COX-1 and has been shown to inhibit carcinogen-induced preneoplastic lesions in mammary organ cultures [204]. Although data are lacking, pterostilbene is thought to be similar to resveratrol by inhibiting cancer cell proliferation and inducing apoptosis [205]. In HER2+ breast cancer cells, pterostilbene inhibited HER2-mediated invasion and metastasis through downregulation of MMP-9 expression and inhibition of p38 and Akt pathways [205]. Pterostilbene has been shown to inhibit BCSCs via Argonaute2-dependent mechanism in a similar manner as resveratrol [203]. Additionally, pterostilbene also suppressed the generation of BCSCs and metastatic potential induced by tumor-associated macrophages [206]. This suppression is potentially mediated by the modulation of the NF-κB/miR488 circuit [206]. Wu et al. found that pterostilbene selectively killed BCSCs isolated from MCF-7, inhibited mammosphere formation and enhanced BCSCs sensitivity to chemotherapeutic drugs through reduction in CD44 expression and inhibition of the hedgehog/Akt/GSK3β signaling [207].

4. Clinical Trials Investigating Effects of Phytoestrogens on Breast Cancer

Based on the promising results of phytoestrogens inhibiting breast cancer in vitro and in vivo, several clinical trials investigated the protective effects of phytoestrogens in breast cancer treatment and prevention. Table 1 comprehensively listed the results of trials on the NIH website that was published between 2008 and 2020. Most phytoestrogens tested were soy isoflavones and its metabolite, S-equol, or flaxseed lignan and its metabolite, enterolactone. As expected, soy isoflavone is the most studied phytoestrogen and the most tested in clinical trials. Soy earned its fame from an epidemiological finding in which women living in Asian countries have lower incidence of breast cancer compared to Western women, and this is thought to be attributed to their diet containing soy phytoestrogens [103,208]. As soy is known to have estrogenic properties, many studies since have investigated the effects of soy isoflavones on breast tissue and breast cancer. However, the results from clinical studies have been conflicting, with soy isoflavones inhibiting breast epithelial cell growth in some trials, stimulating proliferation or having little effect in others. A recent study, including 39 patients with invasive triple negative breast cancer, demonstrated that a short course of oral S-equol was sufficient to inhibit the proliferation of breast tumor cells, as measured by Ki-67, with nearly one-third of patients having up to 20% decrease in Ki-67 expression [209]. Likewise, a prospective study on a cohort of German postmenopausal patients with breast cancer found that higher serum genistein was associated with lower Ki-67 expression in tumors [210]. Some studies further suggested that soy isoflavone supplement in healthy women could reduce breast cancer risk by inducing favorable modification of estrogen metabolism, lowering bone density and modulating blood pressure [211,212,213]. In contrast, Khan et al. found that an oral form of soy isoflavone containing genistein and daidzein did not reduce breast epithelial proliferation in healthy Western women who were at high risk of developing breast cancer; instead it induced a 27% increase in Ki-67 in premenopausal women [214]. Similarly, Shike et al. found soy intake in women with invasive adenocarcinoma induced genes that drive cell cycle progression and proliferation pathways in breast tumor cells, although no significant change was observed in Ki-67 expression [215]. Aside from proliferation, multiple studies found that soy isoflavones were well tolerated even at high dosages and did not induce adverse changes in breast density, DNA damage or apoptosis [216,217,218]. The discrepancy in these studies could be due to dosages of different soy isoflavone mixtures, duration of treatment, individual abilities to metabolize isoflavones, the composition of isoflavone-metabolizing microbiome, menopausal status and/or incidence of breast cancer. For example, people who can produce equol from soy isoflavones can exhibit different responses with soy consumption [219]. Recently, a small study in healthy Belgian and Dutch women found that oral consumption of soy products containing isoflavones led to high levels of genistein and daidzein, specifically in the breast tissue [220], suggesting they may be sufficient to have direct effects on breast tissue. More studies would be needed to delineate which patient population would benefit from soy isoflavone supplements and when.
Aside from soy isoflavones, red clover isoflavones were found to have no significant effects on breast density, endometrial thickness, serum cholesterol, follicle stimulating hormone levels and bone mineral density in a randomized trial of 401 healthy women in the United Kingdom [221]. Similarly, in a double-blinded, randomized trial of 152 premenopausal women at high risk of breast cancer, flaxseed Secoisolariciresinol Diglycoside (SDG) was reported to have no effect on Ki-67 expression in breast tissues [222]. This finding is different from the previous open-labeled, pilot study from the same group in which 49 healthy women took SDG supplement for a year and found their Ki-67 expression reduced from 4% to 2% [223]. Flaxseed had no effect on serum hormone levels or prognostic breast tumor characteristics when combined with an aromatase inhibitor in postmenopausal women with ER+ breast cancer [224].
Table 1. Reported results of clinical trials on phytoestrogens and breast cancer (2008–2020).
Table 1. Reported results of clinical trials on phytoestrogens and breast cancer (2008–2020).
PhytoestrogenTrial DesignSample Size/InterventionsResultsNCT Number/
Population StudiedReferences
Enterolactone and GenisteinObservational prospective cohort study
  • MARIE cohort of 1060–2105 postmenopausal breast cancer patients in Germany
  • Aged 50–74 years
Lifestyle questionnaires and blood samples collected at recruitment 2002–2005 (baseline)
and 2009 (follow-up)
  • Higher genistein concentrations were associated with lower Ki-67 expression in tumors showing >20% Ki-67
  • No associations between enterolactone or genistein and HER2 status
  • Enterolactone concentration inversely associated with all- cause mortality, breast-cancer-specific mortality and distant disease-free survival, likely through mediation of C-reactive protein
  • Higher enterolactone concentrations were associated with improved 5-year survival for postmenopausal breast cancer patients up to 4 years post-diagnosis
  • Higher concentrations of genistein, resveratrol and luteolin at follow-up in long- term survivors were associated with poorer subsequent prognosis
NCT03401034
(Jaskulski et al., 2017 [209])
(Jaskulski et al., 2018 [225])
(Jaskulski et al., 2020 [226])
Estrogenic Botanical SupplementsObservational study
  • Up to 3159 women in the UK with invasive primary breast cancer at 9–15 months post-diagnosis
  • Aged 18 to 75 years
Questionnaires (diet, lifestyle, use of complementary treatments) and blood/urine samples were collected annually for up to 5 years
  • Estrogenic botanical supplement usage doubled after diagnosis (8.4%)
  • Flaxseed and soy/isoflavone were most commonly used
  • Pre-diagnosis phytoestrogen intake was not associated with factors associated with improved breast cancer prognosis
NCT00701584
(Velentzis et al., 2011 [224])
(Swann et al., 2013 [227])
Red Clover IsoflavonesDouble-blind, randomized intervention study
  • 401 healthy women in the UK with at least one first-degree relative with breast cancer, in the UK
  • Aged 35 to 70 years
Red clover isoflavones for 3 years
  • Red clover isoflavones were well tolerated in healthy women
  • No significant differences in breast density, endometrial thickness, serum cholesterol, follicle stimulating hormone levels and bone mineral density
(Powles et al., 2008 [220])
Flaxseed Lignan Secoisolaricires inol Diglycoside (SDG)Double-blind, randomized intervention study, phase IIB
  • 152 premenopausal women who have a >2-fold relative risk of breast cancer compared to women in their age group, in USA
  • Aged 21–49 years
50 mg of (SDG) capsule once daily for 12 months
  • No difference in breast epithelial cells’ Ki-67 expression between SDG and placebo
NCT01276704
(Fabian et al., 2020 [221])
Flaxseed
(with aromatase inhibitor)
2 × 2 factorial, randomized interventional study
  • 24 postmenopausal women with estrogen receptor positive (ER+) breast cancer receiving surgery at Roswell
25 g/day ground flaxseed +/−1 mg/day anastrozole for
13–16 days prior to breast surgery
  • No interaction between flaxseed and aromatase inhibitor anastrozole in serum hormone levels or prognostic breast tumor characteristics
NCT00612560
(McCann et al., 2014 [223])
  • Park Cancer Institute, USA
  • Aged 59–65 years
  • Anastrozole may reduce circulating lignans induced by flaxseed
S-equolOpen-label intervention study, early phase I
  • 39 patients in Texas, USA, with invasive triple-negative breast cancer, confirmed by core needle biopsy
  • Aged 18 and older
S-equol at a dose of 50 mg or 150 mG PO twice daily for 10–21 days
  • S-equol was well tolerated and inhibited proliferation of breast tumor cells, as measured by a decrease in Ki-67 (8% compared to baseline)
  • Up to 20% decrease in Ki- 67 was observed in 28% of S-equol-treated patients
NCT02352025
(Lathrop et al., 2020 [208])
Soy IsoflavonesRandomized intervention study
  • 31 healthy Belgian or Dutch women who were scheduled for an esthetic breast reduction
  • Aged 18 to 62 years
Soy milk (16.98 mg genistein and 5.40 mg daidzein aglycone equivalents per dose) or soy supplement (5.27 mg genistein and 17.56 mg daidzein aglycone equivalents per dose), with three doses daily for 5 days before breast reduction
  • After soy product intake, genistein and total daidzein concentrations reached high levels in breast tissue, which could be sufficient to cause potential health effects
(Bolca et al., 2010 [219])
Soy IsoflavonesDouble-blind, randomized intervention study
  • 85 previously treated breast cancer women at high risk of breast cancer living in CA, USA
  • Aged 30–75 years
Oral soy isoflavones (50 mg/day) for 12 months
  • Treatment increased plasma soy isoflavone levels with minimal adverse effect
  • Soy supplementation did not decrease mammographic density
NCT01219075
(Wu et al., 2015 [215])
Soy IsoflavonesDouble-blind, randomized
  • 80 postmenopausal women with
250 mg of standardized soy extract corresponding to
  • Soy isoflavones did not affect breast density as measured by
(Delmanto et al., 2013 [216])
intervention study
  • vasomotor symptoms in Brazil
  • Aged >45 years
100 mg/day isoflavone for 10 monthsmammography and ultrasound
Soy IsoflavonesRandomized, placebo-controlled intervention study
  • 140 women with invasive breast adenocarcinoma in NY, USA
  • Aged mean 56 ± 12 years
5.8 g soy protein powder twice a day for 7–30 days prior to breast surgery
  • Soy intake induced overexpression of FGFR2 and genes that drive cell cycle and proliferation pathways in breast tumor cells
  • No significant changes in Ki67 or Caspase3
(Shike et al., 2014 [214])
Soy IsoflavonesDouble-blind, randomized intervention study
  • 200 healthy premenopausal women in TX, USA
  • Aged 30 to 42 years
Soy isoflavone tablet (60 mg daidzein, 60 mg genistein and
16.6 mg glycitein) twice per day for five days per week for up to 2 years
  • Isoflavones tended to normalize systolic blood pressure via serum calcium moderation and decreased diastolic blood pressure, independent of calcium level
  • Genistein significantly decreased whole-body bone mineral density at low serum calcium levels
NCT00204490
(Lu et al., 2020 [212]) (Nayeem et al., 2019 [211])
Soy Isoflavones/GenisteinDouble-blind, randomized intervention study, phase IIB
  • 126 healthy women who were at increased risk of developing breast cancer in IL, USA
  • Aged 42–55 years
Oral PTI G-2535 pill (genistein 150 mg, daidzein 74 mg, glycitein 11 mg) once daily up to 6 months
  • Soy isoflavones in healthy, high-risk adult Western women did not reduce breast epithelial proliferation, as measured by Ki-67
  • In premenopausal women, soy induced a 27% increase in Ki-67 in breast epithelial cells post-intervention
NCT00290758
(Khan et al., 2012 [213])
Soy Isoflavones/Genistein/Soy Isoflavones/GenisteinDouble-blind, randomized intervention study, phase
  • 30 healthy non- obese postmenopausal women at no risk of breast cancer, living in NC, USA
  • Aged 45–70 years
Oral genistein (PTI G-2535) twice daily for 84 days
  • High dose of soy isoflavones (900 mg) in postmenopausal women did not cause DNA damages, apoptosis or significant estrogenic effects
NCT00099008
(Pop et al., 2008 [217])
Soy Protein (with seaweed)Double-blind, randomized with crossover intervention study
  • 15 healthy postmenopausal European–American women living in central MA, USA
  • Aged mean 58.8 ± 7.9 years
7 weeks of 5 g/day seaweed (Alaria), plus 2 mg isoflavones/kg body weight during week 7; crossover after 3-week washout
  • Soy and SeaSoy (seaweed plus soy) significantly decreased serum E1, increased urinary excretion of estrogen metabolites and altered phytoestrogen metabolism
NCT01204957
(Teas et al., 2009 [210])
Genistein (with Gemcitabine)Open-label intervention study, phase II
  • 17 women with metastatic stage IV breast cancer in MI, USA
  • Aged 31 to 57
Oral genistein (100 mg) once daily on days −7 to 1, then twice daily from days 1 to 21; IV gemcitabine hydrochloride (1000 mg/m2) on days 1 and 8; course repeated up to 24 weeks
  • Study was closed early due to lack of efficacy
NCT00244933
An important measure would be to assess whether phytoestrogens regulate survival of BCSCs. For example, either recurrence-free survival or overall survival rates would be appropriate endpoints. However, there are few studies that have reported recurrence-free or overall survival outcome results, as time to these outcomes requires years of observations, and these phytoestrogen studies are rather recent. An observational study on a large cohort of ~3159 British women with invasive primary breast cancer reported that women changed their diet after diagnosis and doubled their usage of estrogenic botanical supplement (8.4% of the cohort), with flaxseed and soy isoflavone being the most common [224]. However, pre-diagnosis phytoestrogen intake was not associated with improved breast cancer diagnosis [227]. In a cohort of German postmenopausal breast cancer patients, higher serum enterolactone concentration was associated with lower all-cause mortality, breast-cancer-specific mortality and higher distant disease-free survival [225]. This increase in survival is likely mediated through enterolactone’s modification of the inflammatory marker C-reactive protein [225]. Higher concentrations of enterolactone were also associated with improved 5-year survival for patients in this cohort; however the association was only up to 4 years post-diagnosis [226]. On the other hand, genistein, resveratrol or luteolin concentrations in survivors after long-term follow-up were associated with poorer subsequent diagnosis [226]. So far, only enterolactone is reported to be associated with improved survival in breast cancer patients in observational studies. A number of studies on phytoestrogens, especially soy isoflavones, have yet to report their long-term survival outcomes. It will be important to examine the effect of interventional, placebo-controlled phytoestrogens on survival and long-term recurrence to see whether they would be effective at targeting BCSCs.

5. Conclusions

Although breast cancer treatment has significantly improved patient survival, the latest frontiers of breast cancer research focus on preventing metastasis and recurrence. As more studies unraveled the role and significance of BCSCs in resistance, recurrence and metastasis, it became evident that we must target BCSCs to completely eradicate breast cancer at the root. Numerous therapies tackling BCSCs, from stemness pathway inhibitors to immunotherapies, are being studied and tested in clinical trials. Nonetheless, there is much interest in using diet-based, natural products as chemopreventives and/or treatment adjuvants. Natural products are thought to be less toxic, cause fewer side effects, cheaper and more available/accessible to patients worldwide. Additionally, epidemiological studies report very encouraging results associating certain diets containing soy and other phytoestrogens with lower breast cancer incidence and better outcome. Among natural products studied, phytoestrogens (particularly genistein, S-equol, naringenin, resveratrol and pterostilbene) have emerged as key players with the ability to inhibit BCSCs through various mechanisms.
However, recommendations by primary physicians or oncologists regarding phytoestrogen consumption for pre/postmenopausal women or healthy/cancer patients remain unclear. There are questions about the risk to benefit ratio and which population would benefit from phytoestrogen supplementation. Some data are conflicting between different studies concerning the effect of phytoestrogens on breast cancer. Due to the biphasic nature of phytoestrogens and their binding to ERs, many phytoestrogens exhibited opposing effects on breast cancer in vitro and in vivo depending on the dosage. Furthermore, there are significant differences in phytoestrogen metabolism between mice and humans and between individuals, which further complicate the issue of dosing needed to achieve the desired clinical outcomes. The off-target effects of phytoestrogens on normal stem cells must also be considered, as these cells share many similarities with BCSCs. In addition, there is a lack of controlled interventional studies with phytoestrogens and long-term recurrence-free survival and overall survival outcomes. Although the data thus far are promising, more studies are needed to understand the mechanisms, the interactions with surrounding cells and other breast cancer treatments, as well as the effective clinical dosage in order to establish phytoestrogens as a viable anti-BCSC therapy. Ultimately, the hope is that patients could easily supplement their diet with phytoestrogens to prevent breast cancer and/or recurrence, with little toxicity and side effects.

Author Contributions

Conceptualization, C.O. and M.N.; Writing—original draft preparation, M.N.; Writing—review and editing, C.O. and M.N.; All authors have read and agreed to the published version of the manuscript.

Funding

Mai Nguyen was funded by the National Institute of Health T32 (AI007508-21) Fellowship in Immunology; Clodia Osipo was supported by the Breast Cancer Research Foundation (BCRF-21-003).

Institutional Review Board Statement

Not Applicable.

Informed Consent Statement

Not Applicable.

Data Availability Statement

Not Applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
  2. Zeng, X.; Liu, C.; Yao, J.; Wan, H.; Wan, G.; Li, Y.; Chen, N. Breast cancer stem cells, heterogeneity, targeting therapies and therapeutic implications. Pharmacol. Res. 2021, 163, 105320. [Google Scholar] [CrossRef] [PubMed]
  3. Sin, W.C.; Lim, C.L. Breast cancer stem cells—From origins to targeted therapy. Stem Cell Investig. 2017, 4, 96. [Google Scholar] [CrossRef] [Green Version]
  4. Koren, S.; Bentires-Alj, M. Breast Tumor Heterogeneity: Source of Fitness, Hurdle for Therapy. Mol. Cell 2015, 60, 537–546. [Google Scholar] [CrossRef]
  5. Rabinovich, I.; Sebastião, A.P.M.; Lima, R.S.; de Andrade Urban, C.; Schunemann, E., Jr.; Anselmi, K.F.; Elifio-Esposito, S.; De Noronha, L.; Moreno-Amaral, A.N. Cancer stem cell markers ALDH1 and CD44+/CD24- phenotype and their prognosis impact in invasive ductal carcinoma. Eur. J. Histochem. 2018, 62, 2943. [Google Scholar] [CrossRef] [PubMed]
  6. Kakarala, M.; Wicha, M.S. Implications of the Cancer Stem-Cell Hypothesis for Breast Cancer Prevention and Therapy. J. Clin. Oncol. 2008, 26, 2813–2820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Van Diest, P.J.; Vooijs, M. Mammary Development and Breast Cancer: The Role of Stem Cells. Curr. Mol. Med. 2011, 11, 270–285. [Google Scholar] [CrossRef] [Green Version]
  8. Stingl, J.; Eaves, C.J.; Zandieh, I.; Emerman, J.T. Characterization of bipotent mammary epithelial progenitor cells in normal adult human breast tissue. Breast Cancer Res. Treat. 2001, 67, 93–109. [Google Scholar] [CrossRef]
  9. Khan, S.; Suryavanshi, M.; Kaur, J.; Nayak, D.; Khurana, A.; Manchanda, R.K.; Tandon, C.; Tandon, S. Stem cell therapy: A paradigm shift in breast cancer treatment. World J. Stem Cells 2021, 13, 841–860. [Google Scholar] [CrossRef]
  10. Villadsen, R.; Fridriksdottir, A.; Ronnov-Jessen, L.; Gudjonsson, T.; Rank, F.; LaBarge, M.; Bissell, M.J.; Petersen, O.W. Evidence for a stem cell hierarchy in the adult human breast. J. Cell Biol. 2007, 177, 87–101. [Google Scholar] [CrossRef]
  11. Tharmapalan, P.; Mahendralingam, M.; Berman, H.K.; Khokha, R. Mammary stem cells and progenitors: Targeting the roots of breast cancer for prevention. EMBO J. 2019, 38, e100852. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, S.; Cong, Y.; Wang, D.; Sun, Y.; Deng, L.; Liu, Y.; Martin-Trevino, R.; Shang, L.; McDermott, S.P.; Landis, M.D.; et al. Breast cancer stem cells transition between epithelial and mesenchymal states reflective of their normal counterparts. Stem Cell Rep. 2014, 2, 78–91. [Google Scholar] [CrossRef]
  13. Bao, L.; Cardiff, R.D.; Steinbach, P.; Messer, K.S.; Ellies, L.G. Multipotent luminal mammary cancer stem cells model tumor heterogeneity. Breast Cancer Res. 2015, 17, 137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lim, E.; Vaillant, F.; Wu, D.; Forrest, N.C.; Pal, B.; Hart, A.H.; Asselin-Labat, M.-L.; Gyorki, D.E.; Ward, T.; Partanen, A.; et al. Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat. Med. 2009, 15, 907–913. [Google Scholar] [CrossRef]
  15. Zhang, X.; Powell, K.; Li, L. Breast Cancer Stem Cells: Biomarkers, Identification and Isolation Methods, Regulating Mechanisms, Cellular Origin, and Beyond. Cancers 2020, 12, 3765. [Google Scholar] [CrossRef] [PubMed]
  16. Scioli, M.G.; Storti, G.; D’Amico, F.; Gentile, P.; Fabbri, G.; Cervelli, V.; Orlandi, A. The Role of Breast Cancer Stem Cells as a Prognostic Marker and a Target to Improve the Efficacy of Breast Cancer Therapy. Cancers 2019, 11, 1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Lapidot, T.; Sirard, C.; Vormoor, J.; Murdoch, B.; Hoang, T.; Caceres-Cortes, J.; Minden, M.; Paterson, B.; Caligiuri, M.A.; Dick, J.E. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994, 367, 645–648. [Google Scholar] [CrossRef] [PubMed]
  18. Tanei, T.; Morimoto, K.; Shimazu, K.; Kim, S.J.; Tanji, Y.; Taguchi, T.; Tamaki, Y.; Noguchi, S. Association of Breast Cancer Stem Cells Identified by Aldehyde Dehydrogenase 1 Expression with Resistance to Sequential Paclitaxel and Epirubicin-Based Chemotherapy for Breast Cancers. Clin. Cancer Res. 2009, 15, 4234–4241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Ricardo, S.; Vieira, A.F.; Gerhard, R.; Leitão, D.; Pinto, R.; Cameselle-Teijeiro, J.F.; Milanezi, F.; Schmitt, F.; Paredes, J. Breast cancer stem cell markers CD44, CD24 and ALDH1: Expression distribution within intrinsic molecular subtype. J. Clin. Pathol. 2011, 64, 937–946. [Google Scholar] [CrossRef]
  20. Honeth, G.; Bendahl, P.-O.; Ringnér, M.; SaaL, L.H.; Gruvberger-Saal, S.K.; Lövgren, K.; Grabau, D.; Fernö, M.; Borg, Å.; Hegardt, C. The CD44+/CD24- phenotype is enriched in basal-like breast tumors. Breast Cancer Res. 2008, 10, R53. [Google Scholar] [CrossRef] [Green Version]
  21. Beca, F.; Caetano, P.; Gerhard, R.; Alvarenga, C.A.; Gomes, M.; Paredes, J.; Schmitt, F. Cancer stem cells markers CD44, CD24 and ALDH1 in breast cancer special histological types. J. Clin. Pathol. 2012, 66, 187–191. [Google Scholar] [CrossRef] [PubMed]
  22. Al-Hajj, M.; Wicha, M.S.; Benito-Hernandez, A.; Morrison, S.J.; Clarke, M.F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. USA 2003, 100, 3983–3988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ponti, D.; Costa, A.; Zaffaroni, N.; Pratesi, G.; Petrangolini, G.; Coradini, D.; Pilotti, S.; Pierotti, M.A.; Daidone, M.G. Isolation and In vitro Propagation of Tumorigenic Breast Cancer Cells with Stem/Progenitor Cell Properties. Cancer Res. 2005, 65, 5506–5511. [Google Scholar] [CrossRef] [Green Version]
  24. Fillmore, C.M.; Kuperwasser, C. Human breast cancer cell lines contain stem-like cells that self-renew, give rise to phenotypically diverse progeny and survive chemotherapy. Breast Cancer Res. 2008, 10, R25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Charafe-Jauffret, E.; Ginestier, C.; Iovino, F.; Wicinski, J.; Cervera, N.; Finetti, P.; Hur, M.-H.; Diebel, M.E.; Monville, F.; Dutcher, J.; et al. Breast Cancer Cell Lines Contain Functional Cancer Stem Cells with Metastatic Capacity and a Distinct Molecular Signature. Cancer Res. 2009, 69, 1302–1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Bartucci, M.; Dattilo, R.; Moriconi, C.; Pagliuca, A.; Mottolese, M.; Federici, G.; Di Benedetto, A.; Todaro, M.; Stassi, G.; Sperati, F.; et al. TAZ is required for metastatic activity and chemoresistance of breast cancer stem cells. Oncogene 2014, 34, 681–690. [Google Scholar] [CrossRef]
  27. Li, W.; Ma, H.; Zhang, J.; Zhu, L.; Wang, C.; Yang, Y. Unraveling the roles of CD44/CD24 and ALDH1 as cancer stem cell markers in tumorigenesis and metastasis. Sci. Rep. 2017, 7, 13856. [Google Scholar] [CrossRef] [Green Version]
  28. Yin, H.; Glass, J. The phenotypic radiation resistance of CD44+/CD24(-or low) breast cancer cells is mediated through the enhanced activation of ATM signaling. PLoS ONE 2011, 6, e24080. [Google Scholar] [CrossRef]
  29. Palomeras, S.; Ruiz-Martínez, S.; Puig, T. Targeting Breast Cancer Stem Cells to Overcome Treatment Resistance. Molecules 2018, 23, 2193. [Google Scholar] [CrossRef] [Green Version]
  30. Creighton, C.J.; Li, X.; Landis, M.; Dixon, J.M.; Neumeister, V.M.; Sjolund, A.; Rimm, D.L.; Wong, H.; Rodriguez, A.; Herschkowitz, J.I.; et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc. Natl. Acad. Sci. USA 2009, 106, 13820–13825. [Google Scholar] [CrossRef] [Green Version]
  31. Harrison, H.; Farnie, G.; Howell, S.J.; Rock, R.E.; Stylianou, S.; Brennan, K.R.; Bundred, N.J.; Clarke, R.B. Regulation of Breast Cancer Stem Cell Activity by Signaling through the Notch4 Receptor. Cancer Res. 2010, 70, 709–718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Suman, S.; Das, T.P.; Damodaran, C. Silencing NOTCH signaling causes growth arrest in both breast cancer stem cells and breast cancer cells. Br. J. Cancer 2013, 109, 2587–2596. [Google Scholar] [CrossRef] [PubMed]
  33. Monteiro, J.; Gaspar, C.; Richer, W.; Franken, P.F.; Sacchetti, A.; Joosten, R.; Idali, A.; Brandao, J.; Decraene, C.; Fodde, R. Cancer stemness in Wnt-driven mammary tumorigenesis. Carcinogenesis 2013, 35, 2–13. [Google Scholar] [CrossRef] [Green Version]
  34. Jang, G.B.; Kim, J.Y.; Cho, S.D.; Park, K.S.; Jung, J.Y.; Lee, H.Y.; Hong, I.S.; Nam, J.S. Blockade of Wnt/β-catenin signaling suppresses breast cancer metastasis by inhibiting CSC-like phenotype. Sci. Rep. 2015, 5, 12465. [Google Scholar] [CrossRef] [Green Version]
  35. Liu, S.; Dontu, G.; Mantle, I.D.; Patel, S.; Ahn, N.-S.; Jackson, K.W.; Suri, P.; Wicha, M.S. Hedgehog Signaling and Bmi-1 Regulate Self-renewal of Normal and Malignant Human Mammary Stem Cells. Cancer Res. 2006, 66, 6063–6071. [Google Scholar] [CrossRef] [Green Version]
  36. Smith, S.M.; Lyu, Y.L.; Cai, L. NF-κB affects proliferation and invasiveness of breast cancer cells by regulating CD44 expression. PLoS ONE 2014, 9, e106966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Liu, S.; Ginestier, C.; Charafe-Jauffret, E.; Foco, H.; Kleer, C.G.; Merajver, S.D.; Dontu, G.; Wicha, M.S. BRCA1 regulates human mammary stem/progenitor cell fate. Proc. Natl. Acad. Sci. USA 2008, 105, 1680–1685. [Google Scholar] [CrossRef] [Green Version]
  38. Ithimakin, S.; Day, K.C.; Malik, F.; Zen, Q.; Dawsey, S.J.; Bersano-Begey, T.F.; Quraishi, A.A.; Ignatoski, K.; Daignault, S.; Davis, A.; et al. HER2 Drives Luminal Breast Cancer Stem Cells in the Absence of HER2 Amplification: Implications for Efficacy of Adjuvant Trastuzumab. Cancer Res. 2013, 73, 1635–1646. [Google Scholar] [CrossRef] [Green Version]
  39. Duru, N.; Fan, M.; Candas, D.; Menaa, C.; Liu, H.-C.; Nantajit, D.; Wen, Y.; Xiao, K.; Eldridge, A.; Chromy, B.A.; et al. HER2-Associated Radioresistance of Breast Cancer Stem Cells Isolated from HER2-Negative Breast Cancer Cells. Clin. Cancer Res. 2012, 18, 6634–6647. [Google Scholar] [CrossRef] [Green Version]
  40. Diessner, J.; Bruttel, V.; Stein, R.G.; Horn, E.; Häusler, S.F.M.; Dietl, J.; Hönig, A.; Wischhusen, J. Targeting of preexisting and induced breast cancer stem cells with trastuzumab and trastuzumab emtansine (T-DM1). Cell Death Dis. 2014, 5, e1149. [Google Scholar] [CrossRef] [Green Version]
  41. Baker, A.; Wyatt, D.; Bocchetta, M.; Li, J.; Filipovic, A.; Green, A.; Peiffer, D.S.; Fuqua, S.; Miele, L.; Albain, K.S.; et al. Notch-1-PTEN-ERK1/2 signaling axis promotes HER2+ breast cancer cell proliferation and stem cell survival. Oncogene 2018, 37, 4489–4504. [Google Scholar] [CrossRef] [PubMed]
  42. Korkaya, H.; Paulson, A.; Charafe-Jauffret, E.; Ginestier, C.; Brown, M.; Dutcher, J.; Clouthier, S.G.; Wicha, M.S. Regulation of Mammary Stem/Progenitor Cells by PTEN/Akt/β-Catenin Signaling. PLoS Biol. 2009, 7, e1000121. [Google Scholar] [CrossRef] [PubMed]
  43. Sousa, B.; Ribeiro, A.S.; Paredes, J. Heterogeneity and Plasticity of Breast Cancer Stem Cells. In Advances in Experimental Medicine and Biology; Springer: Berlin/Heidelberg, Germany, 2019; in press. [Google Scholar]
  44. Zu, X.; Zhang, Q.; Cao, R.; Liu, J.; Zhong, J.; Wen, G.; Cao, D. Transforming growth factor-β signaling in tumor initiation, progression and therapy in breast cancer: An update. Cell Tissue Res. 2012, 347, 73–84. [Google Scholar] [CrossRef] [PubMed]
  45. Hung, S.-P.; Yang, M.-H.; Tseng, K.-F.; Lee, O.K. Hypoxia-Induced Secretion of TGF-β1 in Mesenchymal Stem Cell Promotes Breast Cancer Cell Progression. Cell Transplant. 2013, 22, 1869–1882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Buckley, N.E.; Mullan, P.B. BRCA1–Conductor of the Breast Stem Cell Orchestra: The Role of BRCA1 in Mammary Gland Development and Identification of Cell of Origin of BRCA1 Mutant Breast Cancer. Stem Cell Rev. Rep. 2012, 8, 982–993. [Google Scholar] [CrossRef] [PubMed]
  47. Wright, M.H.; Calcagno, A.M.; Salcido, C.D.; Carlson, M.D.; Ambudkar, S.V.; Varticovski, L. Brca1 breast tumors contain distinct CD44+/CD24- and CD133+ cells with cancer stem cell characteristics. Breast Cancer Res. 2008, 10, R10. [Google Scholar] [CrossRef] [Green Version]
  48. Cordenonsi, M.; Zanconato, F.; Azzolin, L.; Forcato, M.; Rosato, A.; Frasson, C.; Inui, M.; Montagner, M.; Parenti, A.R.; Poletti, A.; et al. The Hippo Transducer TAZ Confers Cancer Stem Cell-Related Traits on Breast Cancer Cells. Cell 2011, 147, 759–772. [Google Scholar] [CrossRef]
  49. Britschgi, A.; Duss, S.; Kim, S.; Couto, J.P.; Brinkhaus, H.; Koren, S.; De Silva, D.; Mertz, K.; Kaup, D.; Varga, Z.; et al. The Hippo kinases LATS1 and 2 control human breast cell fate via crosstalk with ERα. Nature 2017, 541, 541–545. [Google Scholar] [CrossRef]
  50. Yamamoto, M.; Taguchi, Y.; Ito-Kureha, T.; Semba, K.; Yamaguchi, N.; Inoue, J.I. NF-κB non-cell-autonomously regulates cancer stem cell populations in the basal-like breast cancer subtype. Nat. Commun. 2013, 4, 2299. [Google Scholar] [CrossRef] [Green Version]
  51. Rinkenbaugh, A.L.; Baldwin, A.S. The NF-κB Pathway and Cancer Stem Cells. Cells 2016, 5, 16. [Google Scholar] [CrossRef]
  52. Liu, M.; Sakamaki, T.; Casimiro, M.C.; Willmarth, N.E.; Quong, A.A.; Ju, X.; Ojeifo, J.; Jiao, X.; Yeow, W.S.; Katiyar, S.; et al. The canonical NF-kappaB pathway governs mammary tumorigenesis in transgenic mice and tumor stem cell expansion. Cancer Res. 2010, 70, 10464–10473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Pratt, M.A.C.; Tibbo, E.; Robertson, S.J.; Jansson, D.; Hurst, K.; Perez-Iratxeta, C.; Lau, R.; Niu, M.Y. The canonical NF-kappaB pathway is required for formation of luminal mammary neoplasias and is activated in the mammary progenitor population. Oncogene 2009, 28, 2710–2722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kendellen, M.F.; Bradford, J.W.; Lawrence, C.L.; Clark, K.S.; Baldwin, A.S. Canonical and non-canonical NF-κB signaling promotes breast cancer tumor-initiating cells. Oncogene 2014, 33, 1297–1305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Schwab, L.P.; Peacock, D.L.; Majumdar, D.; Ingels, J.F.; Jensen, L.C.; Smith, K.D.; Cushing, R.C.; Seagroves, T.N. Hypoxia-inducible factor 1α promotes primary tumor growth and tumor-initiating cell activity in breast cancer. Breast Cancer Res. 2012, 14, R6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Xing, F.; Okuda, H.; Watabe, M.; Kobayashi, A.; Pai, S.K.; Liu, W.; Pandey, P.R.; Fukuda, K.; Hirota, S.; Sugai, T.; et al. Hypoxia-induced Jagged2 promotes breast cancer metastasis and self-renewal of cancer stem-like cells. Oncogene 2011, 30, 4075–4086. [Google Scholar] [CrossRef] [Green Version]
  57. Marotta, L.L.; Almendro, V.; Marusyk, A.; Shipitsin, M.; Schemme, J.; Walker, S.R.; Bloushtain-Qimron, N.; Kim, J.J.; Choudhury, S.A.; Maruyama, R. The JAK2/STAT3 signaling pathway is required for growth of CD44+CD24 stem cell-like breast cancer cells in human tumors. J. Clin. Investig. 2011, 121, 2723–2735. [Google Scholar] [CrossRef]
  58. Wei, W.; Tweardy, D.J.; Zhang, M.; Zhang, X.; Landua, J.; Petrovic, I.; Bu, W.; Roarty, K.; Hilsenbeck, S.G.; Rosen, J.M.; et al. STAT3 Signaling Is Activated Preferentially in Tumor-Initiating Cells in Claudin-Low Models of Human Breast Cancer. Stem Cells 2014, 32, 2571–2582. [Google Scholar] [CrossRef] [Green Version]
  59. Hernández-Vargas, H.; Ouzounova, M.; Le Calvez-Kelm, F.; Lambert, M.-P.; McKay-Chopin, S.; Tavtigian, S.V.; Puisieux, A.; Matar, C.; Herceg, Z. Methylome analysis reveals Jak-STAT pathway deregulation in putative breast cancer stem cells. Epigenetics 2011, 6, 428–439. [Google Scholar] [CrossRef] [Green Version]
  60. Pires, B.R.B.; DE Amorim, S.S.; Souza, L.D.E.; Rodrigues, J.A.; Mencalha, A.L. Targeting Cellular Signaling Pathways in Breast Cancer Stem Cells and its Implication for Cancer Treatment. Anticancer Res. 2016, 36, 5681–5692. [Google Scholar] [CrossRef] [Green Version]
  61. Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Zhang, G.; Wang, X.; Dong, Z.; Chen, F.; et al. Targeting Cancer Stem Cell Pathways for Cancer Therapy. Signal Transduct. Target. Ther. 2020, 5, 8. [Google Scholar] [CrossRef] [Green Version]
  62. Muthusamy, B.P.; Budi, E.H.; Katsuno, Y.; Lee, M.K.; Smith, S.M.; Mirza, A.M.; Akhurst, R.J.; Derynck, R. ShcA Protects against Epithelial–Mesenchymal Transition through Compartmentalized Inhibition of TGF-beta-Induced Smad Activation. PLoS Biol. 2015, 13, e1002325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yu, F.; Yao, H.; Zhu, P.; Zhang, X.; Pan, Q.; Gong, C.; Huang, Y.; Hu, X.; Su, F.; Lieberman, J.; et al. let-7 Regulates Self Renewal and Tumorigenicity of Breast Cancer Cells. Cell 2007, 131, 1109–1123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Yu, F.; Deng, H.; Yao, H.; Liu, Q.; Su, F.; Song, E. Mir-30 reduction maintains self-renewal and inhibits apoptosis in breast tumor-initiating cells. Oncogene 2010, 29, 4194–4204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Wu, D.; Zhang, J.; Lu, Y.; Bo, S.; Li, L.; Wang, L.; Zhang, Q.; Mao, J. miR-140-5p inhibits the proliferation and enhances the efficacy of doxorubicin to breast cancer stem cells by targeting Wnt1. Cancer Gene Ther. 2018, 26, 74–82. [Google Scholar] [CrossRef]
  66. Zhang, H.; Cai, K.; Wang, J.; Wang, X.; Cheng, K.; Shi, F.; Jiang, L.; Zhang, Y.; Dou, J. MiR-7, Inhibited Indirectly by LincRNA HOTAIR, Directly Inhibits SETDB1 and Reverses the EMT of Breast Cancer Stem Cells by Downregulating the STAT3 Pathway. Stem Cells 2014, 32, 2858–2868. [Google Scholar] [CrossRef]
  67. Zhou, M.; Hou, Y.; Yang, G.; Zhang, H.; Tu, G.; Du, Y.E.; Wen, S.; Xu, L.; Tang, X.; Tang, S.; et al. LncRNA-Hh strengthen cancer stem cells generation in twist-positive breast cancer via activation of hedgehog signaling pathway. Stem Cells 2016, 34, 55–66. [Google Scholar] [CrossRef] [Green Version]
  68. Diehn, M.; Cho, R.W.; Clarke, M.F. Therapeutic Implications of the Cancer Stem Cell Hypothesis. Semin. Radiat. Oncol. 2009, 19, 78–86. [Google Scholar] [CrossRef] [Green Version]
  69. Brown, J.; Yonekubo, Y.; Hanson, N.; Sastre-Perona, A.; Basin, A.; Rytlewski, J.; Dolgalev, I.; Meehan, S.; Tsirigos, A.; Beronja, S.; et al. TGF-β-Induced Quiescence Mediates Chemoresistance of Tumor-Propagating Cells in Squamous Cell Carcinoma. Cell Stem Cell 2017, 21, 650–664.e8. [Google Scholar] [CrossRef]
  70. Singh, S.; Brocker, C.; Koppaka, V.; Chen, Y.; Jackson, B.C.; Matsumoto, A.; Thompson, D.C.; Vasiliou, V. Aldehyde dehydrogenases in cellular responses to oxidative/electrophilicstress. Free Radic. Biol. Med. 2013, 56, 89–101. [Google Scholar] [CrossRef] [Green Version]
  71. Petrovska, B.B. Historical review of medicinal plants’ usage. Pharmacogn. Rev. 2012, 6, 1–5. [Google Scholar] [CrossRef] [Green Version]
  72. Taylor, W.F.; Jabbarzadeh, E. The use of natural products to target cancer stem cells. Am. J. Cancer Res. 2017, 7, 1588–1605. [Google Scholar] [PubMed]
  73. Zhou, Q.; Ye, M.; Lu, Y.; Zhang, H.; Chen, Q.; Huang, S.; Su, S. Curcumin Improves the Tumoricidal Effect of Mitomycin C by Suppressing ABCG2 Expression in Stem Cell-Like Breast Cancer Cells. PLoS ONE 2015, 10, e0136694. [Google Scholar] [CrossRef] [PubMed]
  74. Chung, S.S.; Vadgama, J.V. Curcumin and epigallocatechin gallate inhibit the cancer stem cell phenotype via down-regulation of STAT3-NFκB signaling. Anticancer Res. 2015, 35, 39–46. [Google Scholar] [PubMed]
  75. Kakarala, M.; Brenner, D.E.; Korkaya, H.; Cheng, C.; Tazi, K.; Ginestier, C.; Liu, S.; Dontu, G.; Wicha, M.S. Targeting breast stem cells with the cancer preventive compounds curcumin and piperine. Breast Cancer Res. Treat. 2009, 122, 777–785. [Google Scholar] [CrossRef] [Green Version]
  76. Mineva, N.D.; Paulson, K.E.; Naber, S.P.; Yee, A.S.; Sonenshein, G.E. Epigallocatechin-3-Gallate Inhibits Stem-Like Inflammatory Breast Cancer Cells. PLoS ONE 2013, 8, e73464. [Google Scholar] [CrossRef] [Green Version]
  77. Li, Y.; Zhang, T.; Korkaya, H.; Liu, S.; Lee, H.-F.; Newman, B.; Yu, Y.; Clouthier, S.G.; Schwartz, S.J.; Wicha, M.S.; et al. Sulforaphane, a Dietary Component of Broccoli/Broccoli Sprouts, Inhibits Breast Cancer Stem Cells. Clin. Cancer Res. 2010, 16, 2580–2590. [Google Scholar] [CrossRef] [Green Version]
  78. Li, Q.; Eades, G.; Yao, Y.; Zhang, Y.; Zhou, Q. Characterization of a Stem-like Subpopulation in Basal-like Ductal Carcinoma in Situ (DCIS) Lesions. J. Biol. Chem. 2014, 289, 1303–1312. [Google Scholar] [CrossRef] [Green Version]
  79. Dandawate, P.; Subramaniam, D.; Jensen, R.A.; Anant, S. Targeting cancer stem cells and signaling pathways by phytochemicals: Novel approach for breast cancer therapy. Semin. Cancer Biol. 2016, 40, 192–208. [Google Scholar] [CrossRef] [Green Version]
  80. Naujokat, C.; Steinhart, R. Salinomycin as a Drug for Targeting Human Cancer Stem Cells. J. Biomed. Biotechnol. 2012, 2012, 1–17. [Google Scholar] [CrossRef]
  81. Oak, P.S.; Kopp, F.; Thakur, C.; Ellwart, J.W.; Rapp, U.R.; Ullrich, A.; Wagner, E.; Knyazev, P.; Roidl, A. Combinatorial treatment of mammospheres with trastuzumab and salinomycin efficiently targets HER2-positive cancer cells and cancer stem cells. Int. J. Cancer 2012, 131, 2808–2819. [Google Scholar] [CrossRef]
  82. Hermawan, A.; Ikawati, M.; Jenie, R.I.; Khumaira, A.; Putri, H.; Nurhayati, I.P.; Angraini, S.M.; Muflikhasari, H.A. Identification of potential therapeutic target of naringenin in breast cancer stem cells inhibition by bioinformatics and in vitro studies. Saudi Pharm. J. 2020, 29, 12–26. [Google Scholar] [CrossRef] [PubMed]
  83. Rossouw, J.E.; Anderson, G.L.; Prentice, R.L.; LaCroix, A.Z.; Kooperberg, C.; Stefanick, M.L.; Jackson, R.D.; Beresford, S.A.; Howard, B.V.; Johnson, K.C.; et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: Principal results From the Women’s Health Initiative randomized controlled trial. JAMA 2002, 288, 321–333. [Google Scholar] [PubMed] [Green Version]
  84. Moore, T.R.; Franks, R.B.; Fox, C. Review of Efficacy of Complementary and Alternative Medicine Treatments for Menopausal Symptoms. J. Midwifery Women’s Health 2017, 62, 286–297. [Google Scholar] [CrossRef] [PubMed]
  85. Thaung Zaw, J.J.; Howe, P.R.C.; Wong, R.H.X. Postmenopausal health interventions: Time to move on from the Women’s Health Initiative? Ageing Res. Rev. 2018, 48, 79–86. [Google Scholar] [CrossRef]
  86. Bennetts, H.; Underwood, E.; Shier, P. A Specific Breeding Problem of Sheep on Subterranean Clover Pastures in Western Australia. Vet. J. 1946, 102, 348–352. [Google Scholar] [CrossRef]
  87. Senthilkumar, H.A.; Fata, J.E.; Kennelly, E.J. Phytoestrogens: The current state of research emphasizing breast pathophysiology. Phytotherapy Res. 2018, 32, 1707–1719. [Google Scholar] [CrossRef]
  88. Lecomte, S.; Demay, F.; Ferrière, F.; Pakdel, F. Phytochemicals Targeting Estrogen Receptors: Beneficial Rather Than Adverse Effects? Int. J. Mol. Sci. 2017, 18, 1381. [Google Scholar] [CrossRef] [Green Version]
  89. Křížová, L.; Dadáková, K.; Kašparovská, J.; Kašparovský, T. Isoflavones. Molecules 2019, 24, 1076. [Google Scholar] [CrossRef] [Green Version]
  90. Cos, P.; De Bruyne, T.; Apers, S.; Van Dem Berghe, D.; Pieters, L.; Vlietinck, A.J. Phytoestrogens: Recent Developments. Plant. Med. 2003, 69, 589–599. [Google Scholar] [CrossRef] [Green Version]
  91. Glazier, M.G.; Bowman, M.A. A Review of the Evidence for the Use of Phytoestrogens as a Replacement for Traditional Estrogen Replacement Therapy. Arch. Intern. Med. 2001, 161, 1161–1172. [Google Scholar] [CrossRef] [Green Version]
  92. Jung, W.; Yu, O.; Lau, S.M.C.; O’Keefe, D.P.; Odell, J.; Fader, G.; McGonigle, B. Identification and expression of isoflavone synthase, the key enzyme for biosynthesis of isoflavones in legumes. Nat. Biotechnol. 2000, 18, 208–212. [Google Scholar] [CrossRef] [PubMed]
  93. Setchell, K.D.R.; Clerici, C.; Lephart, E.D.; Cole, S.J.; Heenan, C.; Castellani, D.; Wolfe, B.E.; Nechemias-Zimmer, L.; Brown, N.M.; Lund, T.D.; et al. 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] [PubMed] [Green Version]
  94. 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]
  95. Setchell, K.D.R.; Cole, S.J. Method of Defining Equol-Producer Status and Its Frequency among Vegetarians. J. Nutr. 2006, 136, 2188–2193. [Google Scholar] [CrossRef] [Green Version]
  96. Kuiper, G.G.J.M.; Lemmen, J.G.; Carlsson, B.; Corton, J.C.; Safe, S.H.; Van Der Saag, P.T.; Van Der Burg, B.; Gustafsson, J.Å. Interaction of Estrogenic Chemicals and Phytoestrogens with Estrogen Receptor β. Endocrinology 1998, 139, 4252–4263. [Google Scholar] [CrossRef]
  97. Jiang, Y.; Gong, P.; Madak-Erdogan, Z.; Martin, T.; Jeyakumar, M.; Carlson, K.; Khan, I.; Smillie, T.J.; Chittiboyina, A.G.; Rotte, S.C.K.; et al. Mechanisms enforcing the estrogen receptor β selectivity of botanical estrogens. FASEB J. 2013, 27, 4406–4418. [Google Scholar] [CrossRef] [Green Version]
  98. Thomas, C.; Gustafsson, J. The different roles of ER subtypes in cancer biology and therapy. Nat. Cancer 2011, 11, 597–608. [Google Scholar] [CrossRef]
  99. Treeck, O.; Lattrich, C.; Springwald, A.; Ortmann, O. Estrogen receptor beta exerts growth-inhibitory effects on human mammary epithelial cells. Breast Cancer Res. Treat. 2009, 120, 557–565. [Google Scholar] [CrossRef]
  100. Sotoca, A.; Ratman, D.; van der Saag, P.; Ström, A.; Gustafsson, J.; Vervoort, J.; Rietjens, I.; Murk, A. Phytoestrogen-mediated inhibition of proliferation of the human T47D breast cancer cells depends on the ERα/ERβ ratio. J. Steroid Biochem. Mol. Biol. 2008, 112, 171–178. [Google Scholar] [CrossRef]
  101. Pons, D.G.; Nadal-Serrano, M.; Blanquer-Rossello, M.M.; Sastre-Serra, J.; Oliver, J.; Roca, P. Genistein Modulates Proliferation and Mitochondrial Functionality in Breast Cancer Cells Depending on ERalpha/ERbeta Ratio. J. Cell. Biochem. 2014, 115, 949–958. [Google Scholar] [CrossRef]
  102. Beecher, G.R. Overview of Dietary Flavonoids: Nomenclature, Occurrence and Intake. J. Nutr. 2003, 133, 3248S–3254S. [Google Scholar] [CrossRef] [PubMed]
  103. Adlercreutz, H.; Mazur, W. Phyto-oestrogens and Western Diseases. Ann. Med. 1997, 29, 95–120. [Google Scholar] [CrossRef] [PubMed]
  104. Korde, L.A.; Wu, A.H.; Fears, T.; Nomura, A.M.; West, D.W.; Kolonel, L.N.; Pike, M.C.; Hoover, R.N.; Ziegler, R.G. Childhood Soy Intake and Breast Cancer Risk in Asian American Women. Cancer Epidemiol. Biomark. Prev. 2009, 18, 1050–1059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Shu, X.O.; Jin, F.; Dai, Q.; Wen, W.; Potter, J.; Kushi, L.; Ruan, Z.; Gao, Y.T.; Zheng, W. Soyfood intake during adolescence and subsequent risk of breast cancer among Chinese women. Cancer Epidemiol. Biomark. Prev. 2001, 10, 483–488. [Google Scholar]
  106. Thanos, J.; Cotterchio, M.; Boucher, B.A.; Kreiger, N.; Thompson, L.U. Adolescent dietary phytoestrogen intake and breast cancer risk (Canada). Cancer Causes Control 2006, 17, 1253–1261. [Google Scholar] [CrossRef]
  107. Wu, A.H.; Wan, P.; Hankin, J.; Tseng, C.-C.; Yu, M.C.; Pike, M.C. Adolescent and adult soy intake and risk of breast cancer in Asian-Americans. Carcinogenesis 2002, 23, 1491–1496. [Google Scholar] [CrossRef]
  108. Warri, A.; Saarinen, N.; Makela, S.M.; Hilakiviclarke, L. The role of early life genistein exposures in modifying breast cancer risk. Br. J. Cancer 2008, 98, 1485–1493. [Google Scholar] [CrossRef] [Green Version]
  109. Chi, F.; Wu, R.; Zeng, Y.-C.; Xing, R.; Liu, Y.; Xu, Z.-G. Post-diagnosis Soy Food Intake and Breast Cancer Survival: A Meta-analysis of Cohort Studies. Asian Pac. J. Cancer Prev. 2013, 14, 2407–2412. [Google Scholar] [CrossRef] [Green Version]
  110. Shu, X.O.; Zheng, Y.; Cai, H.; Gu, K.; Chen, Z.; Zheng, W.; Lu, W. Soy Food Intake and Breast Cancer Survival. JAMA 2009, 302, 2437–2443. [Google Scholar] [CrossRef] [Green Version]
  111. Liu, R.; Yu, X.; Chen, X.; Zhong, H.; Liang, C.; Xu, X.; Xu, W.; Cheng, Y.; Wang, W.; Yu, L.; et al. Individual factors define the overall effects of dietary genistein exposure on breast cancer patients. Nutr. Res. 2019, 67, 1–16. [Google Scholar] [CrossRef]
  112. Ahn, S.-Y.; Jo, M.S.; Lee, D.; Baek, S.-E.; Baek, J.; Yu, J.S.; Jo, J.; Yun, H.; Kang, K.S.; Yoo, J.-E.; et al. Dual effects of isoflavonoids from Pueraria lobata roots on estrogenic activity and anti-proliferation of MCF-7 human breast carcinoma cells. Bioorganic Chem. 2018, 83, 135–144. [Google Scholar] [CrossRef] [PubMed]
  113. Han, R.; Gu, S.; Zhang, Y.; Luo, A.; Jing, X.; Zhao, L.; Zhao, X.; Zhang, L. Estrogen promotes progression of hormone-dependent breast cancer through CCL2-CCR2 axis by upregulation of Twist via PI3K/AKT/NF-κB signaling. Sci. Rep. 2018, 8, 9575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Hsieh, C.Y.; Santell, R.C.; Haslam, S.Z.; Helferich, W.G. Estrogenic Effects of Genistein on the Growth of Estrogen Receptor-positive Human Breast Cancer (MCF-7) Cells in vitro and in vivo. Cancer Res. 1998, 58, 3833. [Google Scholar] [PubMed]
  115. Bhat, S.S.; Prasad, S.K.; Shivamallu, C.; Prasad, K.S.; Syed, A.; Reddy, P.; Cull, C.A.; Amachawadi, R.G. Genistein: A Potent Anti-Breast Cancer Agent. Curr. Issues Mol. Biol. 2021, 43, 1502–1517. [Google Scholar] [CrossRef]
  116. Yu, X.; Zhu, J.; Mi, M.; Chen, W.; Pan, Q.; Wei, M. Anti-angiogenic genistein inhibits VEGF-induced endothelial cell activation by decreasing PTK activity and MAPK activation. Med. Oncol. 2010, 29, 349–357. [Google Scholar] [CrossRef]
  117. Jiang, H.; Fan, J.; Cheng, L.; Hu, P.; Liu, R. The anticancer activity of genistein is increased in estrogen receptor beta 1-positive breast cancer cells. OncoTargets Ther. 2018, 11, 8153–8163. [Google Scholar] [CrossRef] [Green Version]
  118. Zhao, Q.; Zhao, M.; Parris, A.B.; Xing, Y.; Yang, X. Genistein targets the cancerous inhibitor of PP2A to induce growth inhibition and apoptosis in breast cancer cells. Int. J. Oncol. 2016, 49, 1203–1210. [Google Scholar] [CrossRef] [Green Version]
  119. Montales, M.T.E.; Rahal, O.M.; Kang, J.; Rogers, T.J.; Prior, R.L.; Wu, X.; Simmen, R.C. Repression of mammosphere formation of human breast cancer cells by soy isoflavone genistein and blueberry polyphenolic acids suggests diet-mediated targeting of cancer stem-like/progenitor cells. Carcinogenesis 2012, 33, 652–660. [Google Scholar] [CrossRef]
  120. Fan, P.; Fan, S.; Wang, H.; Mao, J.; Shi, Y.; Ibrahim, M.M.; Ma, W.; Yu, X.; Hou, Z.; Wang, B.; et al. Genistein decreases the breast cancer stem-like cell population through Hedgehog pathway. Stem Cell Res. Ther. 2013, 4, 146. [Google Scholar] [CrossRef] [Green Version]
  121. Montales, M.T.; Rahal, O.M.; Nakatani, H.; Matsuda, T.; Simmen, R.C. Repression of mammary adipogenesis by genistein limits mammosphere formation of human MCF-7 cells. J. Endocrinol. 2013, 218, 135–149. [Google Scholar] [CrossRef]
  122. Liu, Y.; Zou, T.; Wang, S.; Chen, H.; Su, D.; Fu, X.; Zhang, Q.; Kang, X. Genistein-induced differentiation of breast cancer stem/progenitor cells through a paracrine mechanism. Int. J. Oncol. 2016, 48, 1063–1072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Lauricella, M.; Carlisi, D.; Giuliano, M.; Calvaruso, G.; Cernigliaro, C.; Vento, R.; D’anneo, A. The analysis of estrogen receptor-α positive breast cancer stem-like cells unveils a high expression of the serpin proteinase inhibitor PI-9: Possible regulatory mechanisms. Int. J. Oncol. 2016, 49, 352–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Yuan, B.; Cheng, L.; Gupta, K.; Chiang, H.C.; Gupta, H.B.; Sareddy, G.R.; Wang, D.; Lathrop, K.; Elledge, R.; Wang, P.; et al. Tyrosine phosphorylation regulates ERβ ubiquitination, protein turnover, and inhibition of breast cancer. Oncotarget 2016, 7, 42585–42597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Taghizadeh, B.; Ghavami, L.; Nikoofar, A.; Goliaei, B. Equol as a potent radiosensitizer in estrogen receptor-positive and -negative human breast cancer cell lines. Breast Cancer 2013, 22, 382–390. [Google Scholar] [CrossRef] [PubMed]
  126. Magee, P.J.; Raschke, M.; Steiner, C.; Duffin, J.G.; Pool-Zobel, B.L.; Jokela, T.; Wahala, K.; Rowland, I.R. Equol: A Comparison of the Effects of the Racemic Compound with That of the Purified S-Enantiomer on the Growth, Invasion, and DNA Integrity of Breast and Prostate Cells In Vitro. Nutr. Cancer 2006, 54, 232–242. [Google Scholar] [CrossRef]
  127. Magee, P.; McGlynn, H.; Rowland, I. Differential effects of isoflavones and lignans on invasiveness of MDA-MB-231 breast cancer cells in vitro. Cancer Lett. 2004, 208, 35–41. [Google Scholar] [CrossRef]
  128. Kim, T.; Choi, E.J. Equol induced apoptosis via cell cycle arrest in human breast cancer MDA-MB-453 but not MCF-7 cells. Mol. Med. Rep. 2008, 1, 239–244. [Google Scholar] [CrossRef] [Green Version]
  129. Choi, E.J.; Ahn, W.S.; Bae, S.M. Equol induces apoptosis through cytochrome c-mediated caspases cascade in human breast cancer MDA-MB-453 cells. Chem. Interact. 2009, 177, 7–11. [Google Scholar] [CrossRef]
  130. Zhang, J.; Ren, L.; Yu, M.; Liu, X.; Ma, W.; Huang, L.; Li, X.; Ye, X. S-equol inhibits proliferation and promotes apoptosis of human breast cancer MCF-7 cells via regulating miR-10a-5p and PI3K/AKT pathway. Arch. Biochem. Biophys. 2019, 672, 108064. [Google Scholar] [CrossRef]
  131. Ju, Y.H.; Fultz, J.; Allred, K.F.; Doerge, D.R.; Helferich, W.G. Effects of dietary daidzein and its metabolite, equol, at physiological concentrations on the growth of estrogen-dependent human breast cancer (MCF-7) tumors implanted in ovariectomized athymic mice. Carcinogenesis 2006, 27, 856–863. [Google Scholar] [CrossRef]
  132. Onoda, A.; Ueno, T.; Uchiyama, S.; Hayashi, S.-I.; Kato, K.; Wake, N. Effects of S-equol and natural S-equol supplement (SE5-OH) on the growth of MCF-7 in vitro and as tumors implanted into ovariectomized athymic mice. Food Chem. Toxicol. 2011, 49, 2279–2284. [Google Scholar] [CrossRef] [PubMed]
  133. Tonetti, D.A.; Zhang, Y.; Zhao, H.; Lim, S.B.; Constantinou, A.I. The effect of the phytoestrogens genistein, daidzein, and equol on the growth of tamoxifen-resistant T47D/PKC alpha. Nutr Cancer 2007, 58, 222–229. [Google Scholar] [CrossRef] [PubMed]
  134. Martínez-Montemayor, M.M.; Otero-Franqui, E.; Martinez, J.; Mota-Peynado, A.D.L.; Cubano, L.S.; Dharmawardhane, S. Individual and combined soy isoflavones exert differential effects on metastatic cancer progression. Clin. Exp. Metastasis 2010, 27, 465–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. de la Parra, C.; Otero-Franqui, E.; Martinez-Montemayor, M.; Dharmawardhane, S. The soy isoflavone equol may increase cancer malignancy via up-regulation of eukaryotic protein synthesis initiation factor eIF4G. J. Biol. Chem. 2012, 287, 41640–41650. [Google Scholar] [CrossRef] [Green Version]
  136. de la Parra, C.; Otero-Franqui, E.; Martinez-Montemayor, M.; Dharmawardhane, S. Equol, an isoflavone metabolite, regulates cancer cell viability and protein synthesis initiation via c-Myc and eIF4G. J. Biol. Chem. 2015, 290, 6047–6057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Amin, I.; Majid, S.; Farooq, A.; Wani, H.A.; Noor, F.; Khan, R.; Shakeel, S.; Bhat, S.A.; Ahmad, A.; Madkhali, H.; et al. Chapter 8-Naringenin (4,5,7-trihydroxyflavanone) as a potent neuroprotective agent: From chemistry to medicine. In Studies in Natural Products Chemistry; Attaur, R., Ed.; Elsevier: Amsterdam, The Netherlands, 2020; pp. 271–300. [Google Scholar]
  138. Srinivasan, S.; Vinothkumar, V.; Murali, R. Chapter 22-Antidiabetic Efficacy of Citrus Fruits with Special Allusion to Flavone Glycosides. In Bioactive Food as Dietary Interventions for Diabetes, 2nd ed.; Watson, R.R., Preedy, V.R., Eds.; Academic Press: Cambridge, MA, USA, 2019; pp. 335–346. [Google Scholar]
  139. Singh, R.S.; Singh, T.; Pandey, A. Chapter 1-Microbial Enzymes—An Overview. In Advances in Enzyme Technology; Elsevier: Amsterdam, The Netherlands, 2019; pp. 1–40. [Google Scholar]
  140. Choudhury, R.; Chowrimootoo, G.; Srai, K.; Debnam, E.; Rice-Evans, C.A. Interactions of the flavonoid naringenin in the gastrointestinal tract and the influence of glycosylation. Biochem. Biophys. Res. Commun. 1999, 265, 410–415. [Google Scholar] [CrossRef]
  141. Felgines, C.; Texier, O.; Morand, C.; Manach, C.; Scalbert, A.; Régerat, F.; Rémésy, C. Bioavailability of the flavanone naringenin and its glycosides in rats. Am. J. Physiol. Liver Physiol. 2000, 279, G1148–G1154. [Google Scholar] [CrossRef]
  142. Memariani, Z.; Abbas, S.Q.; ul Hassan, S.S.; Ahmadi, A.; Chabra, A. Naringin and naringenin as anticancer agents and adjuvants in cancer combination therapy: Efficacy and molecular mechanisms of action, a comprehensive narrative review. Pharmacol. Res. 2021, 171, 105264. [Google Scholar] [CrossRef]
  143. Helle, J.; Kräker, K.; Bader, M.I.; Keiler, A.M.; Zierau, O.; Vollmer, G.; Welsh, J.; Kretzschmar, G. Assessment of the proliferative capacity of the flavanones 8-prenylnaringenin, 6-(1.1-dimethylallyl)naringenin and naringenin in MCF-7 cells and the rat mammary gland. Mol. Cell. Endocrinol. 2014, 392, 125–135. [Google Scholar] [CrossRef]
  144. Guthrie, N.; Carroll, K.K. Inhibition of Mammary Cancer by Citrus Flavonoids. Flavonoids Living Syst. 1998, 439, 227–236. [Google Scholar] [CrossRef]
  145. Zhang, F.; Dong, W.; Zeng, W.; Zhang, L.; Zhang, C.; Qiu, Y.; Wang, L.; Yin, X.; Zhang, C.; Liang, W. Naringenin prevents TGF-β1 secretion from breast cancer and suppresses pulmonary metastasis by inhibiting PKC activation. Breast Cancer Res. 2016, 18, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Harmon, A.W.; Patel, Y.M. Naringenin Inhibits Glucose Uptake in MCF-7 Breast Cancer Cells: A Mechanism for Impaired Cellular Proliferation. Breast Cancer Res. Treat. 2004, 85, 103–110. [Google Scholar] [CrossRef] [PubMed]
  147. Sun, Y.; Gu, J. Study on effect of naringenin in inhibiting migration and invasion of breast cancer cells and its molecular mechanism. China J. Chin. Mater. Med. 2015, 40, 1144–1150. [Google Scholar]
  148. Wang, R.; Wang, J.; Dong, T.; Shen, J.; Gao, X.; Zhou, J. Naringenin has a chemoprotective effect in MDA-MB-231 breast cancer cells via inhibition of caspase-3 and -9 activities. Oncol. Lett. 2019, 17, 1217–1222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Zhao, Z.; Jin, G.; Ge, Y.; Guo, Z. Naringenin inhibits migration of breast cancer cells via inflammatory and apoptosis cell signaling pathways. Inflammopharmacology 2019, 27, 1021–1036. [Google Scholar] [CrossRef]
  150. Qi, Z.; Kong, S.; Zhao, S.; Tang, Q. Naringenin inhibits human breast cancer cells (MDA-MB-231) by inducing programmed cell death, caspase stimulation, G2/M phase cell cycle arrest and suppresses cancer metastasis. Cell. Mol. Biol. 2021, 67, 8–13. [Google Scholar] [CrossRef] [PubMed]
  151. Rajamani, S.; Radhakrishnan, A.; Sengodan, T.; Thangavelu, S. Augmented anticancer activity of naringenin-loaded TPGS polymeric nanosuspension for drug resistive MCF-7 human breast cancer cells. Drug Dev. Ind. Pharm. 2018, 44, 1752–1761. [Google Scholar] [CrossRef]
  152. 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]
  153. Qin, L.; Jin, L.; Lu, L.; Lu, X.; Zhang, C.; Zhang, F.; Liang, W. Naringenin reduces lung metastasis in a breast cancer resection model. Protein Cell 2011, 2, 507–516. [Google Scholar] [CrossRef]
  154. Hatkevich, T.; Ramos, J.; Santos-Sanchez, I.; Patel, Y.M. A naringenin–tamoxifen combination impairs cell proliferation and survival of MCF-7 breast cancer cells. Exp. Cell Res. 2014, 327, 331–339. [Google Scholar] [CrossRef]
  155. Peiffer, D.S.; Ma, E.; Wyatt, D.; Albain, K.S.; Osipo, C. DAXX-inducing phytoestrogens inhibit ER+ tumor initiating cells and delay tumor development. NPJ Breast Cancer 2020, 6, 1–12. [Google Scholar] [CrossRef] [PubMed]
  156. Zhang, Y.-Z.; Yang, J.-Y.; Wu, R.-X.; Fang, C.; Lu, H.; Li, H.-C.; Li, D.-M.; Zuo, H.-L.; Ren, L.-P.; Liu, X.-Y.; et al. Network Pharmacology–Based Identification of Key Mechanisms of Xihuang Pill in the Treatment of Triple-Negative Breast Cancer Stem Cells. Front. Pharmacol. 2021, 12, 714628. [Google Scholar] [CrossRef] [PubMed]
  157. Shen, T.; Wang, X.-N.; Lou, H.-X. Natural stilbenes: An overview. Nat. Prod. Rep. 2009, 26, 916–935. [Google Scholar] [CrossRef] [PubMed]
  158. Langcake, P. Disease resistance of Vitis spp. and the production of the stress metabolites resveratrol, ε-viniferin, α-viniferin and pterostilbene. Physiol. Plant Pathol. 1981, 18, 213–226. [Google Scholar] [CrossRef]
  159. Langcake, P.; Pryce, R.J. The production of resveratrol by Vitis vinifera and other members of the Vitaceae as a response to infection or injury. Physiol. Plant Pathol. 1976, 9, 77–86. [Google Scholar] [CrossRef]
  160. Soleas, G.J.; Diamandis, E.P.; Goldberg, D.M. Resveratrol: A molecule whose time has come? And gone? Clin. Biochem. 1997, 30, 91–113. [Google Scholar] [CrossRef]
  161. Siemann, E.; Creasy, L. Concentration of the phytoalexin resveratrol in wine. Am. J. Enol. Vitic. 1992, 43, 49–52. [Google Scholar]
  162. Basly, J.-P.; Marre-Fournier, F.; Le Bail, J.-C.; Habrioux, G.; Chulia, A.J. Estrogenic/antiestrogenic and scavenging properties of (E)- and (Z)-resveratrol. Life Sci. 2000, 66, 769–777. [Google Scholar] [CrossRef]
  163. Lee, P.-S.; Chiou, Y.-S.; Ho, C.-T.; Pan, M.-H. Chemoprevention by resveratrol and pterostilbene: Targeting on epigenetic regulation. BioFactors 2017, 44, 26–35. [Google Scholar] [CrossRef]
  164. Langcake, P.; Cornford, C.; Pryce, R. Identification of pterostilbene as a phytoalexin from Vitis vinifera leaves. Phytochemistry 1979, 18, 1025–1027. [Google Scholar] [CrossRef]
  165. Zhang, L.; Wen, X.; Li, M.; Li, S.; Zhao, H. Targeting cancer stem cells and signaling pathways by resveratrol and pterostilbene. BioFactors 2017, 44, 61–68. [Google Scholar] [CrossRef] [PubMed]
  166. Lin, H.-S.; Yue, B.-D.; Ho, P.C. Determination of pterostilbene in rat plasma by a simple HPLC-UV method and its application in pre-clinical pharmacokinetic study. Biomed. Chromatogr. 2009, 23, 1308–1315. [Google Scholar] [CrossRef] [PubMed]
  167. Bowers, J.L.; Tyulmenkov, V.V.; Jernigan, S.C.; Klinge, C.M. Resveratrol acts as a mixed agonist/antagonist for estrogen receptors alpha and beta. Endocrinology 2000, 141, 3657–3667. [Google Scholar] [CrossRef] [PubMed]
  168. Bhat, K.P.; Lantvit, D.; Christov, K.; Mehta, R.G.; Moon, R.C.; Pezzuto, J.M. Estrogenic and antiestrogenic properties of resveratrol in mammary tumor models. Cancer Res. 2001, 61, 7456–7463. [Google Scholar]
  169. Lu, R.; Serrero, G. Resveratrol, a natural product derived from grape, exhibits antiestrogenic activity and inhibits the growth of human breast cancer cells. J. Cell. Physiol. 1999, 179, 297–304. [Google Scholar] [CrossRef]
  170. Pan, C.; Hu, Y.; Li, J.; Wang, Z.; Huang, J.; Zhang, S.; Ding, L. Estrogen Receptor-α36 Is Involved in Pterostilbene-Induced Apoptosis and Anti-Proliferation in In Vitro and In Vivo Breast Cancer. PLoS ONE 2014, 9, e104459. [Google Scholar] [CrossRef] [Green Version]
  171. Tolba, M.F.; Abdel-Rahman, S.Z. Pterostilbine, an active component of blueberries, sensitizes colon cancer cells to 5-fluorouracil cytotoxicity. Sci. Rep. 2015, 5, 15239. [Google Scholar] [CrossRef] [Green Version]
  172. Robb, E.L.; Stuart, J.A. The stilbenes resveratrol, pterostilbene and piceid affect growth and stress resistance in mammalian cells via a mechanism requiring estrogen receptor beta and the induction of Mn-superoxide dismutase. Phytochemistry 2014, 98, 164–173. [Google Scholar] [CrossRef]
  173. Song, Z.; Han, S.; Pan, X.; Gong, Y.; Wang, M. Pterostilbene mediates neuroprotection against oxidative toxicity via oestrogen receptor α signalling pathways. J. Pharm. Pharmacol. 2015, 67, 720–730. [Google Scholar] [CrossRef]
  174. Buckland, G.; Travier, N.; Cottet, V.; González, C.A.; Lujan-Barroso, L.; Agudo, A.; Trichopoulou, A.; Lagiou, P.; Trichopoulos, D.; Peeters, P.; et al. Adherence to the mediterranean diet and risk of breast cancer in the European prospective investigation into cancer and nutrition cohort study. Int. J. Cancer 2012, 132, 2918–2927. [Google Scholar] [CrossRef]
  175. Fung, T.T.; Hu, F.B.; McCullough, M.L.; Newby, P.K.; Willett, W.C.; Holmes, M.D. Diet Quality Is Associated with the Risk of Estrogen Receptor–Negative Breast Cancer in Postmenopausal Women. J. Nutr. 2006, 136, 466–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Trichopoulou, A.; Bamia, C.; Lagiou, P.; Trichopoulos, D. Conformity to traditional Mediterranean diet and breast cancer risk in the Greek EPIC (European Prospective Investigation into Cancer and Nutrition) cohort. Am. J. Clin. Nutr. 2010, 92, 620–625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Murtaugh, M.A.; Sweeney, C.; Giuliano, A.R.; Herrick, J.S.; Hines, L.; Byers, T.; Baumgartner, K.B.; Slattery, M.L. Diet patterns and breast cancer risk in Hispanic and non-Hispanic white women: The Four-Corners Breast Cancer Study. Am. J. Clin. Nutr. 2008, 87, 978–984. [Google Scholar] [CrossRef] [Green Version]
  178. Grosso, G.; Buscemi, S.; Galvano, F.; Mistretta, A.; Marventano, S.; La Vela, V.; Drago, F.; Gangi, S.; Basile, F.; Biondi, A. Mediterranean diet and cancer: Epidemiological evidence and mechanism of selected aspects. BMC Surg. 2013, 13, S14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Santos-Buelga, C.; González-Manzano, S.; González-Paramás, A.M. Wine, Polyphenols, and Mediterranean Diets. What Else Is There to Say? Molecules 2021, 26, 5537. [Google Scholar] [CrossRef] [PubMed]
  180. Renaud, S.; De Lorgeril, M. Wine, alcohol, platelets, and the French paradox for coronary heart disease. Lancet 1992, 339, 1523–1526. [Google Scholar] [CrossRef]
  181. Catalgol, B.; Batirel, S.; Taga, Y.; Ozer, N.K. Resveratrol: French Paradox Revisited. Front. Pharmacol. 2012, 3, 141. [Google Scholar] [CrossRef] [Green Version]
  182. Levi, F.; Pasche, C.; Lucchini, F.; Ghidoni, R.; Ferraroni, M.; La Vecchia, C. Resveratrol and breast cancer risk. Eur. J. Cancer Prev. 2005, 14, 139–142. [Google Scholar] [CrossRef]
  183. Semba, R.D.; Ferrucci, L.; Bartali, B.; Urpi, M.; Zamora-Ros, R.; Sun, K.; Cherubini, A.; Bandinelli, S.; Andres-Lacueva, C. Resveratrol Levels and All-Cause Mortality in Older Community-Dwelling Adults. JAMA Intern. Med. 2014, 174, 1077–1084. [Google Scholar] [CrossRef] [Green Version]
  184. Poschner, S.; Maier-Salamon, A.; Thalhammer, T.; Jäger, W. Resveratrol and other dietary polyphenols are inhibitors of estrogen metabolism in human breast cancer cells. J. Steroid Biochem. Mol. Biol. 2019, 190, 11–18. [Google Scholar] [CrossRef]
  185. Gehm, B.D.; McAndrews, J.M.; Chien, P.-Y.; Jameson, J.L. Resveratrol, a polyphenolic compound found in grapes and wine, is an agonist for the estrogen receptor. Proc. Natl. Acad. Sci. USA 1997, 94, 14138–14143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Nakagawa, H.; Kiyozuka, Y.; Uemura, Y.; Senzaki, H.; Shikata, N.; Hioki, K.; Tsubura, A. Resveratrol inhibits human breast cancer cell growth and may mitigate the effect of linoleic acid, a potent breast cancer cell stimulator. J. Cancer Res. Clin. Oncol. 2001, 127, 258–264. [Google Scholar] [CrossRef] [PubMed]
  187. Chen, F.-P.; Chien, M.-H.; Chern, I.Y.-Y. Impact of lower concentrations of phytoestrogens on the effects of estradiol in breast cancer cells. Climacteric 2015, 18, 1–8. [Google Scholar] [CrossRef] [PubMed]
  188. Poschner, S.; Maier-Salamon, A.; Zehl, M.; Wackerlig, J.; Dobusch, D.; Meshcheryakova, A.; Mechtcheriakova, D.; Thalhammer, T.; Pachmann, B.; Jäger, W. Resveratrol Inhibits Key Steps of Steroid Metabolism in a Human Estrogen-Receptor Positive Breast Cancer Model: Impact on Cellular Proliferation. Front. Pharmacol. 2018, 9, 742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Murias, M.; Miksits, M.; Aust, S.; Spatzenegger, M.; Thalhammer, T.; Szekeres, T.; Jaeger, W. Metabolism of resveratrol in breast cancer cell lines: Impact of sulfotransferase 1A1 expression on cell growth inhibition. Cancer Lett. 2008, 261, 172–182. [Google Scholar] [CrossRef]
  190. Nguyen, T.H.; Mustafa, F.B.; Pervaiz, S.; Ng, F.S.; Lim, L.H. ERK1/2 activation is required for resveratrol-induced apoptosis in MDA-MB-231 cells. Int. J. Oncol. 2008, 33, 81–92. [Google Scholar] [CrossRef]
  191. Chin, Y.-T.; Hsieh, M.-T.; Yang, S.-H.; Tsai, P.-W.; Wang, S.-H.; Wang, C.-C.; Lee, Y.-S.; Cheng, G.-Y.; Huangfu, W.-C.; London, D.; et al. Anti-proliferative and gene expression actions of resveratrol in breast cancer cells in vitro. Oncotarget 2014, 5, 12891–12907. [Google Scholar] [CrossRef] [Green Version]
  192. Castillo-Pichardo, L.; Martinez-Montemayor, M.; Martínez, J.E.; Wall, K.M.; Cubano, L.A.; Dharmawardhane, S. Inhibition of mammary tumor growth and metastases to bone and liver by dietary grape polyphenols. Clin. Exp. Metastasis 2009, 26, 505–516. [Google Scholar] [CrossRef]
  193. Suh, J.; Kim, D.-H.; Surh, Y.-J. Resveratrol suppresses migration, invasion and stemness of human breast cancer cells by interfering with tumor-stromal cross-talk. Arch. Biochem. Biophys. 2018, 643, 62–71. [Google Scholar] [CrossRef]
  194. Banerjee, S.; Bueso-Ramos, C.; Aggarwal, B.B. Suppression of 7, 12-dimethylbenz (a) anthracene-induced mammary carcinogenesis in rats by resveratrol: Role of nuclear factor-κB, cyclooxygenase 2, and matrix metalloprotease 9. Cancer Res. 2002, 62, 4945. [Google Scholar]
  195. Provinciali, M.; Re, F.; Donnini, A.; Orlando, F.; Bartozzi, B.; Di Stasio, G.; Smorlesi, A. Effect of resveratrol on the development of spontaneous mammary tumors in HER-2/neu transgenic mice. Int. J. Cancer 2005, 115, 36–45. [Google Scholar] [CrossRef] [PubMed]
  196. Garvin, S.; Öllinger, K.; Dabrosin, C. Resveratrol induces apoptosis and inhibits angiogenesis in human breast cancer xenografts in vivo. Cancer Lett. 2006, 231, 113–122. [Google Scholar] [CrossRef] [PubMed]
  197. Whitsett, T.; Carpenter, M.; Lamartiniere, C.A. Resveratrol, but not EGCG, in the diet suppresses DMBA-induced mammary cancer in rats. J. Carcinog. 2006, 5, 15. [Google Scholar] [CrossRef]
  198. Chatterjee, M.; Das, S.; Janarthan, M.; Ramachandran, H.K.; Chatterjee, M. Role of 5-lipoxygenase in resveratrol mediated suppression of 7, 12-dimethylbenz (α) anthracene-induced mammary carcinogenesis in rats. Eur. J. Pharmacol. 2011, 668, 99–106. [Google Scholar] [CrossRef] [PubMed]
  199. Fu, Y.; Chang, H.; Peng, X.; Bai, Q.; Yi, L.; Zhou, Y.; Zhu, J.; Mi, M. Resveratrol Inhibits Breast Cancer Stem-Like Cells and Induces Autophagy via Suppressing Wnt/β-Catenin Signaling Pathway. PLoS ONE 2014, 9, e102535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Pandey, P.R.; Okuda, H.; Watabe, M.; Pai, S.K.; Liu, W.; Kobayashi, A.; Xing, F.; Fukuda, K.; Hirota, S.; Sugai, T.; et al. Resveratrol suppresses growth of cancer stem-like cells by inhibiting fatty acid synthase. Breast Cancer Res. Treat. 2010, 130, 387–398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Singh, B.; Shoulson, R.; Chatterjee, A.; Ronghe, A.; Bhat, N.K.; Dim, D.C.; Bhat, H.K. Resveratrol inhibits estrogen-induced breast carcinogenesis through induction of NRF2-mediated protective pathways. Carcinogenesis 2014, 35, 1872–1880. [Google Scholar] [CrossRef] [Green Version]
  202. Hagiwara, K.; Kosaka, N.; Yoshioka, Y.; Takahashi, R.-U.; Takeshita, F.; Ochiya, T. Stilbene derivatives promote Ago2-dependent tumour-suppressive microRNA activity. Sci. Rep. 2012, 2, 314. [Google Scholar] [CrossRef] [Green Version]
  203. Rimando, A.M.; Cuendet, M.; Desmarchelier, C.; Mehta, R.G.; Pezzuto, J.M.; Duke, S.O. Cancer Chemopreventive and Antioxidant Activities of Pterostilbene, a Naturally Occurring Analogue of Resveratrol. J. Agric. Food Chem. 2002, 50, 3453–3457. [Google Scholar] [CrossRef]
  204. Pan, M.H.; Lin, Y.T.; Lin, C.L.; Wei, C.S.; Ho, C.T.; Chen, W.J. Suppression of Heregulin-β1/HER2-Modulated Invasive and Aggressive Phenotype of Breast Carcinoma by Pterostilbene via Inhibition of Matrix Metalloproteinase-9, p38 Kinase Cascade and Akt Activation. Evid.-Based Complement. Altern. Med. 2011, 2011, 562187. [Google Scholar] [CrossRef] [Green Version]
  205. Mak, K.K.; Wu, A.T.; Lee, W.H.; Chang, T.C.; Chiou, J.F.; Wang, L.S.; Wu, C.H.; Huang, C.Y.F.; Shieh, Y.S.; Chao, T.Y.; et al. Pterostilbene, a bioactive component of blueberries, suppresses the generation of breast cancer stem cells within tumor microenvironment and metastasis via modulating NF-κB/microRNA 448 circuit. Mol. Nutr. Food Res. 2013, 57, 1123–1134. [Google Scholar] [CrossRef] [PubMed]
  206. Wu, C.-H.; Hong, B.-H.; Ho, C.-T.; Yen, G.-C. Targeting Cancer Stem Cells in Breast Cancer: Potential Anticancer Properties of 6-Shogaol and Pterostilbene. J. Agric. Food Chem. 2015, 63, 2432–2441. [Google Scholar] [CrossRef] [PubMed]
  207. Adlercreutz, H. Phyto-oestrogens and cancer. Lancet Oncol. 2002, 3, 364–373. [Google Scholar] [CrossRef]
  208. Lathrop, K.I.; Kaklamani, V.G.; Brenner, A.J.; Li, R.; Nazarullah, A.; Hackman, S.; Thomas, C.; Gelfond, J.; Rodriguez, M.; Elledge, R. Novel estrogen receptor beta agonist S-equol decreases tumor proliferation in patients with triple negative breast cancer (TNBC). J. Clin. Oncol. 2020, 38, 560. [Google Scholar] [CrossRef]
  209. Jaskulski, S.; Jung, A.Y.; Rudolph, A.; Johnson, T.; Thöne, K.; Herpel, E.; Sinn, P.; Chang-Claude, J. Genistein and enterolactone in relation to Ki-67 expression and HER2 status in postmenopausal breast cancer patients. Mol. Nutr. Food Res. 2017, 61, 1700449. [Google Scholar] [CrossRef] [PubMed]
  210. Teas, J.; Hurley, T.G.; Hebert, J.R.; Franke, A.A.; Sepkovic, D.W.; Kurzer, M.S. Dietary Seaweed Modifies Estrogen and Phytoestrogen Metabolism in Healthy Postmenopausal Women. J. Nutr. 2009, 139, 939–944. [Google Scholar] [CrossRef] [Green Version]
  211. Nayeem, F.; Chen, N.-W.; Nagamani, M.; Anderson, K.E.; Lu, L.-J.W. Daidzein and genistein have differential effects in decreasing whole body bone mineral density but had no effect on hip and spine density in premenopausal women: A 2-year randomized, double-blind, placebo-controlled study. Nutr. Res. 2019, 68, 70–81. [Google Scholar] [CrossRef]
  212. Lu, L.-J.W.; Chen, N.-W.; Nayeem, F.; Nagamani, M.; Anderson, K.E. Soy isoflavones interact with calcium and contribute to blood pressure homeostasis in women: A randomized, double-blind, placebo controlled trial. Eur. J. Nutr. 2020, 59, 2369–2381. [Google Scholar] [CrossRef]
  213. Khan, S.A.; Chatterton, R.T.; Michel, N.; Bryk, M.; Lee, O.; Ivancic, D.; Heinz, R.; Zalles, C.M.; Helenowski, I.B.; Jovanovic, B.D.; et al. Soy Isoflavone Supplementation for Breast Cancer Risk Reduction: A Randomized Phase II Trial. Cancer Prev. Res. 2012, 5, 309–319. [Google Scholar] [CrossRef] [Green Version]
  214. Shike, M.; Doane, A.S.; Russo, L.; Cabal, R.; Reis-Filo, J.; Gerald, W.; Cody, H.; Khanin, R.; Bromberg, J.; Norton, L. The Effects of Soy Supplementation on Gene Expression in Breast Cancer: A Randomized Placebo-Controlled Study. JNCI J. Natl. Cancer Inst. 2014, 106, 189. [Google Scholar] [CrossRef] [Green Version]
  215. Wu, A.H.; Spicer, D.V.; Garcia, A.A.; Tseng, C.-C.; Hovanessian-Larsen, L.; Sheth, P.; Martin, S.E.; Hawes, D.; Russell, C.; Macdonald, H.; et al. Double-Blind Randomized 12-Month Soy Intervention Had No Effects on Breast MRI Fibroglandular Tissue Density or Mammographic Density. Cancer Prev. Res. 2015, 8, 942–951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Delmanto, A.; Nahas-Neto, J.; Traiman, P.; Uemura, G.; Pessoa, E.C.; Nahas, E.A.P. Effects of soy isoflavones on mammographic density and breast parenchyma in postmenopausal women: A randomized, double-blind, placebo-controlled clinical trial. Menopause 2013, 20, 1049–1054. [Google Scholar] [CrossRef] [PubMed]
  217. Pop, E.A.; Fischer, L.M.; Coan, A.D.; Gitzinger, M.; Nakamura, J.; Zeisel, S.H. Effects of a high daily dose of soy isoflavones on DNA damage, apoptosis, and estrogenic outcomes in healthy postmenopausal women: A phase I clinical trial. Menopause 2008, 15, 684–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Setchell, K.D.R.; 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] [Green Version]
  219. Bolca, S.; Urpi-Sarda, M.; Blondeel, P.; Roche, N.; Vanhaecke, L.; Possemiers, S.; Al-Maharik, N.; Botting, N.; De Keukeleire, D.; Bracke, M.; et al. Disposition of soy isoflavones in normal human breast tissue. Am. J. Clin. Nutr. 2010, 91, 976–984. [Google Scholar] [CrossRef] [Green Version]
  220. Powles, T.J.; Howell, A.; Evans, G.; McCloskey, E.V.; Ashley, S.; Greenhalgh, R.; Affen, J.; Flook, L.A.; Tidy, A. Red clover isoflavones are safe and well tolerated in women with a family history of breast cancer. Menopause Int. 2008, 14, 6–12. [Google Scholar] [CrossRef]
  221. Fabian, C.J.; Khan, S.A.; Garber, J.E.; Dooley, W.C.; Yee, L.D.; Klemp, J.R.; Nydegger, J.L.; Powers, K.R.; Kreutzjans, A.L.; Zalles, C.M.; et al. Randomized Phase IIB Trial of the Lignan Secoisolariciresinol Diglucoside in Premenopausal Women at Increased Risk for Development of Breast Cancer. Cancer Prev. Res. 2020, 13, 623–634. [Google Scholar] [CrossRef] [Green Version]
  222. Fabian, C.J.; Kimler, B.F.; Zalles, C.M.; Klemp, J.R.; Petroff, B.K.; Khan, Q.J.; Sharma, P.; Setchell, K.D.R.; Zhao, X.; Phillips, T.A.; et al. Reduction in Ki-67 in Benign Breast Tissue of High-Risk Women with the Lignan Secoisolariciresinol Diglycoside. Cancer Prev. Res. 2010, 3, 1342–1350. [Google Scholar] [CrossRef] [Green Version]
  223. McCann, S.E.; Edge, S.B.; Hicks, D.G.; Thompson, L.U.; Morrison, C.D.; Fetterly, G.; Andrews, C.; Clark, K.; Wilton, J.; Kulkarni, S. A pilot study comparing the effect of flaxseed, aromatase inhibitor, and the combination on breast tumor biomarkers. Nutr. Cancer 2014, 66, 566–575. [Google Scholar] [CrossRef] [Green Version]
  224. Velentzis, L.S.; Keshtgar, M.R.; Woodside, J.; Leathem, A.J.; Titcomb, A.; Perkins, K.A.; Mazurowska, M.; Anderson, V.; Wardell, K.; Cantwell, M.M. Significant changes in dietary intake and supplement use after breast cancer diagnosis in a UK multicentre study. Breast Cancer Res. Treat. 2011, 128, 473–482. [Google Scholar] [CrossRef] [Green Version]
  225. Jaskulski, S.; Jung, A.Y.; Behrens, S.; Johnson, T.; Kaaks, R.; Thöne, K.; Flesch-Janys, D.; Sookthai, D.; Chang-Claude, J. Circulating enterolactone concentrations and prognosis of postmenopausal breast cancer: Assessment of mediation by inflammatory markers. Int. J. Cancer 2018, 143, 2698–2708. [Google Scholar] [CrossRef] [PubMed]
  226. Jaskulski, S.; Jung, A.Y.; Huebner, M.; Poschet, G.; Hell, R.; Hüsing, A.; Gonzalez-Maldonado, S.; Behrens, S.; Obi, N.; Becher, H.; et al. Prognostic associations of circulating phytoestrogens and biomarker changes in long-term survivors of postmenopausal breast cancer. Nutr. Cancer 2019, 72, 1155–1169. [Google Scholar] [CrossRef] [PubMed]
  227. Swann, R.; Perkins, K.A.; Velentzis, L.S.; Ciria, C.; Dutton, S.J.; Mulligan, A.A.; Woodside, J.V.; Cantwell, M.M.; Leathem, A.J.; Robertson, C.E.; et al. The DietCompLyf study: A prospective cohort study of breast cancer survival and phytoestrogen consumption. Maturitas 2013, 75, 232–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Structure and Function of Phytoestrogens on Breast Cancer Stem Cells. (A). Chemical structures of different classes of phytoestrogens compared to 17β-estradiol. Colors highlight different subclasses of phytoestrogens. (B). Phytoestrogens either inhibit or activate different signaling pathways to suppress breast cancer stem cells (BSCS). Specifically, inhibition of hedgehog, CD44 or adipogenesis by genistein, pterostilbene or resveratrol reduces survival of BCSC. Activation of ERβ, DAXX, FAS, argonaute2 or miRNAs by S-equol, genistein, naringenin, resveratrol or pterostilbene suppresses survival of BCSC. Red indicates inhibition of pathways. Blue indicates activation of pathways.
Figure 1. Structure and Function of Phytoestrogens on Breast Cancer Stem Cells. (A). Chemical structures of different classes of phytoestrogens compared to 17β-estradiol. Colors highlight different subclasses of phytoestrogens. (B). Phytoestrogens either inhibit or activate different signaling pathways to suppress breast cancer stem cells (BSCS). Specifically, inhibition of hedgehog, CD44 or adipogenesis by genistein, pterostilbene or resveratrol reduces survival of BCSC. Activation of ERβ, DAXX, FAS, argonaute2 or miRNAs by S-equol, genistein, naringenin, resveratrol or pterostilbene suppresses survival of BCSC. Red indicates inhibition of pathways. Blue indicates activation of pathways.
Ijms 23 06813 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nguyen, M.; Osipo, C. Targeting Breast Cancer Stem Cells Using Naturally Occurring Phytoestrogens. Int. J. Mol. Sci. 2022, 23, 6813. https://doi.org/10.3390/ijms23126813

AMA Style

Nguyen M, Osipo C. Targeting Breast Cancer Stem Cells Using Naturally Occurring Phytoestrogens. International Journal of Molecular Sciences. 2022; 23(12):6813. https://doi.org/10.3390/ijms23126813

Chicago/Turabian Style

Nguyen, Mai, and Clodia Osipo. 2022. "Targeting Breast Cancer Stem Cells Using Naturally Occurring Phytoestrogens" International Journal of Molecular Sciences 23, no. 12: 6813. https://doi.org/10.3390/ijms23126813

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