This article was downloaded by: [New York University] On: 25 May 2015, At: 06:06 Publisher: Routledge Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH, UK

Nutrition and Cancer Publication details, including instructions for authors and subscription information: http://www.tandfonline.com/loi/hnuc20

Anticarcinogenic Effects of Dietary Phytoestrogens and Their Chemopreventive Mechanisms a

a

Kyung-A Hwang & Kyung-Chul Choi a

Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea Published online: 21 May 2015.

Click for updates To cite this article: Kyung-A Hwang & Kyung-Chul Choi (2015): Anticarcinogenic Effects of Dietary Phytoestrogens and Their Chemopreventive Mechanisms, Nutrition and Cancer, DOI: 10.1080/01635581.2015.1040516 To link to this article: http://dx.doi.org/10.1080/01635581.2015.1040516

PLEASE SCROLL DOWN FOR ARTICLE Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and should be independently verified with primary sources of information. Taylor and Francis shall not be liable for any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of the Content. This article may be used for research, teaching, and private study purposes. Any substantial or systematic reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http:// www.tandfonline.com/page/terms-and-conditions

Nutrition and Cancer, 0(0), 1–8 Copyright Ó 2015, Taylor & Francis Group, LLC ISSN: 0163-5581 print / 1532-7914 online DOI: 10.1080/01635581.2015.1040516

Anticarcinogenic Effects of Dietary Phytoestrogens and Their Chemopreventive Mechanisms Kyung-A Hwang and Kyung-Chul Choi

Downloaded by [New York University] at 06:06 25 May 2015

Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea

Phytoestrogens are phenolic compounds derived from plants and exert an estrogenic as well as an antiestrogenic effect and also various biological efficacies. Chemopreventive properties of phytoestrogens has emerged from epidemiological observations indicating that the incidence of some cancers including breast and prostate cancers is much lower in Asian people, who consume significantly higher amounts of phytoestrogens than Western people. There are 4 main classes of phytoestrogens: isoflavones, stilbenes, coumestans, and lignans. Currently, resveratrol is recognized as another major phytoestrogen present in grape and red wine and has been studied in many biological studies. Phytoestrogens have biologically diverse profitabilities and advantages such as low cytotoxicity to patients, lack of side effects in clinical trials, and pronounced benefits in a combined therapy. In this review, we highlighted the effects of genistein, daidzein, and resveratrol in relation with their anticarcinogenic activity. A lot of in vitro and in vivo results on their chemopreventive properties were presented along with the underlying mechanisms. Besides well-known mechanisms such as antioxidant property and apoptosis, newly elucidated anticarcinogenic modes of action including epigenetic modifications and topoisomerase inhibition have been provided to examine the possibility of phytoestrogens as promising reagents for cancer chemoprevention and/or treatment and to suggest the importance of plant-based diet of phytoestrogens.

INTRODUCTION Phytoestrogens are a class of biologically active phenolic compounds derived from plants and have structures that are similar to the principal mammalian estrogen, 17b-estradiol (E2) (1). They exert an estrogenic as well as an antiestrogenic effect and also various biological efficacies such as antioxidant property and chemoprevention against cancers. Especially, the importance of phytoestrogens in chemopreventive properties Submitted 17 February 2014; accepted in final form 9 April 2015. Address correspondence to Kyung-Chul Choi, Laboratory of Veterinary Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea. Phone: +82-43-261-3664. Fax: +82-43-267-3150. E-mail: [email protected]

has emerged from epidemiological observations, indicating that the incidence of some cancers including breast and prostate cancers is much lower in Asian people, who consume significantly higher amounts of phytoestrogens than Westerners (2). According to the reports, Asian women have a threefold lower breast cancer risk than women in the United States (3). Also, the incidence and mortality of prostate cancer shows the highest rates in Western countries and the lowest rates in Asian countries (4). With respect to the diet of phytoestrogens, there are considerable differences between the amounts of phytoestrogen intake of people in 2 parts of area. The average daily intake of phytoestrogens of Asian population is estimated to be between 20 and 50 mg. In contrast, the average daily consumption of phytoestrogens in the United States and Europe is measured to be 0.15~3 mg and 0.49~1 mg, respectively (1,5). These results are thought to be related with traditional dietary habits; plant-based diet of Asian population and meat-based diet of Western population. In fact, phytoestrogens are the most prevalent compounds found in ordinary edible plants such as vegetables, fruits, legumes, and teas (6). Therefore, they are commonly ingested in Asian populations from daily diets without going through special process, which is one of the most profitable aspects in the availability of phytoestrogens. There are 4 main classes of phytoestrogens: isoflavones, stilbenes, coumestans, and lignans (1,6). Among these classes, isoflavones and lignans are more intensely studied than the other 2 classes (7). Isoflavones are mainly present in legumes such as soybean, kala channa, mung bean, red lentils, and red clover. Genistein and daidzein are the 2 major isoflavones found in soy beans (7). The dietary sources of lignans are flaxseed, sea weed, whole grains, oil seeds, fruits, and vegetables. Secoisolariciresinol diglucoside and matairesinol are the 2 major lignans (7). Besides, resveratrol is currently recognized as a major phytoestrogen present in grape and red wine and has been intensely studied for its effectiveness. In this review, we highlight the effects and underlying mechanisms of important phytoestrogens such as genistein, daidzein, and resveratrol in relation with their anticarcinogenic activity, because these 2 1

Downloaded by [New York University] at 06:06 25 May 2015

2

K-A. HWANG AND K-C. CHOI

isoflavones and resveratrol are the most actively studied phytoestrogens in this field (8). Low cytotoxicity to patients, lack of side effects in clinical trials, and pronounced benefits in combined therapy with other treatments have activated the research for anticarcinogenic effects of phytoestrogens (7). Especially, the role of phytoestrogens in combined therapy was magnified in many other studies due to their adjuvant interactions with hormonal therapies using selective estrogen receptor modulators and aromatase inhibitors (9–13). For example, equol enhanced the anticancer activity of tamoxifen, an antagonist of estrogen receptor (ER), in estrogen dependent MCF-7 breast cancer cells, and a diet supplemented with soybean seeds synergistically increased tamoxifen’s suppressive effect in rat mammary tumor models (10,13). In addition, in the clinical trials on women with breast cancer, soy food consumption along with tamoxifen use was significantly associated with the decreased risk of death and recurrence (14), and high dietary intake of soy isoflavones was also correlated with lower risk of recurrence in those receiving anastrozole as endocrine therapy (15). However, these secondary preventive effects of phytoestrogens in combined therapies were not included in this review for illuminating the sole effect of each phytoestrogen. Overall, this review is aimed to examine the possiblility of genistein, daidzein, and resveratrol as natural food derived reagents for cancer chemoprevention and/or treatment by providing diverse anticancer efficacies and relevant mechanisms including newly elucidated ones and to suggest the importance of plant-based diet including phytoestrogens.

GENISTEIN Isoflavonoids are in a subclass of flavonoids, which are ketone or nonketone polyhydroxy polyphenol compounds as plant secondary metabolites, and fulfill many functions such as plant pigmentation, UV filtration and symbiotic nitrogen fixation. Genistein and daidzein are the mostly studied isoflavonoids derived from legumes. First, genistein, 40 ,5,7-trihydroxyisoflavone, is the predominant isoflavone in human diet, because it is mainly included in soybeans, peas, lentils and other beans (16). As a phytoestrogen, genistein has a potency of E2 by interacting with 2 isoforms of ERs: ERa and ERb. Genistein was shown to more closely interact with ERb than ERa from a luciferase reporter gene assay, showing that it has one-third the potency of E2 when it interacts with ERb and one-thousandth of the potency of E2 when it interacts with ERa (17). This higher binding affinity for ERb of genistein has been related with its action as an estrogen antagonist and chemopreventive activity. A higher binding affinity for ERb and ERb-mediated inhibition of ERa signaling pathway are associated with antiproliferative and antitumor properties, and a large series of phytoestrogens including genistein was investigated to have this tendency (18). The antiproliferative effect of genistein

was identified in the experiments with Sprague-Dawley rats, in which neonatal prepubertal exposure of E2 increased the number of terminal end buds and cell proliferation in mammary tissue; however, genistein reduced the number of terminal end buds and cell proliferation (19,20). We also have proven the antitumor effect of genistein in the recent studies. In BG-1 ovarian cancer cell line, E2 and bisphenol A (BPA), a typical endocrine disrupting chemical (EDC), identically increased the cell proliferation (21). On the other hand, the additional introduction of genistein significantly reduced the cell proliferation induced by E2 or BPA at 5.0 £ 10¡5 M and 2.5 £ 10¡5 M, respectively (21). This phenomenon was identified in the mouse xenograft model previously transplanted with BG-1 cell line. The cancer burden of E2 or BPA-administrated mice increased; however, when genistein was cotreated with E2 or BPA, the tumor volume was evidently reduced (22). From these results, genistein was confirmed to have a chemopreventive activity by abolishing the tumor progressive risk caused by E2 and BPA in ER-dependent BG-1 ovarian cancer. Genistein, furthermore, showed an anticarcinogenic effect on other cancers. It significantly inhibited the cell growth of human bladder cancer cell line 253 JB-V (23) and human renal carcinoma cell lines SMKT R-1, R-2, R-3, and R4 (24). In animal cancer models, genistein (15 mg/kg/day) or soybean-based diet reduced tumor cell migration of B16FO melanoma cells and angiogenesis in Balb/c mice (25). In addition, dietary soy at 10% and 20% (w/w) significantly was demonstrated to reduce the lung invasiveness of MDA-MB-435 human breast cancer cells (26). In another in vivo study using SD rats, dietary genistein (250 mg/kg diet) showed the protective effect against mammary and prostate cancers by regulating specific sex steroid receptors and growth factor signaling pathways (27). Anticarcinogenic mechanisms of genistein have been explained by its action on several biochemical targets. First, genistein is a potent inhibitor of protein tyrosine kinase (PTK), especially that of the epidermal growth factor receptor (EGFR) (28). Generally, PTKs play a central role in the control of cellular proliferation by transmitting mitogenic signals and its inactivation might be applied in the discovery of new anticancer compounds (29). Through dissecting signal transduction pathways of PTK, genistein can inhibit the growth of a wide variety of tumor cell types. For instance, EGF mediated growth stimulation of AGS gastric cancer cells was dosedependently reversed by co-incubation with genistein (30). Genistein also exerted multiple suppressive effects on the proliferation of ER-positive breast carcinoma cell lines by inhibiting estrogen-induced PTK activity (31). Second, the inhibition of topoisomerase II (Topo II), an important protein involved in the processes of DNA replication and cell proliferation and ribosomal S6 kinase (RS6K) activities, is associated with antiproliferative efficacy of genistein (32). Genistein inhibits Topo II and RS6K by stabilizing a cleavable Topo–DNA complex and modulating mRNA translation in vitro (33–35).

Downloaded by [New York University] at 06:06 25 May 2015

ANTIPROLIFERATIVE EFFECT OF PHYTOESTROGENS ON CANCERS

Stabilization of this complex may lead to protein-linked DNA strand breaks, cell growth suppression, and differentiation induction of several malignant cell lines (36). A recent study indicates that genistein may be an anticancer food component because it prevented the proliferation of HCT116 human colon carcinoma cells and halted the cell cycle in G2/M phase by inhibiting cellular Topo II, not Topo I, and DNA polymerases (37). Genistein has a potent capability to decrease the production of reactive oxygen species (ROS) by tumor cell types and cells of the immune system (28). Because the production of ROS, especially by activated cells of the immune system, has been postulated to play a role in tumor promotion, the ROS scavenging or antioxidant effect of genistein might be related with anticarcinogenesis. For example, in LNCaP and PC-3 prostate cancer cell lines, the antioxidant effect of genistein prevented cancer progression, metastasis, and invasion (38). Genistein has been shown to inhibit both the priming events necessary for high level ROS production and agonist-stimulated ROS production (39–41). In addition, recent studies also show that genistein can contribute to cancer prevention as an epigenome modifier (42). At the epigenetic level, DNA methylation is thought to inhibit transcription of genes by regulating alterations in chromatin structure, and its patterns have been used as a marker for the study of cancer-related epigenetics (42,43). In animal experiments, genistein was positively correlated with changes in prostate DNA methylation at CpG islands of specific mouse genes. Thus, genistein, may be involved in preventing the development of certain prostate and mammary cancers by maintaining a protective DNA methylation profile (44). From another epigenetic point of view, genistein was shown to increase histone acetylation, which is responsible for a loosened chromatin structure and the transcription activation machinery (45) and induce tumor suppressor gene expression, leading to cancer prevention (46, 47). Finally, the downregulation or inhibition of some growth factors including transforming growth factor-beta (TGF-b) and EGF is also associated with anticarcinogenic activity of genistein (48,49). In insulin-like growth factor-1 (IGF-1)-stimulated PC-3 prostate cancer cells, genistein also inhibited cell growth by suppressing downstream of IGF-1R activation (50). In our previous study, genistein was shown to effectively reverse the increased proliferation of BG-1 ovarian cancer by suppressing the crosstalk between ERa and IGF-1 receptor (IGF-1R) signaling pathways upregulated by E2 or BPA (22).

DAIDZEIN The next abundant isoflavone following genistein in soy beans is daidzein, which is 7-hydroxy-3-(4-hydroxyphenyl) chromen-4one. As in the case of genistein, daidzein has been also garnered interest for its antiproliferative and anticancer effects. Previously, daidzein has been shown to be a natural compound capable of inducing tumor cell death in various

3

types of cancer such as pancreatic, colon, breast, and ovarian cancers at concentrations over 5 or 10 mM (51–54). Recently, chemopreventive effects of daidzein on prostate cancer have been reported (55–57). Besides, it was investigated that equol, which is converted from daidzein by the intestinal flora (58), also decreased the risk of prostate cancer (59). In general, since equol is known to have a higher affinity to ER than daidzein does, equol producers might be expected to have greater efficacy of daidzein than nonequol producers : people who have plasma equol concentrations of < 40 nmol/L can be classified as nonequol producers; people having concentrations of >83 nmol/L can be defined as equol producers (60). Also, in the productive capacity of equol, Asians are reported to produce significantly higher amount of equol after eating foods containing soy isoflavones than Westerners do (61). In the previous study, equol (5, 10, and 50 mM) and daidzein (0.1, 1, and 5 mM) showed to decrease cell migration and invasion in human prostate cancer DU 145 cells (62). In addition to in vitro cellular models, daidzein exerted its anticancer effects in in vivo animal models. In 7,12-dimethylbenz[a]anthracene administered mice, daidzein represented antihepatic cancer effect by modulating the induction of cytochrome p450 CYP1A1 and CYP1B1 enzymes (63). Also in the previous study, daidzein effectively inhibited the growth of human leukemia HL cells in the subrenal capsules of mice and in diffusion chambers implanted into the peritoneal cavities of mice (64). 9,10-dimethyl-1,2-dibenzanthracene-induced mammary tumors in rats were inhibited by daidzein, and tumor latency was significantly increased in mammary tumor virus-neu mice at concentration ranges of 5250 mg/kg (65, 66). The potential anticancer mechanisms of daidzein can be considered as cell cycle arrest and apoptosis (67). It was shown that daidzein caused cell cycle arrest at G1 and G2/M phases and significantly induced apoptosis by increasing caspase-9 activity and decreasing cyclin D expression (53). Jin et al. (67) revealed that daidzein-induced apoptosis was mediated through the generation of ROS that perturbs mitochondrial function, leading to the increase in mitochondrial permeability, cytochrome C release, and the activation of caspases, which play critical roles in the execution of apoptosis (68). Moreover, daidzein was demonstrated to inhibit ovarian cancer cell growth by inducing apoptosis through caspase-3 activation (53). As a different type of apoptosis, daidzein was found to augment the effect of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) mediated apoptotic death in LNCaP prostate cancer cells (56). In this study, daidzein did not alter the expression of death receptors TRAIL-R1 and TRAIL-R2 but significantly increased TRAIL-induced disruption of mitochondrial membrane potential. Also in prostate cancer, the androgen receptor (AhR) dependent inhibition of CYP1 enzymes, which are involved in carcinogenesis via the activation of procarcinogenic compounds to carcinogenic metabolites, may explain other anticancer mechanisms of daidzein (63, 69). In terms of DNA topoisomerases, daidzein

4

K-A. HWANG AND K-C. CHOI

Downloaded by [New York University] at 06:06 25 May 2015

was also reported to inhibit the enzyme action, leading to cancer cell death (70). However, in HCT116 human colon carcinoma cells, daidzein did not inhibit Topo II unlike genistein, which induced the decrease in cell proliferation due to its inhibition of this enzyme (37).

RESVERATROL Resveratrol, a trans-3,40 ,5-trihydroxystilbene, is a plantderived polyphenolic stilbene and present in the skin of red grapes, other fruits, peanuts, and dietary supplements (71). Generally, red wine contains resveratrol on the order of 0.1– 14.3 mg/l (72). Resveratrol is categorized as a phytoestrogen due to its capability to compete with natural estrogens for binding to ERa and thus modulating the biological responses exerted by this receptor (73). Although the biological effects of resveratrol remain controversial due to its both estrogenic and antiestrogenic properties depending on cellular environment and the presence of coregulator, broad-spectrum beneficial health effects of resveratrol have been currently emphasized. Especially, resveratrol has obtained much attention due to its potential chemopreventive properties resulting from targeting multiple signaling pathways that promote cancer cell survival and tumor growth (74). Resveratrol can be regarded as an ideal molecule because several reports have shown that it does not damage nonmalignant cells although it may trigger direct cytotoxic effects on cancer cells (75–77). Many studies have demonstrated the antiproliferative and antitumor effects of resveratrol in some human cancers. For instance, resveratrol was found to inhibit the growth of prostate cancer cell lines, LNCaP, DU-145, and PC-3 cells at 2~40 mM (74) and revealed anticancer effect on A549 nonsmall cell lung cancer cells exposed to benzo(a)pyrene at 10 mM (78). In MCF-7 breast cancer cells, resveratrol showed antiproliferative effects by inducing apoptosis through a mitochondria/caspase pathway at concentrations less than 10 mM (79). In addition, resveratrol was found to significantly suppress the BG-1 ovarian cancer cell growth stimulated by E2 or BPA at 50 mM as did genistein in our recent study (80). In addition, resveratrol effectively reversed the BG-1 cell proliferation induced by E2 or BPA via inversely down-regulating the expressions of ERa, IGF-1R, p-IRS-1, p-Akt1/2/3, and cyclin D1 at both transcriptional and translational levels, suggesting that resveratrol is a novel candidate for the prevention of tumor progression caused by BPA via an effective inhibition of the cross-talk of ERa and IGF-1R signaling pathways (81). A previous study reported that topical resveratrol applications prevented skin cancer development in mice treated with a carcinogen (82). In vivo evaluations using animal models also showed that resveratrol effectively inhibited the growth of diverse cancers. Resveratrol inhibited the growth of lung cancer in a dose-dependent manner in nude mice xenografted with A549 cells at the administration levels of 15, 30, and 60 mg/kg (83). In orthotopic PCa xenografts, resveratrol

significantly inhibited tumor growth, progression, local invasion, and spontaneous metastasis of prostate cancer at 50 mg/ kg (84). Resveratrol-bovine serum albumin nanoparticles represented anticancer activity on xenografted nude mice at 50, 100, and 200 mg/kg, which were established by injecting a suspension of the human primary ovarian cancer cells, SKOV3 (85). Underlying mechanisms facilitated by resveratrol have been investigated with its chemopreventive actions. As a potential antioxidant, resveratrol has been shown to exert a strong inhibitory effect on the formation of free radicals and to reduce oxidative stress in PC-3 and DU-145 cells, leading to decrease the growth of prostate cancer (86). Resveratrol-mediated apoptosis is a main mechanism of its chemopreventive action. This type of apoptosis is associated with the activation of p53, a cellular tumor antigen or a tumor suppressor, and the induction of death receptor Fas/CD95/APO-1 in various cancer cells (87). For some cancer cell lines, such as breast cancer MCF-7, adenocarcinomic alveolar basal epithelia A549, and non-small lung cancer H460, resveratrol reduced the viability of these cell lines in a dose- and a time-dependent manner by partially increasing p53 protein level and involving the activation of caspases 9 and 7 and the cleavage of poly (ADP) ribose polymerase (PARP) (88). A significant increase of Fas ligand and Fas-associated protein with death domain by resveratrol was shown to mediate apoptosis and cell death in MCF-7 breast cancer cells (42). Other antitumor mechanisms of resveratrol include modulation of the transcription factor NF-kB (89, 90), inhibition of the cytochrome P450 isoenzyme CYP1A1 (91), alterations in androgenic actions (86, 92), and expression and activity of cyclooxygenase (COX) enzymes (93). Current preclinical and mechanistic data available from

TABLE 1 Chemopreventive mechanisms of genistein, daidzein, and resveratrol Phytoestrogen Genistein

Daidzein

Resveratrol

Mode of action

Ref.

PTK inhibition Topoisomerase inhibition Antioxidant property Epigenome modification Growth factor signaling downregulation Apoptosis CYP1 inhibition Topoisomerase inhibition Antioxidation Apoptosis NF-kB modulation CYP1 inhibition COX modulation

28, 30–31 33–37 28, 38–41 44–47 22, 48–50, 53, 56, 67 63, 69 70 86 42, 87, 88 89, 90 91 93

ANTIPROLIFERATIVE EFFECT OF PHYTOESTROGENS ON CANCERS

5

Downloaded by [New York University] at 06:06 25 May 2015

TABLE 2 Chemopreventive effects of genistein, daidzein, and resveratrol in laboratory animals Animal

Dose

Balb/c mouse

Genistein, 100 mg/kg/day

8 wk

Balb/c & C57BL/ 6 mouse

Genistein, 10 mg/kg/day

5 days

Nude mouse

12 wk

SD rat

Soy bean chips,10~20% (wt/wt) in diet Genistein, 25, 250 mg/kg diet

ICR mouse

Daidzein, 5, 25 mg/kg

SD rat

Daidzein, 200 mg/kg in diet

MMTV-neu mouse Daidzein, 250 mg/kg in diet Nude mouse Nude mouse

Nude mouse

Resveratrol, 15, 30, 60 mg/ kg/day Resveratrol, 50 mg/kg/day

Resveratrol, 50, 100, 200 mg/ kg/wk

Exposure time Administration

21 days

4 wk 120 days

34 wk 15 days 8 wk

4 wk

Major outcome

Intraperitoneal injection Inhibition of BPA-induced ovarian cancer growth Intraperitoneal injection Inhibition of mammarycarcinomainduced angiogenesis Feeding Inhibition of lung metastasis of breast cancer Feeding Suppression of DMBAinduced mammary tumor development Oral administration Inhibition of DMBA-induced carcinogenesis Feeding Reduction in DMBA-induced mammary tumor multiplicity Feeding Delay of mammary tumorigenesis Intravenous injection Inhibition of lung cancer growth Intraperitoneal injection Inhibition of prostate tumor growth, progression, and metastasis Intraperitoneal injection Inhibition of ovarian tumor growth

Ref. 22 25

26 27

63 65

66 83 84

85

SD D Sprague-Dawley.

numerous in vitro and in vivo studies can be implied to support anti-cancer effects of resveratrol to apply its potential as an anticancer agent in human populations (71).

CONCLUSION Genistein and daidzein, the 2 predominant isoflavones in soy products, and resveratrol derived from grapes and other vegetables are the most potent phytoestrogens that can be used for chemopreventive purposes against cancers because a great number of evidences have supported their efficacies. In addition to their excellent anticancer activity, low cytotoxicity for normal cells and easy ingestion from diet might spur the development of natural therapeutic agents including phytoestrogens. However, the controversies about their chemopreventive effect have existed due to some negative reasons (52,94). For genistein, it stimulates cancer cell growth at low concentration (10 mM) (95). This fact poses concern that plasma phytoestrogen concentrations of >10 mM cannot be achieved by dietary intake, and thus low phytoestrogen levels in the body may stimulate cancer growth. The

serum levels of >10 mM cannot be attainable even in Asian people who consume a respectable amount of phytoestrogens. In fact, the average value of plasma daidzein and genistein concentration of Japanese women who intake high isoflavone diet including tofu, natto, and miso was merely 72.46 and 206.09 nmol/L (0.07 and 0.2 mM), respectively (96). For resveratrol, it was also known to have poor bioavailability from the measurement of circulating levels in the body (97). In spite of those facts, the epidemiological study definitely shows that the incidence of some cancers is much lower in Asians than Westerners as referred above. Therefore, besides the serum levels of phytoestrogens, other factors such as tissue phytoestrogen concentration, which is currently unknown, and the timing of exposure to phytoestrogenic compounds are needed to be known (95). In addition, because the synergistic effect of mixed intake of phytoestrogens from daily diet strengthens the chemopreventive efficacies (95, 98), the serum concentration of each phytoestrogen may not be an absolute factor for estimating anticancer effects of phytoestrogens in vivo. As reviewed in this article, each phytoestrogen has its own ingenious or common chemopreventive mechanism (Table 1) and actually shows anticarcinogenic effects by inhibiting

Downloaded by [New York University] at 06:06 25 May 2015

6

K-A. HWANG AND K-C. CHOI

chemically induced tumor development or cancer progression in in vivo animal studies as shown in Table 2. Another remarkable point is that various modes of action of phytoestrogens have been successively found these days. For instance, genistein has been found to induce epigenetic modifications such as DNA methylation and histone acetylation, leading to cancer cell death. The possibility of genistein and daidzein to act as Topo poison was also presented. Along with general mechanisms such as antioxidant property and apoptosis, newly elucidated anticarcinogenic modes of action can raise statue of phytoestrogens as a promising reagent for cancer chemoprevention and/or treatment. Another point of this article is that genistein can be a candidate to negate the tumor progressive risk of BPA, a chemical EDC, by suppressing the crosstalk between ERa and IGF-1R signaling pathways. The removal of the carcinogenic risk of EDCs may be anticipated from a diet of phytoestrogens. Future studies on phytoestrogens may be needed to focus on the estimation for humans and to provide the natural strategies against cancers.

FUNDING This work was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (MEST) of the Republic of Korea (2014R1A1A2055295).

REFERENCES 1. Sirtori CR, Arnoldi A, and Johnson SK: Phytoestrogens: End of a tale? Ann Med 37, 423–438, 2005. 2. Adlercreutz H: Phytoestrogens and breast cancer. J Steroid Biochem Mol Biol 83, 113–118, 2002. 3. Ursin G, Bernstein L, and Pike MC: Breast cancer. Cancer Surv 19–20, 241–264, 1994. 4. Van Poppel H and Tombal B: Chemoprevention of prostate cancer with nutrients and supplements. Cancer Manag Res 3, 91–100, 2011. 5. Adlercreutz H: Epidemiology of phytoestrogens. Baillieres Clin Endocrinol Metab 12, 605–623, 1998. 6. Moon YJ, Wang X, and Morris ME: Dietary flavonoids: Effects on xenobiotic and carcinogen metabolism. Toxicol In Vitro 20, 187–210, 2006. 7. Virk-Baker MK, Nagy TR, and Barnes S: Role of phytoestrogens in cancer therapy. Planta Med 76, 1132–1142, 2010. 8. Liu MM, Huang Y, and Wang J: Developing phytoestrogens for breast cancer prevention. Anticancer Agents Med Chem 12, 1306–1313, 2012. 9. Mason JK and Thompson LU: Flaxseed and its lignan and oil components: Can they play a role in reducing the risk of and improving the treatment of breast cancer? Appl Physiol Nutr Metab 39, 663–678, 2014. 10. Charalambous C, Pitta CA, and Constantinou AI: Equol enhances tamoxifen’s anti-tumor activity by induction of caspase-mediated apoptosis in MCF-7 breast cancer cells. BMC Cancer 13, 238, 2013. 11. Li Y, Meeran SM, Patel SN, Chen H, Hardy TM, et al.: Epigenetic reactivation of estrogen receptor-alpha (ERalpha) by genistein enhances hormonal therapy sensitivity in ERalpha-negative breast cancer. Mol Cancer 12, 9, 2013.

12. Lindahl G, Saarinen N, Abrahamsson A, and Dabrosin C: Tamoxifen, flaxseed, and the lignan enterolactone increase stroma- and cancer cellderived IL-1Ra and decrease tumor angiogenesis in estrogen-dependent breast cancer. Cancer Res 71, 51–60, 2011. 13. Mishra R, Bhadauria S, Murthy PK, and Murthy PS: Glycine soya diet synergistically enhances the suppressive effect of tamoxifen and inhibits tamoxifen-promoted hepatocarcinogenesis in 7,12-dimethylbenz[alpha] anthracene-induced rat mammary tumor model. Food Chem Toxicol 49, 434–440, 2011. 14. Shu XO, Zheng Y, Cai H, Gu K, Chen Z, et al.: Soy food intake and breast cancer survival. Jama 302, 2437–2443, 2009. 15. Kang X, Zhang Q, Wang S, Huang X, and Jin S: Effect of soy isoflavones on breast cancer recurrence and death for patients receiving adjuvant endocrine therapy. Cmaj 182, 1857–1862, 2010. 16. Perabo FG, Von Low EC, Ellinger J, von Rucker A, Muller SC, et al.: Soy isoflavone genistein in prevention and treatment of prostate cancer. Prostate Cancer Prostatic Dis 11, 6–12, 2008. 17. Kuiper GG, Lemmen JG, Carlsson B, Corton JC, Safe SH, et al.: Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology 139, 4252–4263, 1998. 18. Pelekanou V and Leclercq G: Recent insights into the effect of natural and environmental estrogens on mammary development and carcinogenesis. Int J Dev Biol 55, 869–878, 2011. 19. Whitten PL and Patisaul HB: Cross-species and interassay comparisons of phytoestrogen action. Environ Health Perspect 109(Suppl 1), 5–20, 2001. 20. Murrill WB, Brown NM, Zhang JX, Manzolillo PA, Barnes S, et al.: Prepubertal genistein exposure suppresses mammary cancer and enhances gland differentiation in rats. Carcinogenesis 17, 1451–1457, 1996. 21. Hwang KA, Kang NH, Yi BR, Lee HR, Park MA, et al.: Genistein, a soy phytoestrogen, prevents the growth of BG-1 ovarian cancer cells induced by 17beta-estradiol or bisphenol A via the inhibition of cell cycle progression. Int J Oncol 42, 733–740, 2012. 22. Hwang KA, Park MA, Kang NH, Yi BR, Hyun SH, et al.: Anticancer effect of genistein on BG-1 ovarian cancer growth induced by 17 betaestradiol or bisphenol A via the suppression of the crosstalk between estrogen receptor alpha and insulin-like growth factor-1 receptor signaling pathways. Toxicol Appl Pharmacol 272, 637–646, 2013. 23. Singh AV, Franke AA, Blackburn GL, and Zhou JR: Soy phytochemicals prevent orthotopic growth and metastasis of bladder cancer in mice by alterations of cancer cell proliferation and apoptosis and tumor angiogenesis. Cancer Res 66, 1851–1858, 2006. 24. Sasamura H, Takahashi A, Miyao N, Yanase M, Masumori N, et al.: Inhibitory effect on expression of angiogenic factors by antiangiogenic agents in renal cell carcinoma. Br J Cancer 86, 768–773, 2002. 25. Farina HG, Pomies M, Alonso DF, and Gomez DE: Antitumor and antiangiogenic activity of soy isoflavone genistein in mouse models of melanoma and breast cancer. Oncol Rep 16, 885–891, 2006. 26. Connolly JM, Liu XH, and Rose DP: Effects of dietary menhaden oil, soy, and a cyclooxygenase inhibitor on human breast cancer cell growth and metastasis in nude mice. Nutr Cancer 29, 48–54, 1997. 27. Lamartiniere CA, Cotroneo MS, Fritz WA, Wang J, Mentor-Marcel R, et al.: Genistein chemoprevention: Timing and mechanisms of action in murine mammary and prostate. J Nutr 132, 552S–558S, 2002. 28. Peterson G: Evaluation of the biochemical targets of genistein in tumor cells. J Nutr 125, 784S–789S, 1995. 29. Bhuva HA and Kini SG: Synthesis, anticancer activity and docking of some substituted benzothiazoles as tyrosine kinase inhibitors. J Mol Graph Model 29, 32–37, 2010. 30. Piontek M, Hengels KJ, Porschen R, and Strohmeyer G: Antiproliferative effect of tyrosine kinase inhibitors in epidermal growth factor-stimulated growth of human gastric cancer cells. Anticancer Res 13, 2119–2123, 1993.

Downloaded by [New York University] at 06:06 25 May 2015

ANTIPROLIFERATIVE EFFECT OF PHYTOESTROGENS ON CANCERS 31. Shao Z, Wu J, and Shen Z: [Genistein exerts multiple suppressive effects on human breast carcinoma cells]. Zhonghua Zhong Liu Za Zhi 22, 362– 365, 2000. 32. Wei H, Bowen R, Zhang X, and Lebwohl M: Isoflavone genistein inhibits the initiation and promotion of two-stage skin carcinogenesis in mice. Carcinogenesis 19, 1509–1514, 1998. 33. Dean NM, Kanemitsu M, and Boynton AL: Effects of the tyrosine-kinase inhibitor genistein on DNA synthesis and phospholipid-derived second messenger generation in mouse 10T1/2 fibroblasts and rat liver T51B cells. Biochem Biophys Res Commun 165, 795–801, 1989. 34. Peterson G, Barnes S: Genistein inhibition of the growth of human breast cancer cells: independence from estrogen receptors and the multi-drug resistance gene. Biochem Biophys Res Commun 179, 661–667, 1991. 35. Zhou N, Yan Y, Li W, Wang Y, Zheng L, et al.: Genistein inhibition of topoisomerase IIalpha expression participated by Sp1 and Sp3 in HeLa cell. Int J Mol Sci 10, 3255–3268, 2009. 36. Gewirtz DA: Does bulk damage to DNA explain the cytostatic and cytotoxic effects of topoisomerase II inhibitors? Biochem Pharmacol 42, 2253–2258, 1991. 37. Mizushina Y, Shiomi K, Kuriyama I, Takahashi Y, and Yoshida H: Inhibitory effects of a major soy isoflavone, genistein, on human DNA topoisomerase II activity and cancer cell proliferation. Int J Oncol 43, 1117– 1124, 2013. 38. Suzuki K, Koike H, Matsui H, Ono Y, Hasumi M, et al.: Genistein, a soy isoflavone, induces glutathione peroxidase in the human prostate cancer cell lines LNCaP and PC-3. Int J Cancer 99, 846–852, 2002. 39. Akimaru K, Utsumi T, Sato EF, Klostergaard J, Inoue M, et al.: Role of tyrosyl phosphorylation in neutrophil priming by tumor necrosis factoralpha and granulocyte colony stimulating factor. Arch Biochem Biophys 298, 703–709, 1992. 40. Tanimura M, Kobuchi H, Utsumi T, Yoshioka T, Kataoka S, et al.: Neutrophil priming by granulocyte colony stimulating factor and its modulation by protein kinase inhibitors. Biochem Pharmacol 44, 1045–1052, 1992. 41. Wei H, Wei L, Frenkel K, Bowen R, and Barnes S: Inhibition of tumor promoter-induced hydrogen peroxide formation in vitro and in vivo by genistein. Nutr Cancer 20, 1–12, 1993. 42. Chen FP and Chien MH: Phytoestrogens induce apoptosis through a mitochondria/caspase-pathway in human breast cancer cells. Climacteric 17, 385–392, 2014. 43. Wietrzyk J, Grynkiewicz G, and Opolski A: Phytoestrogens in cancer prevention and therapy–mechanisms of their biological activity. Anticancer Res 25, 2357–2366, 2005. 44. Day JK, Bauer AM, DesBordes C, Zhuang Y, Kim BE, et al.: Genistein alters methylation patterns in mice. J Nutr 132, 2419S–2423S, 2002. 45. Esteller M: Epigenetics provides a new generation of oncogenes and tumour-suppressor genes. Br J Cancer 96(Suppl.), R26–R30, 2007. 46. Majid S, Kikuno N, Nelles J, Noonan E, Tanaka Y, et al.: Genistein induces the p21WAF1/CIP1 and p16INK4a tumor suppressor genes in prostate cancer cells by epigenetic mechanisms involving active chromatin modification. Cancer Res 68, 2736–2744, 2008. 47. Kikuno N, Shiina H, Urakami S, Kawamoto K, Hirata H, et al.: Genistein mediated histone acetylation and demethylation activates tumor suppressor genes in prostate cancer cells. Int J Cancer 123, 552–560, 2008. 48. Zhao R, Xiang N, Domann FE, and Zhong W: Effects of selenite and genistein on G2/M cell cycle arrest and apoptosis in human prostate cancer cells. Nutr Cancer 61, 397–407, 2009. 49. Banerjee S, Li Y, Wang Z, and Sarkar FH: Multi-targeted therapy of cancer by genistein. Cancer Lett 269, 226–242, 2008. 50. Lee J, Ju J, Park S, Hong SJ, and Yoon S: Inhibition of IGF-1 signaling by genistein: modulation of E-cadherin expression and downregulation of beta-catenin signaling in hormone refractory PC-3 prostate cancer cells. Nutr Cancer 64, 153–162, 2012.

7

51. Guo JM, Xiao BX, Dai DJ, Liu Q, and Ma HH: Effects of daidzein on estrogen-receptor-positive and negative pancreatic cancer cells in vitro. World J Gastroenterol 10, 860–863, 2004. 52. Guo JM, Xiao BX, Liu DH, Grant M, Zhang S, et al.: Biphasic effect of daidzein on cell growth of human colon cancer cells. Food Chem Toxicol 42, 1641–1646, 2004. 53. Gercel-Taylor C, Feitelson AK, and Taylor DD: Inhibitory effect of genistein and daidzein on ovarian cancer cell growth. Anticancer Res 24, 795–800, 2004. 54. Choi EJ and Kim GH: Daidzein causes cell cycle arrest at the G1 and G2/ M phases in human breast cancer MCF-7 and MDA-MB-453 cells. Phytomedicine 15, 683–690, 2008. 55. Dong X, Xu W, Sikes RA, and Wu C: Apoptotic effects of cooked and in vitro digested soy on human prostate cancer cells. Food Chem 135, 1643– 1652, 2012. 56. Szliszka E and Krol W: Soy isoflavones augment the effect of TRAILmediated apoptotic death in prostate cancer cells. Oncol Rep 26, 533– 541, 2011. 57. Singh-Gupta V, Zhang H, Yunker CK, Ahmad Z, Zwier D, et al.: Daidzein effect on hormone refractory prostate cancer in vitro and in vivo compared to genistein and soy extract: potentiation of radiotherapy. Pharm Res 27, 1115–1127, 2010. 58. Magee PJ: Is equol production beneficial to health? Proc Nutr Soc 70, 10– 18, 2011. 59. Sugiyama Y, Masumori N, Fukuta F, Yoneta A, Hida T, et al.: Influence of isoflavone intake and equol-producing intestinal flora on prostate cancer risk. Asian Pac J Cancer Prev 14, 1–4, 2013. 60. Setchell KD, Brown NM, and Lydeking-Olsen E: The clinical importance of the metabolite equol-a clue to the effectiveness of soy and its isoflavones. J Nutr 132, 3577–3584, 2002. 61. Song KB, Atkinson C, Frankenfeld CL, Jokela T, Wahala K, et al.: Prevalence of daidzein-metabolizing phenotypes differs between Caucasian and Korean American women and girls. J Nutr 136, 1347–1351, 2006. 62. Zheng W, Zhang Y, Ma D, Shi Y, Liu C, et al.: (C/¡)Equol inhibits invasion in prostate cancer DU145 cells possibly via down-regulation of matrix metalloproteinase-9, matrix metalloproteinase-2 and urokinasetype plasminogen activator by antioxidant activity. J Clin Biochem Nutr 51, 61–67, 2012. 63. Choi EJ and Kim T: Daidzein modulates induction of hepatic CYP1A1, 1B1, and AhR by 7,12-dimethylbenz[a]anthracene in mice. Arch Pharm Res 31, 1115–1119, 2008. 64. Jing Y, Nakaya K, and Han R: Differentiation of promyelocytic leukemia cells HL-60 induced by daidzein in vitro and in vivo. Anticancer Res 13, 1049–1054, 1993. 65. Constantinou AI, Lantvit D, Hawthorne M, Xu X, van Breemen RB, et al.: Chemopreventive effects of soy protein and purified soy isoflavones on DMBA-induced mammary tumors in female Sprague-Dawley rats. Nutr Cancer 41, 75–81, 2001. 66. Jin Z and MacDonald RS: Soy isoflavones increase latency of spontaneous mammary tumors in mice. J Nutr 132, 3186–3190, 2002. 67. Jin S, Zhang QY, Kang XM, Wang JX, and Zhao WH: Daidzein induces MCF-7 breast cancer cell apoptosis via the mitochondrial pathway. Ann Oncol 21, 263–268, 2010. 68. Li M, Kondo T, Zhao QL, Li FJ, Tanabe K, et al.: Apoptosis induced by cadmium in human lymphoma U937 cells through Ca2C-calpain and caspase-mitochondria- dependent pathways. J Biol Chem 275, 39702–39709, 2000. 69. Androutsopoulos VP, Spyrou I, Ploumidis A, Papalampros AE, Kyriakakis M, et al.: Expression Profile of CYP1A1 and CYP1B1 Enzymes in Colon and Bladder Tumors. PLoS One 8, e82487, 2013. 70. Russo P, Del Bufalo A, and Cesario A: Flavonoids acting on DNA topoisomerases: recent advances and future perspectives in cancer therapy. Curr Med Chem 19, 5287–5293, 2012.

Downloaded by [New York University] at 06:06 25 May 2015

8

K-A. HWANG AND K-C. CHOI

71. Athar M, Back JH, Tang X, Kim KH, Kopelovich L, et al.: Resveratrol: a review of preclinical studies for human cancer prevention. Toxicol Appl Pharmacol 224, 274–283, 2007. 72. Baur JA and Sinclair DA: Therapeutic potential of resveratrol: The in vivo evidence. Nat Rev Drug Discov 5, 493–506, 2006. 73. Chakraborty S, Levenson AS, and Biswas PK: Structural insights into Resveratrol’s antagonist and partial agonist actions on estrogen receptor alpha. BMC Struct Biol 13, 27, 2013. 74. Cimino S, Sortino G, Favilla V, Castelli T, Madonia M, et al.: Polyphenols: Key issues involved in chemoprevention of prostate cancer. Oxid Med Cell Longev 2012, 632959, 2012. 75. Marcsek ZL, Kocsis Z, Szende B, Tompa A: Effect of formaldehyde and resveratrol on the viability of Vero, HepG2 and MCF-7 cells. Cell Biol Int 31, 1214–1219, 2007. 76. Reagan-Shaw S, Mukhtar H, and Ahmad N: Resveratrol imparts photoprotection of normal cells and enhances the efficacy of radiation therapy in cancer cells. Photochem Photobiol 84, 415–421, 2008. 77. Baarine M, Thandapilly SJ, Louis XL, Mazue F, Yu L, et al.: Pro-apoptotic versus anti-apoptotic properties of dietary resveratrol on tumoral and normal cardiac cells. Genes Nutr 6, 161–169, 2011. 78. Ulasli SS, Celik S, Gunay E, Ozdemir M, Hazman O, et al.: Anticancer effects of thymoquinone, caffeic acid phenethyl ester and resveratrol on A549 non-small cell lung cancer cells exposed to benzo(a)pyrene. Asian Pac J Cancer Prev 14, 6159–6164, 2013. 79. Chen FP and Chien MH: Phytoestrogens induce apoptosis through a mitochondria/caspase-pathway in human breast cancer cells. Climacteric, 17, 385-392, 2014. 80. Kang NH, Hwang KA, Kim TH, Hyun SH, Jeung EB, et al.: Induced growth of BG-1 ovarian cancer cells by 17beta-estradiol or various endocrine disrupting chemicals was reversed by resveratrol via downregulation of cell cycle progression. Mol Med Rep 6, 151–156, 2012. 81. Kang NH, Hwang KA, Lee HR, Choi DW, and Choi KC: Resveratrol regulates the cell viability promoted by 17beta-estradiol or bisphenol A via down-regulation of the cross-talk between estrogen receptor alpha and insulin growth factor-1 receptor in BG-1 ovarian cancer cells. Food Chem Toxicol 59, 373–379, 2013. 82. Jang M, Cai L, Udeani GO, Slowing KV, Thomas CF, et al.: Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 275, 218–220, 1997. 83. Yin HT, Tian QZ, Guan L, Zhou Y, Huang XE, et al.: In vitro and in vivo evaluation of the antitumor efficiency of resveratrol against lung cancer. Asian Pac J Cancer Prev 14, 1703–1706, 2013. 84. Li K, Dias SJ, Rimando AM, Dhar S, Mizuno CS, et al.: Pterostilbene acts through metastasis-associated protein 1 to inhibit tumor growth, progression and metastasis in prostate cancer. PLoS One 8, e57542, 2013.

85. Guo L, Peng Y, Yao J, Sui L, Gu A, et al.: Anticancer activity and molecular mechanism of resveratrol-bovine serum albumin nanoparticles on subcutaneously implanted human primary ovarian carcinoma cells in nude mice. Cancer Biother Radiopharm 25, 471–477, 2010. 86. Ratan HL, Steward WP, Gescher AJ, and Mellon JK: Resveratrol–a prostate cancer chemopreventive agent? Urol Oncol 7, 223–227, 2002. 87. Athar M, Back JH, Kopelovich L, Bickers DR, and Kim AL: Multiple molecular targets of resveratrol: Anti-carcinogenic mechanisms. Arch Biochem Biophys 486, 95–102, 2009. 88. Ferraz da Costa DC, Casanova FA, Quarti J, Malheiros MS, Sanches D, et al.: Transient transfection of a wild-type p53 gene triggers resveratrolinduced apoptosis in cancer cells. PLoS One 7, e48746, 2012. 89. Leiro J, Arranz JA, Fraiz N, Sanmartin ML, Quezada E, et al.: Effect of cis-resveratrol on genes involved in nuclear factor kappa B signaling. Int Immunopharmacol 5, 393–406, 2005. 90. Gupta SC, Kannappan R, Reuter S, Kim JH, and Aggarwal BB: Chemosensitization of tumors by resveratrol. Ann N Y Acad Sci 1215, 150–160, 2011. 91. Chun YJ, Kim MY, and Guengerich FP: Resveratrol is a selective human cytochrome P450 1A1 inhibitor. Biochem Biophys Res Commun 262, 20– 24, 1999. 92. Benitez DA, Pozo-Guisado E, Alvarez-Barrientos A, Fernandez-Salguero PM, and Castellon EA: Mechanisms involved in resveratrol-induced apoptosis and cell cycle arrest in prostate cancer-derived cell lines. J Androl 28, 282–293, 2007. 93. Tang HY, Shih A, Cao HJ, Davis FB, Davis PJ, et al.: Resveratrolinduced cyclooxygenase-2 facilitates p53-dependent apoptosis in human breast cancer cells. Mol Cancer Ther 5, 2034–2042, 2006. 94. Mahmoud AM, Zhu T, Parray A, Siddique HR, Yang W, et al.: Differential effects of genistein on prostate cancer cells depend on mutational status of the androgen receptor. PLoS One 8, e78479, 2013. 95. Magee PJ and Rowland IR: Phyto-oestrogens, their mechanism of action: current evidence for a role in breast and prostate cancer. Br J Nutr 91, 513–531, 2004. 96. Arai Y, Uehara M, Sato Y, Kimira M, Eboshida A, et al.: Comparison of isoflavones among dietary intake, plasma concentration and urinary excretion for accurate estimation of phytoestrogen intake. J Epidemiol 10, 127–135, 2000. 97. Carter LG, D’Orazio JA, and Pearson KJ: Resveratrol and cancer: focus on in vivo evidence. Endocr Relat Cancer 21, R209–R25, 2014. 98. Dong X, Xu W, Sikes RA, and Wu C: Combination of low dose of genistein and daidzein has synergistic preventive effects on isogenic human prostate cancer cells when compared with individual soy isoflavone. Food Chem 141, 1923–1933, 2013.

Anticarcinogenic Effects of Dietary Phytoestrogens and Their Chemopreventive Mechanisms.

Phytoestrogens are phenolic compounds derived from plants and exert an estrogenic as well as an antiestrogenic effect and also various biological effi...
107KB Sizes 0 Downloads 12 Views