Next Article in Journal
A Comparison of the Effects of Oral Glutamine Dipeptide, Glutamine, and Alanine on Blood Amino Acid Availability in Rats Submitted to Insulin-Induced Hypoglycemia
Next Article in Special Issue
n-3 Polyunsaturated Fatty Acids and Mechanisms to Mitigate Inflammatory Paracrine Signaling in Obesity-Associated Breast Cancer
Previous Article in Journal
Severe Periodontitis Is Inversely Associated with Coffee Consumption in the Maintenance Phase of Periodontal Treatment
Previous Article in Special Issue
Effects of Perioperative Supplementation with Omega-3 Fatty Acids on Leukotriene B4 and Leukotriene B5 Production by Stimulated Neutrophils in Patients with Colorectal Cancer: A Randomized, Placebo-Controlled Intervention Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Use of Dietary Supplements to Alleviate Androgen Deprivation Therapy Side Effects during Prostate Cancer Treatment

1
Division of Experimental Urology, Department of Urology, Innsbruck Medical University, Innsbruck, A-6020 Austria
2
Department of Dietetics, University of Applied Sciences Tyrol, Innsbruck A-6020, Austria
*
Author to whom correspondence should be addressed.
Nutrients 2014, 6(10), 4491-4519; https://doi.org/10.3390/nu6104491
Submission received: 4 August 2014 / Revised: 7 September 2014 / Accepted: 19 September 2014 / Published: 21 October 2014
(This article belongs to the Special Issue Nutrition and Cancer)

Abstract

:
Prostate cancer (PCa), the most commonly diagnosed cancer and second leading cause of male cancer death in Western societies, is typically androgen-dependent, a characteristic that underlies the rationale of androgen deprivation therapy (ADT). Approximately 90% of patients initially respond to ADT strategies, however many experience side effects including hot flashes, cardiotoxicity, metabolic and musculoskeletal alterations. This review summarizes pre-clinical and clinical studies investigating the ability of dietary supplements to alleviate adverse effects arising from ADT. In particular, we focus on herbal compounds, phytoestrogens, selenium (Se), fatty acids (FA), calcium, and Vitamins D and E. Indeed, there is some evidence that calcium and Vitamin D can prevent the development of osteoporosis during ADT. On the other hand, caution should be taken with the antioxidants Se and Vitamin E until the basis underlying their respective association with type 2 diabetes mellitus and PCa tumor development has been clarified. However, many other promising supplements have not yet been subjected large-scale clinical trials making it difficult to assess their efficacy. Given the demographic trend of increased PCa diagnoses and dependence on ADT as a major therapeutic strategy, further studies are required to objectively evaluate these supplements as adjuvant for PCa patients receiving ADT.

1. Introduction

Prostate Cancer (PCa) is the most commonly diagnosed male cancer and the second leading cause of cancer death among men in Western societies [1,2]. Radical prostatectomy or primary radiation therapy are the preferred treatment modalities in men with locally confined PCa. For advanced tumors or tumor recurring after primary surgery or radiation therapy the androgen receptor (AR) and its signaling network are the prime targets of therapy. The androgen receptor orchestrates crucial oncogenic factors in PCa etiology since androgens drive proliferation, differentiation, and survival of benign and malignant prostate cells [3]. Hence, upon initial diagnosis, 80%–90% of PCa are androgen-dependent [4], an observation that underlies the rationale of androgen deprivation therapy (ADT), the current mainstay systemic treatment for advanced PCa [5]. Although highly effective, ADT is associated with considerable side effects that negatively affect the patient’s quality of life [6,7]. These adverse events include hot flashes [8], metabolic effects such as an induced metabolic syndrome (MetS) [9,10,11,12] including insulin resistance [13,14], cardiovascular (CV) diseases [15,16], musculoskeletal side effects characterized by reduced lean body mass and muscle strength, and osteoporosis [17,18,19,20] as well as depression and sexual dysfunction. Although several medical regimens have been developed [21], their impact on minimizing ADT side effects and improving quality of life is still under discussion. Recent statistics revealed an increasing use of complementary and alternative substances by PCa patients [22]. Indeed, approximately one in four patients with PCa uses at least one complementary or alternative method with the primary aim of ameliorating ADT-induced adverse effects [23,24]. In particular, herbal and dietary supplements appeal to patients because they are perceived as being “natural” with fewer side effects than prescription medicines. Despite the widespread use of alternatives to medical treatment options, little is known about their safety, efficacy and mechanism of action. The limitation of clinical studies investigating this issue leads to a lack of information concerning the use of different types of alternative interventions. This article focuses on the metabolic and musculoskeletal side effects of ADT, which are not alleviated by current treatment strategies. In particular, we discuss several adjuvant dietary options including herbs, phytoestrogens, selenium (Se), fatty acids (FA), calcium, and Vitamins D and E, whose use in the treatment of ADT side effects is supported by scientific evidence derived either from cell-based models, animal models or clinical trials.

2. ADT in the Treatment of PCa

The androgenic hormones testosterone (T) and 5α-dihydrotestosterone (DHT), which mediate their action through the AR, are essential for normal prostate development but also contribute to prostate tumor growth by regulating cell proliferation and differentiation. The concept of hormonal manipulation using ADT to restrict PCa growth was first described in 1941 by Huggins and Hodges [25] and is based on the observation that 80%–90% of PCa are androgen dependent. Since then, multiple strategies have been established to reduce serum androgen levels or to interfere with their function by inhibiting the AR. Current strategies for hormonal blockade used in the treatment of PCa have been reviewed recently [26] and include bilateral orchiectomy, luteinizing hormone-releasing hormone (LHRH) agonists or antagonists and anti-androgens (Figure 1 and Table 1).
Figure 1. Mechanisms of action of androgen deprivation therapy (ADT) for blockage of the hypothalamus-pituary axis.
Figure 1. Mechanisms of action of androgen deprivation therapy (ADT) for blockage of the hypothalamus-pituary axis.
Nutrients 06 04491 g001
Current ADT strategies used in the treatment of PCa include bilateral orchiectomy (surgical castration), LHRH agonists or antagonists and anti-androgens (medical castration). (1) Bilateral orchiectomy is the surgical removal of both testicles inhibiting the production of testicular testosterone (T) and estradiol; (2) LHRH analogs reversibly decrease T production by the testis resulting in the down-regulation of LHRH receptors and thus reduced levels of luteinizing hormone (LH), follicle stimulating hormone (FSH) and T; (3) Anti-androgens (including flutamide, nilutamide, and bicalutamide) may bind competitively to the AR in cells thereby inhibiting its activation, or (abiraterone) block de novo androgen synthesis, both leading to apoptosis and reduced prostate tumor growth.
Table 1. Treatment options for hormone reduction.
Table 1. Treatment options for hormone reduction.
ModalityDrugMechanismSide Effects
Surgical Castration
Bilateral Orchiectomy-Surgical removal of testicles, ↓THot flashes, reduced muscle mass and energy, anemia and osteoporosis
Medical Castration
LHRH InterferenceLHRH agonists
Leuproreline acetate (Trenantone®, Eligard®)
Goserelin (Zoladex®)
Triptorelin (Trelstar®)
Histrelin (Vantas®)
LHRH receptor downregulation after initial flare, ↓LH, ↓FSH, ↓THot flashes, reduced muscle mass and energy, anemia and osteoporosis, flare phenomenon, CV events, cardiotoxicity
LHRH antagonists
Degarelix (Firmagon®)
Blockade of LHRH receptor, ↓LH, ↓FSH, ↓THot flashes, reduced muscle mass and energy, anemia and osteoporosis, CV events, histamine release
AntiandrogensNon-Steroidal
Flutamide (Eulexin®)
Bicalutamide (Casodex®)
Nilutamide (Nilandron®)
Enzalutamide (Xtandi®)
Antagonizes AR in target tissues, ↑TGynecomastia, hepatotoxicity (flutamide), visual and respiratory disturbance and alcohol intolerance (nilutamide), GI problems ,fatigue and hot flushes (enzalutamide)
Steroidal
Cyproterone acetate
(Androcur®, Cyprostat®)
Antagonizes AR in target tissues, suppress LHRH secretion, ↓LH, ↓TGynecomastia, cardiovascular events, fluid retention, GI problems
Abiraterone acetate (Zytiga®)Inhibition of Cyp17A1 enzyme, suppresses T, estrogen and glucocorticoid biosynthesisVomiting, GI problems, swellings, weakness, cough, high blood pressure
LHRH, luteinizing hormone-releasing hormone; LH, luteinizing hormone; T, testosterone; AR, androgen receptor; GI, gastrointestinal; CV, cardiovascular.
Although accompanied by fewer side effects than “medical castration”, the use of surgical bilateral orchiectomy is currently limited in developed countries. Rather, medical-based approaches to achieve castration levels of circulating androgens are preferred. In contrast to surgical castration, medical castration using LHRH analogs reversibly decrease T production by the testis and are therefore the preferred treatment modality. There are two different classes of LHRH analogs: LHRH agonists and antagonists. LHRH agonists stimulate the LHRH receptors in the pituitary gland resulting in a temporary increase in LH and FSH secretion, which in turn causes an initial rise in T production (the so-called “flare phenomenon [27]”). However, constant LHRH stimulation leads to a negative feedback loop resulting in the down-regulation of LHRH receptors and thus reduced levels of LH, FSH and T. In contrast to LHRH agonists, LHRH antagonists competitively bind to their receptors in the pituitary gland thereby blocking their activation by the natural ligand, inducing a rapid decrease in LH, FSH, and T levels without any flare.
Anti-androgens differ mechanistically from the above-described castration therapies as they do not alter androgen induction by direct modification of the hypothalamic-pituitary-gonadal axis in the brain. Rather, most anti-androgens (including flutamide, nilutamide, and bicalutamide) bind competitively to the AR in cells inhibiting its activation, leading to apoptosis and reduced prostate tumor growth. By contrast, abiraterone blocks de novo androgen synthesis via irreversible inhibition of CYP17-A1, a rate-limiting enzyme that catalyzes the conversion of cholesterol to androgen and estrogen precursors. The new-generation drugs abiraterone and enzalutamide have been developed during the past 5 years [28] and thus knowledge concerning their side effects is still limited. Since serum T and estrogen levels are maintained for all anti-androgen drugs that target the AR, hypogonadal side effects are generally less pronounced [29]. However, anti-androgens are often used in combination with LHRH analogs, thus this article will not discuss the side effect profile of anti-androgen monotherapy.

3. Adverse Effects of ADT

Frequent side effects of ADT that result in poor quality of life include hot flashes, metabolic effects such as gynacomastia as well as an increased body mass index, insulin resistance, metabolic syndrome (MetS), cardiovascular (CV) diseases, and musculoskeletal effects including reduced muscle mass, osteoporosis, and also sexual dysfunction (summarized Table 2) [1,7,30,31,32,33,34,35] Of these adverse events, metabolic and musculoskeletal effects are the most prevalent and distressing side effects reported by patients [21].
ADT targets gonadal function. Consequently, hypogonadism is prevalent in PCa patients undergoing ADT compared to those that undergo surgery and/or radiation therapy or compared to age-matched controls [7]. Thus, ADT induces a profound hypogonadism, which in turn is responsible for increased body mass index, increased fat mass, reduced lean body mass and muscle strength, and osteoporosis. Besides the desired physiological consequences of ADT in reducing serum androgens, hormonal castration is also associated with a decrease in circulating estrogens that are synthesized from androgens by peripheral aromatization (Figure 2). Despite having normal to elevated serum T levels, men with congenital aromatase deficiency (and thus, non-detectable serum estrogen levels) have a high prevalence of osteoporosis, insulin resistance and metabolic syndrome (MetS) [36], an observation that underscores the importance of estrogens in men. Moreover, it is decreased estrogen rather than T levels that are responsible for decreased bone density, accelerated rate of bone loss, and increased fracture incidence [37]. Thus, side effects induced by ADT leading to hot flashes, osteoporosis, MetS and higher CV events are related to androgen as well as estrogen deficiencies [35,38]. However, the relative contribution of T and estrogen to these adverse effects remains unclear.
ADT is associated with multiple adverse effects, many of which are related to androgen as well as estrogen deficiency that occur as a result of treatment [30,35].
Table 2. Summary of dietary supplements for the management of androgen deprivation therapy (ADT) side effects.
Table 2. Summary of dietary supplements for the management of androgen deprivation therapy (ADT) side effects.
Side EffectPostulated ManagementEfficacyReference
Hot FlushesHerbs
Black cohoshMinimal, ↓sweating symptoms, may ↓hot flashes in women.[39,40]
Dong quaiNo benefit in women or men.[41]
GinsengMinimal effects in women.[42]
Phyto-Estrogens
Soy-IsoflavonsNo effect with supplements in men; possible impact with dietary source seen in women.[43,44,45,46,47,48,49]
FlaxseedUnknown, initial studies showed impact on hormonal levels and serum lipids.[50]
Vitamin EPreviously recommended, but increased risk for diabetes. Might also increase the risk for PCa (SELECT trial).[43,51,52,53,54,55,56,57,58]
OsteoporosisHerbs
Black cohoshPotentially effective in preclinical studies (only studied in female animals).[40,59]
Phyto-estrogens
Soy-IsoflavonesPotentially effective in preclinical studies (castrated male animals).[60,61]
Fatty acids
Omega-3 FA(CLA)Potentially effective in preclinical studies (male and female animals).[62,63,64,65,66]
Calcium and
Vitamin D
Effective in men and postmenopausal women.[67,68,69,70,71,72]
Cardiovascular EventsHerbs
GinsengNo benefit.[42]
GarlicShowed to reduce blood pressure.[73,74]
Phyto-Estrogens
Soy-IsoflavonesNo effect. Might reduce ↓LDL-cholesterol.[46,75,76]
FlaxseedPotentially effective, reduced ↓LDL-cholesterol in postmenopausal women.[77]
Fatty acids
Omega-3 FANo benefit. Postulated mechanism: ↓TG[78,79,80,81,82]
SeleniumNo effect. Might have adverse effects on diabetes. Increased PCa risk (SELECT trial).[24,56,58, 83,84,85,86,87,88,89,90]
Vitamin ENegative association with CV health. Might increase PCa risk. No benefit in women.[43,51,55, 57,58,86, 91,92]
CV, cardiovascular; CLA, conjugated linoleic acid; LDL, low density lipoprotein; TG, triglyceride.
Figure 2. Side effects associated with androgen deprivation therapy (ADT).
Figure 2. Side effects associated with androgen deprivation therapy (ADT).
Nutrients 06 04491 g002

3.1. Metabolic and Cardiovascular Side Effects

A number of prospective studies have shown that ADT increases abdominal fat and serum triglycerides (TG) and decreases insulin sensitivity [93]. Additionally, cross-sectional studies have shown that men receiving ADT are more likely to meet the diagnostic criteria of MetS [94,95]. Moreover, long term ADT therapy caused significantly higher levels of fasting insulin and glucose compared to men with PCa not on ADT as well as to age-matched controls [96]. A retrospective study involving 44 PCa patients who received ADT showed an increase in fasting blood glucose, total cholesterol, LDL cholesterol, and TG, but a decrease in HDL cholesterol in these patients [34]. In addition, longitudinal studies indicate that lower T levels in men independently predict MetS [97,98] and type 2 diabetes mellitus (T2DM) [99]. Because MetS is independently associated with CV mortality [100], it is plausible that the positive association between lower serum T levels and MetS may at least in part explain the higher CV mortality in men with PCa receiving ADT (with therapy-induced hypogonadism as the likely trigger of these events) [101]. The protective role of T on the CV system and its relationship to MetS, CV morbidity and mortality has been recently reviewed [98]. Since CV disease has become the most common cause of non-PCa-related death among this patient group [102], the potential risks of ADT with regards to CV events should be carefully weighed against the expected benefits. Moreover, since metabolic complications predominantly occur within 3–6 months after starting ADT therapy, close observation of patients especially during the first year of ADT is highly recommended [93].

3.2. Musculoskeletal Side Effects

Given that 25–40% of patients already have osteoporosis at PCa diagnosis, it must be considered that the symptoms will worsen upon administration of ADT. In general, two different forms of osteoporosis are known [103]. Whereas primary osteoporosis is caused by malnutrition and genetic predisposition, secondary osteoporosis predominantly arises in men due to hypogonadism, including that experienced by PCa patients receiving ADT [104]. Longitudinal studies report that approximately 35% of men on LHRH agonists experience at least one skeletal fracture and approximately 20% will be diagnosed as osteoporotic or osteopenic within 7 years of starting therapy [105]. The absolute excess risk of fractures ranges from 5%–7% over an average of 6 years follow-up compared to men not receiving ADT [31,106]. In particular, treatment with either LHRH analogs or orchiectomy is associated with a significant decline in bone mineral density (BMD) and increased risk of bone fractures [31,106]. In this context, it should be noted that bone fractures in men with PCa have been associated with higher mortality rates [107]. Cross-sectional studies have shown that BMD at the hip and lumbar spine decreases with the duration of continuous ADT [108]. However, BMD appears to initially decline at a fast rate. For example, greater declines of BMD were observed among recent versus chronic ADT users with stronger declines in both groups in contrast to ADT naïve men [109]. BMD declined at multiple sites including the lumbar spine and hip in the first year after starting ADT, however subsequent changes in the second year were much smaller and no longer statistically significant [110]. A larger study followed 618 men on ADT for up to 7 years with annual dual-energy X-ray absorptiometry (DXA) scans [111]. Whilst steady annual declines in BMD among men with normal BMD at baseline were noted, only 38 men with normal BMD at baseline still remained in the study by 3 years [111]. Thus, due to the lack of longitudinal studies, it is currently not possible to determine whether BMD loss beyond the first year of ADT is attenuated.

4. Dietary Supplements

The use of dietary supplements to alleviate side effects of ADT is an attractive approach for the majority of PCa patients. Indeed, an array of dietary supplements proposed to be useful in intervening with side effects due to declining hormone levels in men and women are promoted typically via media that lack scientific evidence. However, it should be noted that many of these supplements have been studied in the context of hormone deprivation during menopause or in the course of breast cancer therapy in women with no clinical studies carried out in men thus far (e.g., black cohosh, ginseng, garlic). Thus, it is difficult to conclude whether these dietary supplements will exhibit a similar efficacy in PCa patients receiving ADT. Of the few dietary supplement studies that have been performed in men, most were primarily aimed at reducing the risk of PCa development or reducing the risk of disease progression after initial diagnosis or first line treatment. Thus, studies in men receiving ADT are rare. Consequently, it is inappropriate to assume that beneficial (or non-beneficial) effects on PCa development or progression will similarly translate to modifying the adverse effects associated with ADT.

4.1. Herbs

Herbal supplemental approaches to manage metabolic and musculoskeletal side effects like hot flashes and osteoporosis have been extensively evaluated in breast cancer and menopausal women in small randomized trials with varying success [112,113]. Thus, it seems evident to evaluate the potential of the most promising of these herbs for the treatment of side effects in men who are being treated for PCa with ADT.

4.1.1. Black Cohosh (Cimicifuga racemosa)

The herbal supplement black cohosh, which is approved for the treatment of menopausal symptoms by the German health authority (Commission E), has also shown to have MetS-preventive and bone-protective properties in recent preclinical studies [59,114]. A randomized, placebo-controlled trial found no difference in the frequency of hot flashes but a significantly lower incidence of sweating in menopausal women with a history of breast cancer in the black cohosh group compared to placebo [39]. This finding is particularly pertinent given that a significant placebo effect is consistently observed in investigations of hot flashes, with the placebo effect reportedly sufficient to reduce hot flashes by up to 75% [115]. On the other hand, significant evidence of potentially beneficial effects on bone tissue was shown, although it should be noted that all clinical studies were conducted in females [59,116]. Of interest is one study that was carried out in orchidectomized male rats and showed to prevent osteoporosis in vivo [117].
The mechanism underlying the activity of black cohosh remains poorly understood. There is a lack of scientific evidence regarding its proposed estrogenic activity. However, it was recently shown to attenuate nucleoside uptake into cells and thus may have an impact on tumor treatment by nucleoside analogs [118]. One may speculate that its modulation of adenosine signaling may be beneficial for CV side effects of ADT [119], but this has yet to be investigated. Despite the lack of knowledge regarding the mechanism of action of black cohosh, its long history of use reveals that black cohosh is well tolerated. The use of black cohosh in PCa patients undergoing ADT may be warranted for hot flushes, CV and osteoporotic side effects. Importantly, further systematic studies assessing the safety and efficacy of black cohosh in alleviating ADT-induced side effects may be worth pursuing.

4.1.2. Dong Quai (Angelica sinensis)

Dong quai is a traditional Chinese herbal remedy most commonly used in the treatment of female reproductive problems. According to our knowledge, there are no pre-clinical studies addressing its effects in PCa. However, a small randomized clinical trial was conducted in men receiving ADT where dong quai was shown to be ineffective in reducing hot flashes [41]. Similarly, randomized trials in women also found no effect of dong quai on hot flashes beyond a placebo, irrespective of whether the herb was used alone or as part of a complex multi-ingredient intervention [120]. Taken together, the current evidence does not support the use of dong quai in patients undergoing ADT.

4.1.3. Ginseng (Panax ginseng)

Ginseng extract is widely used in traditional Chinese medicine and was reported to reduce fatigue, insomnia and depression in post-menopausal women, although there was no significant benefit on hot flashes [121]. However, a recent review of studies examining the efficacy of ginseng on menopausal symptoms highlighted the poor quality and bias of many randomized clinical trials conducted to date, raising doubt as to the usefulness of this herb in managing menopause symptoms [42]. Nonetheless, several pre-clinical and clinical studies have shown that ginseng may possess anti-cancer and hypoglycemic properties, the latter of potential benefit in ameliorating MetS in men receiving ADT [122,123].

4.1.4. Garlic (Allium sativum)

Garlic is frequently used as a dietary supplement for the treatment of hyperlipidemia, heart disease, and hypertension [124]. In addition, there is evidence that garlic is associated with blood pressure reduction in patients with elevated systolic blood pressure (10–12 mmHg systolic, 6–9 mmHg diastolic) but not in normotensive patients [73,74]. In this respect, it is conceivable that garlic may also reduce CV effects in PCa patients undergoing ADT. However, there is currently insufficient data to support this hypothesis and further studies that specifically address this question would be required [125].

4.2. Phytoestrogens

In the past, suppression of T was achieved using high doses of estrogens (estradiol) or selective estrogen receptor modulators [126,127,128,129]. However, these treatments were prone to severe and even fatal CV side effects [130,131]. Consequently, these treatments have been replaced by other therapeutics such as LHRH analogs. Phytoestrogens (plant estrogens) are non-steroidal naturally occurring phenolic compounds with known estrogenic effects and estrogen receptor (ER-β and/or ERα) binding properties [132,133,134]. Thus, these “mild” estrogens could possibly serve as natural alternatives with potentially fewer side effects. Indeed, phytoestrogens have been shown to improve metabolic health, reduce CV risk, and improve BMD and brain function [60,75,135]. There are three classes of phyto-estrogens, which are categorized according to their chemical structure as isoflavones, lignans or coumestans. Isoflavones are the largest and also the most extensively studied group of phytoestrogens, which includes genistein, daidzein and glycitein. Isoflavones are found in highest amounts in soybeans, flaxseed and legumes. Soy is stably integrated into Asian diets where daily intake is at least 40 times higher than among Western populations [136]. This is considered to be one of the factors contributing to a much lower incidence of prostate and breast cancer in Asian countries [137]. In addition, increased consumption of isoflavones has been associated with decreased incidence of diabetes and heart disease in South and East Asia [138,44]. In clinical trials, isoflavones have shown an improvement in hot flash severity, glycemic control, MetS and inflammatory profile in both men and postmenopausal women [45,76,139]. However, in a recent double-blinded, randomized, placebo-controlled pilot study, phytoestrogens failed to improve metabolic or inflammatory parameters of men with PCa during ADT [47]. This is consistent with another pilot study conducted in 33 men where high dose isoflavones showed no significant improvement in cognition, vasomotor symptoms or any other aspect of quality of life measures compared to placebo in androgen deprived men [140]. Thus, there is currently no clinical evidence for the proposed improvement of ADT-induced side effects by phytoestrogens, although limitations of these studies including small cohort sizes and short treatment durations should be taken into account.

4.3. Selenium (Se)

Se is an essential trace element, which is incorporated as selenocysteine into selenoproteins, many of which are reactive oxygen species (ROS) scavenging enzymes, such as glutathione peroxidase and thioredoxin reductase [141,142]. Thus, Se may be useful in decreasing oxidative stress and low-grade inflammation. Notably, plasma biomarkers of oxidative stress and low-grade inflammation are associated with MetS, obesity and insulin resistance, conditions that are common side effects of ADT [143]. Se was shown to have strong anti-proliferative and pro-apoptotic effects on human PCa cells and to improve severe metabolic side effects in patients [144,83]. Moreover, cancer prevention studies indicated that Se decreases ROS and is associated with decreased incidence of PCa [145,146]. Taken together, these observations provided a strong mechanistic rationale to combine ADT and Se for the treatment of PCa [5] and led to a number of epidemiological studies and clinical trials [83,51]. However, epidemiological studies have suggested that supranutritional Se intake and high plasma Se levels are not necessarily preventive against cancer, and may even be a possible risk factor for developing T2DM [83]. For example, supplementation with Se and/or Vitamin E in the large-scale Selenium and Vitamin E Cancer Prevention Trial (SELECT) did not prevent the development of PCa, rather, the incidence of newly diagnosed T2DM increased among the Se-supplemented participants [83,51]. Whilst the Nutritional Prevention of Cancer (NPC) study reported a decreased risk of PCa among Se supplemented men, an increased risk of T2DM was also observed in participants with baseline plasma Se levels in the top tertile [147]. Since then, several longitudinal studies have failed to support a causal role of Se in T2DM, although cross-sectional studies continued to find significant associations between circulating Se levels and T2DM [148]. For example, serum Se was observed to be associated with adipocytokines, such as TNF-α, VCAM-1, leptin, FABP-4, and MCP-1 [149,150] and adiponectin [151]. Although on the other hand, an analysis across randomized groups showed that Se supplementation had no effect on adiponectin levels after six months of treatment [84]. Thus, it appears likely that the reported link between Se and T2DM is due to indirect effects of Se-containing ROS scavenging enzymes, affecting the hydrogen peroxide level that in turn modulates both glucose-induced insulin secretion and insulin-induced signaling [152,153]. Moreover, Se homeostasis is modulated by factors related to carbohydrate metabolism, suggesting that low serum Se levels may themselves be a consequence of dysregulated energy metabolism in T2DM [153]. In addition, there appears to be a significant interaction between dietary intake of phytoestrogens and Se with important implications for heart disease, cancer, diabetes, and other conditions related to body weight [154].
From the current prospective—especially since to date there has been no study specifically evaluating the supplementation of Se in PCa patients receiving ADT—a combination of ADT with Se cannot be recommended given that many patients develop pre-diabetes.

4.4. Fatty Acids (FA)

The relative amounts and different types of dietary FA are thought to play critical roles in PCa associated MetS. Total FA intake and the ratio of omega-3 (ω-3) to omega-6 (ω-6) polyunsaturated FA (PUFAs) in the Western diet have increased significantly since the Industrial Revolution [155]. The effects of ω-3 PUFAs on CV disease, cancer as well as MetS have been investigated extensively [78,156,157,158,159,160,161]. Consequently, the United States Food and Drug Administration (FDA) has approved ω-3 PUFAs for the prevention of CV adverse outcomes by the postulated mechanism of lowering serum triglyceride levels [162,163]. Additionally, three common dietary ω-3 FA—alpha-linolenic acid (ALA), eicosa-pentaenoic acid (EPA), and docosahexaenoic acid (DHA)—were proposed to exhibit anti-inflammatory properties [50,79,164]. Since inflammation is a major risk factor for the development of CV disease, these ω-3 FA may reduce the risk of CV disease [50,79,164]. Various pre-clinical, epidemiological and clinical studies have investigated the influence of ω-3 and ω-6 FA on the development and progression of PCa [80,155,158,161,165,166,167,168,169,170,171,172,173]. However, these studies have yielded contradictory results. In particular, dietary intake of long-chain ω-3 PUFA or its individual components (EPA, DHA, docosapentaenoic acid (DPA) and ALA) have been associated with PCa risk and progression [80,155,158,161,165,166,167,168,169,170,171,172,173]. On the other hand, encouraging results were obtained from clinical trials showing potential anti-inflammatory effects of ω-3 FA [77,174,175,176,177]. Other trials investigating higher doses of ω-3 FA (>1 g/day EPA and/or DHA) in populations at high risk for CV disease reported improvements (i.e., reduced concentrations) of selected inflammatory markers [81,178,179]. However, it should be noted that in these studies the same dosage of ω-3 FA showed mixed response rates in healthy adults, and no beneficial effect in patients that received lower doses (<1 g/day) [81,174,175,178,179,180,181,182,183,184,185]. A single study has reported that very high intakes (6.6 g/day), which are well beyond the current recommendations (500 mg/day–1 g/day), may raise blood concentrations of some inflammation markers such as soluble tumor necrosis factor receptors 1 and 2 (sTNF-Rs 1 and 2) [79,185]. It is likely that differences in dosage, study population characteristics, the source of ω-3 FA, study duration, and background diet may explain the inconsistencies in these findings. In this respect, it may be noted that the majority of clinical trials have used marine sources of ω-3 FA (EPA and DHA from fish), whereas few have examined FA from plants (ALA from flax oil) and only two trials compared both sources [175,82]. The heterogeneous results from these studies might also be due to variations in measuring FA consumption of individuals and different techniques assessing their diet [186,187,188,189,190]. The main mechanisms underlying the purported anticancer effects of modulating dietary fat appear to be through reduced insulin-like growth factor signaling and alterations in membrane ω-6 to ω-3 FA ratios leading to suppressed COX-2-dependent PGE-2 production and reduction of inflammation via modification of the eicosanoid pathway [191,192,193,194,195]. Moreover, decreased PGE-2 levels are expected to decrease estrogen production and further also modify androgen production [190].
In summary, although ω-3 FA exhibits some anti-inflammatory potential, there is still a lack of consensus regarding their optimal use and dosage. Moreover, results from the aforementioned studies have to be interpreted with caution, since the metabolic conditions during ADT are different. Further research is warranted to better elucidate the mechanism of action and ideal consumption of ω-3 FA for potential CV health benefits during ADT.

4.5. Calcium and Vitamin D

Osteoporosis is a common and one of the most debilitating side effects of ADT. The most important nutritional factors contributing to osteoporosis include deficiencies in Vitamin D and calcium. Measurement of Vitamin D levels in osteoporotic males by a large multi-center study in the US (MrOs) revealed a deficiency in 26% and an insufficiency in 72% of subjects [54]. Because deterioration of BMD occurs soon after initiation of ADT therapy [67], the European Association of Urology does not specify recommendations but states that calcium supplementation is protective [196]. The National Comprehensive Cancer Network cite the National Osteoporosis foundation guidelines that recommend calcium (1000 mg daily) and Vitamin D (800–1000 IU daily) supplementation for all men aged 50 or above [197]. Notably however, data supporting this recommendation are lacking as shown in a recent systematic review [196]. One cross-sectional study suggested an association between low calcium intake and a greater likelihood of osteoporosis in men with PCa of whom 71 % were undergoing ADT. Unfortunately, Vitamin D use was not examined in this study [198]. Another trial reported a positive association of calcium and Vitamin D use (examined together) on hip and lumbar spine BMD in men on ADT [199]. Importantly, results from 12 different clinical trials revealed that the commonly recommended doses, of 500–1000 mg calcium and 200–500 IU Vitamin D per day still result in BMD loss in men receiving ADT [196]. Alibhai et al. examined long-term effects of calcium and Vitamin D in a prospective 3-year matched cohort study comparing PCa patients with and without ADT. This study found that Vitamin D but not calcium may be protective particularly in the first year of ADT [32]. In a multivariate analysis, it was further shown that the mean daily calcium intake in men receiving ADT was significantly lower in men who suffered from osteoporosis compared to those without osteoporosis [198].
In summary, calcium and Vitamin D supplementation is a recommended complementary therapy not only in elderly men with osteoporosis but also in men undergoing ADT even though the long-term impact of ADT on BMD and the value of calcium and Vitamin D in ameliorating negative effects remains to be elucidated more precisely.

4.6. Vitamin E

Vitamin E is a potent antioxidant, which is of interest to ameliorate hot flushes and CV associated side effects of ADT. Vitamin E has a long history in the treatment of pre-eclampsia (characterized by high blood pressure in pregnant women), premenstrual syndrome, painful periods, menopausal syndrome, hot flashes associated with breast cancer, and breast cysts even though randomized controlled clinical studies did not reveal any benefit [52,53,86,200]. Moreover, the Physicians Health Study II concluded that Vitamin E does not reduce the risk of major CV events (non-fatal myocardial infarction, non-fatal stroke, or CV disease death) [57]. Similarly, the Women’s Health Study, which comprised approximately 40,000 healthy women, found that Vitamin E did not reduce the risk of death or major CV events. Interestingly however, there was a significant reduction in the secondary endpoint of CV deaths and in major CV events among a subgroup of women aged 65 or over [55]. The Women’s Antioxidant Cardiovascular Study found that there were no overall effects of Vitamin E on CV events among women at high risk of CV disease [91]. Moreover, the intake of Vitamin E has been shown to increase all-cause mortality and may even have negative effects on CV health [92].
There have been a number of studies in men, which have purported a positive effect of Vitamin E on hot flushes and high blood pressure [201]. However, most of these studies have yielded inconclusive or conflicting findings or a lack of benefit for its use, illustrating the need for studies of higher quality in this area. Thus, clinical trials have failed to recapitulate the promising findings of in vitro and many observational studies. Possible reasons for this discrepancy may be that clinical trials are too short in duration to reverse the results of decades of oxidative stress contributing to atherosclerosis or that the antioxidants selected for study were chosen for their ease of availability rather than proven efficacy (Vitamin E) [202]. Recent evidence from the SELECT trial revealed an increased risk for PCa in the Vitamin E supplemented group. Taken together, current evidence does not support a beneficial effect for Vitamin E, and its use as a supplementary treatment is therefore not recommended [56,88].

5. Discussion

The administration of ADT is associated with a diverse set of known side effects that have a significant impact on patient quality of life, overall health, and mortality. Some of the dietary supplements discussed in this review may be beneficial for patients undergoing ADT. The value of calcium and Vitamin D supplementation remains to be elucidated more precisely; however, because of their long and safe history of usage they may be recommended in the prevention of osteoporosis during ADT. Phytoestrogens were shown to prevent hot flashes and other climacteric complaints and exert anti-osteoporotic effects in women. However, positive effects on CV health are still questionable and require further elucidation, especially with respect to their effects in PCa patients receiving ADT. In addition, further clinical trials evaluating the efficacy of isoflavones must be conducted before their use for relieving ADT-induced side effects can be advocated. We can conclude that dietary interventions with herbal substances may in fact be helpful in the treatment of adverse effects arising from ADT [43]; however, more clinical randomized studies in PCa patients on ADT are highly warranted to support these findings. The long history of use and lack of adverse effects of black cohosh in the treatment of climacteric complaints in women is particularly encouraging. Further evaluation of its proposed osteo-protective and anti-metabolic effects in conjunction with ADT in randomized and controlled clinical studies is also warranted. Recent data obtained in the large SELECT trial suggest that combined supplementation of Vitamin E and supranutritional Se may increase the risk of PCa, making this a non-recommended treatment for men receiving ADT. From the current perspective, a combination of ADT with Se, which has been associated with an increased risk for the development of T2DM, cannot be recommended given that many patients develop pre-diabetes.

6. Conclusions

In summary, dietary supplements are active compounds with as yet mostly poorly defined effects. As such, the unregulated self-prescription of active compounds such as soy, Se or Vitamin E may in some cases even prove to be harmful or negatively interfere with cancer treatment. Given the prevalent use of alternative dietary supplements in PCa patients, there is an urgent need to (1) perform rigorous research to determine the precise physiological effects of these different supplements with respect to relieving side effects of ADT; (2) conduct clinical trials of these supplements in men undergoing ADT and (3) establish more open lines of communication between patients and physicians regarding the use of dietary supplements and their integration into conventional treatment strategies.

Acknowledgments

We thank Natalie Sampson for reading the manuscript and performing English corrections. This work was supported by the Competence Center Oncotyrol (project 2.3.3) within the scope of the Austrian Competence Center for Excellent Technologies (COMET)—program conducted by the Austrian Research Promotion Agency (FFG) and a Category B research funding from ÖKH-KG/Tirol given to Andrea Dueregger. Andrea Dueregger was partly funded by autonomous province Bozen (Grant Number: 37/40.3) given to Petra Massoner.

Author Contributions

Andrea Dueregger and Iris E. Eder designed and wrote the review. Isabel Heidegger wrote parts of the manuscript. Bernhard Perktold, Reinhold Ramoner and Helmut Klocker were significantly involved in designing the review regarding its content and also in editing the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References and Notes

  1. Siegel, R.; Naishadham, D.; Jemal, A. Cancer statistics, 2012. Cancer J. Clin. 2012, 62, 10–29. [Google Scholar] [CrossRef]
  2. Siegel, R.; Ma, J.; Zou, Z.; Jemal, A. Cancer statistics, 2014. Cancer J. Clin. 2014, 64, 9–29. [Google Scholar] [CrossRef]
  3. Heinlein, C.A.; Chang, C. Androgen receptor in prostate cancer. Endocr. Rev. 2004, 25, 276–308. [Google Scholar] [CrossRef] [PubMed]
  4. Denis, L.J. The role of active treatment in early prostate cancer. Radiother. Oncol. 2000, 57, 251–258. [Google Scholar] [CrossRef] [PubMed]
  5. Lieberman, R. Androgen deprivation therapy for prostate cancer chemoprevention: Current status and future directions for agent development. Urology 2001, 58, 83–90. [Google Scholar] [CrossRef] [PubMed]
  6. Molina, A.; Belldegrun, A. Novel therapeutic strategies for castration resistant prostate cancer: Inhibition of persistent androgen production and androgen receptor mediated signaling. J. Urol. 2011, 185, 787–794. [Google Scholar] [CrossRef] [PubMed]
  7. Basaria, S.; Davda, M.N.; Travison, T.G.; Ulloor, J.; Singh, R.; Bhasin, S. Risk factors associated with cardiovascular events during testosterone administration in older men with mobility limitation. J. Gerontol. Series A Biol. Sci. Med. Sci. 2013, 68, 153–160. [Google Scholar] [CrossRef]
  8. Hot flashes are a sudden wave of mild or intense body heat caused by rushes of hormonal changes resulting from decreased levels of estrogen. They can occur at any time and may last from a few seconds to half an hour. They are due to blood vessel dilation and constriction. Their frequency is increased in menopausal women and men receiving ADT.
  9. The “metabolic” or “insulin-resistance” syndrome (MetS) is a cluster of cardio-vascular (CV) risks related to insulin resistance [10,11]. Diagnosis of the syndrome is defined by the adult treatment panel III (ATP III) and the World health organization (WHO) as a combination of low HDL, increased waist circumference, increased triglycerides, increased fasting glucose and hypertension [11]. The syndrome itself is further associated with various metabolic abnormalities, including insulin resistance as well as other factors such as male hypogonadism [12].
  10. Ferrannini, E. Metabolic syndrome: A solution in search of a problem. J. Clin. Endocrinol. Metab. 2007, 92, 396–398. [Google Scholar] [CrossRef] [PubMed]
  11. Kahn, R.; Buse, J.; Ferrannini, E.; Stern, M. The metabolic syndrome: Time for a critical appraisal: Joint statement from the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2005, 28, 2289–2304. [Google Scholar] [CrossRef] [PubMed]
  12. Cho, L.W. Metabolic syndrome. Singap. Med. J. 2011, 52, 779–785. [Google Scholar]
  13. Insulin resistance is defined clinically as the inability of a known quantity of exogenous or endogenous insulin to increase glucose uptake and utilization in an individual as much as it does in a normal population [14].
  14. Lebovitz, H.E. Insulin resistance: Definition and consequences. Exp. Clin. Endocrinol. Diabetes 2001, 109, 135–148. [Google Scholar] [CrossRef] [PubMed]
  15. Cardiovascular events refer to any incidents that may cause damage to the heart muscle involving the heart and/or blood vessels which include: arterial embolic or thrombotic events, hemorrhagic or ischemic cerebrovascular conditions, myocardial infarction, and other ischemic heart disease. Severe CV events include conditions such as myocardial infarction or congestive heart failure which may be fatal [16].
  16. WHO Fact Sheet—Cardiovascular diseases (CVDs). Available online: Http://www.who.int/mediacentre/factsheets/fs317/en/ (accessed on 4 September 2014).
  17. Osteoporosis is a skeletal disorder characterized by compromised bone strength predisposing to an increased risk for bone fractures. Bone strength reflects the integration of two main features: bone mineral density (BMD) and bone quality [18]. Osteoporosis is defined by the WHO as a BMD of 2.5 standard deviations or more below the mean peak bone mass (average of young, healthy adults) as measured by dual-energy X-ray absorptiometry (DXA). A BMD value allows fracture risk to be calculated using FRAX or CAROC assessment algorithms, which incorporate a group of clinical risk factors in addition to femoral neck BMD [19,20].
  18. Kilbanski, A.; Adams-Campbell, L.; Bassford, T.; Blair, S.N.; Boden, S.D.; Dickersin, K.; Gifford, D.R.; Glasse, L.; Goldring, S.R.; Hruska, K.; et al. Osteoporosis prevention, diagnosis, and therapy. JAMA J. American Medical Association 2001, 285, 785–795. [Google Scholar] [CrossRef]
  19. Leslie, W.D.; Lix, L.M.; Johansson, H.; Oden, A.; McCloskey, E.; Kanis, J.A. Independent clinical validation of a Canadian FRAX tool: Fracture prediction and model calibration. J. Bone Miner. Res. 2010, 25, 2350–2358. [Google Scholar] [CrossRef] [PubMed]
  20. Leslie, W.D.; Tsang, J.F.; Lix, L.M. Simplified system for absolute fracture risk assessment: Clinical validation in Canadian women. J. Bone Miner. Res. 2009, 24, 353–360. [Google Scholar] [CrossRef] [PubMed]
  21. Grossmann, M.; Zajac, J.D. Androgen deprivation therapy in men with prostate cancer: How should the side effects be monitored and treated? Clin. Endocrinol. 2011, 74, 289–293. [Google Scholar]
  22. Truant, T.L.; Porcino, A.J.; Ross, B.C.; Wong, M.E.; Hilario, C.T. Complementary and alternative medicine (CAM) use in advanced cancer: A systematic review. J. Support. Oncol. 2013, 11, 105–113. [Google Scholar] [CrossRef] [PubMed]
  23. Philippou, Y.; Hadjipavlou, M.; Khan, S.; Rane, A. Complementary and alternative medicine (CAM) in prostate and bladder cancer. BJU Int. 2013, 112, 1073–1079. [Google Scholar] [CrossRef] [PubMed]
  24. Klempner, S.J.; Bubley, G. Complementary and alternative medicines in prostate cancer: From bench to bedside? The oncologist 2012, 17, 830–837. [Google Scholar] [CrossRef]
  25. Huggins, C.; Hodges, C.V. Studies on prostatic cancer: I. The effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. J. Urol. 2002, 168, 9–12. [Google Scholar] [CrossRef] [PubMed]
  26. Schroder, F.; Crawford, E.D.; Axcrona, K.; Payne, H.; Keane, T.E. Androgen deprivation therapy: Past, present and future. BJU Int. 2012, 109, 1–12. [Google Scholar] [CrossRef] [PubMed]
  27. The initial administration of an LHRH agonist first causes an increase of LH with a corresponding rise in serum T over 1–2 weeks. This increase in T may initially stimulate and worsen the disease and related symptoms. To prevent this phenomenon, administration of an anti-androgen or estrogen for one week before and during the first few weeks of LHRH agonist therapy is often employed.
  28. Rodrigues, D.N.; Butler, L.M.; Estelles, D.L.; de Bono, J.S. Molecular pathology and prostate cancer therapeutics: From biology to bedside. J. Pathol. 2014, 232, 178–184. [Google Scholar] [CrossRef] [PubMed]
  29. Kolvenbag, G.J.; Iversen, P.; Newling, D.W. Antiandrogen monotherapy: A new form of treatment for patients with prostate cancer. Urology 2001, 58, 16–23. [Google Scholar] [CrossRef] [PubMed]
  30. Grossmann, M.; Zajac, J.D. Management of side effects of androgen deprivation therapy. Endocrinol. Metab. Clin. N. Am. 2011, 40, 655–671. [Google Scholar] [CrossRef]
  31. Alibhai, S.M.; Duong-Hua, M.; Sutradhar, R.; Fleshner, N.E.; Warde, P.; Cheung, A.M.; Paszat, L.F. Impact of androgen deprivation therapy on cardiovascular disease and diabetes. J. Clin. Oncol. 2009, 27, 3452–3458. [Google Scholar] [CrossRef] [PubMed]
  32. Alibhai, S.M.; Mohamedali, H.Z.; Gulamhusein, H.; Panju, A.H.; Breunis, H.; Timilshina, N.; Fleshner, N.; Krahn, M.D.; Naglie, G.; Tannock, I.F.; et al. Changes in bone mineral density in men starting androgen deprivation therapy and the protective role of vitamin D. Osteoporos. Int. 2013, 24, 2571–2579. [Google Scholar] [CrossRef] [PubMed]
  33. Yuan, J.Q.; Xu, T.; Zhang, X.W.; Yu, L.P.; Li, Q.; Liu, S.J.; Huang, X.B.; Wang, X.F. Metabolic syndrome and androgen deprivation therapy in metabolic complications of prostate cancer patients. Chin. Med. J. 2012, 125, 3725–3729. [Google Scholar] [PubMed]
  34. Saglam, H.S.; Kose, O.; Kumsar, S.; Budak, S.; Adsan, O. Fasting blood glucose and lipid profile alterations following twelve-month androgen deprivation therapy in men with prostate cancer. Sci. World J. 2012, 2012. [Google Scholar] [CrossRef]
  35. Freedland, S.J.; Eastham, J.; Shore, N. Androgen deprivation therapy and estrogen deficiency induced adverse effects in the treatment of prostate cancer. Prostate Cancer Prostatic Dis. 2009, 12, 333–338. [Google Scholar] [CrossRef] [PubMed]
  36. Carani, C.; Qin, K.; Simoni, M.; Faustini-Fustini, M.; Serpente, S.; Boyd, J.; Korach, K.S.; Simpson, E.R. Effect of testosterone and estradiol in a man with aromatase deficiency. N Engl. J. Med. 1997, 337, 91–95. [Google Scholar] [CrossRef] [PubMed]
  37. Khosla, S. Update in male osteoporosis. J. Clin. Endocrinol. Metab. 2010, 95, 3–10. [Google Scholar] [CrossRef] [PubMed]
  38. Lee, R.J.; Saylor, P.J.; Smith, M.R. Contemporary therapeutic approaches targeting bone complications in prostate cancer. Clin. Genitourin. Cancer 2010, 8, 29–36. [Google Scholar] [CrossRef] [PubMed]
  39. Jacobson, J.S.; Troxel, A.B.; Evans, J.; Klaus, L.; Vahdat, L.; Kinne, D.; Lo, K.M.; Moore, A.; Rosenman, P.J.; Kaufman, E.L.; et al. Randomized trial of black cohosh for the treatment of hot flashes among women with a history of breast cancer. J. Clin. Oncol. 2001, 19, 2739–2745. [Google Scholar] [PubMed]
  40. Fritz, H.; Seely, D.; McGowan, J.; Skidmore, B.; Fernandes, R.; Kennedy, D.A.; Cooley, K.; Wong, R.; Sagar, S.; Balneaves, L.G.; et al. Black cohosh and breast cancer: A systematic review. Integr. Cancer Ther. 2014, 13, 12–29. [Google Scholar] [CrossRef] [PubMed]
  41. Al-Bareeq, R.J.; Ray, A.A.; Nott, L.; Pautler, S.E.; Razvi, H. Dong Quai (Angelica sinensis) in the treatment of hot flashes for men on androgen deprivation therapy: Results of a randomized double-blind placebo controlled trial. Can. Urol. Assoc. J. 2010, 4, 49–53. [Google Scholar] [PubMed]
  42. Kim, M.S.; Lim, H.J.; Yang, H.J.; Lee, M.S.; Shin, B.C.; Ernst, E. Ginseng for managing menopause symptoms: A systematic review of randomized clinical trials. J. Ginseng Res. 2013, 37, 30–36. [Google Scholar] [CrossRef] [PubMed]
  43. Moyad, M.A. Complementary/alternative therapies for reducing hot flashes in prostate cancer patients: Reevaluating the existing indirect data from studies of breast cancer and postmenopausal women. Urology 2002, 59, 20–33. [Google Scholar] [CrossRef]
  44. Zhang, X.; Shu, X.O.; Gao, Y.T.; Yang, G.; Li, Q.; Li, H.; Jin, F.; Zheng, W. Soy food consumption is associated with lower risk of coronary heart disease in Chinese women. J. Nutr. 2003, 133, 2874–2878. [Google Scholar] [PubMed]
  45. Azadbakht, L.; Kimiagar, M.; Mehrabi, Y.; Esmaillzadeh, A.; Padyab, M.; Hu, F.B.; Willett, W.C. Soy inclusion in the diet improves features of the metabolic syndrome: A randomized crossover study in postmenopausal women. Am. J. Clin. Nutr. 2007, 85, 735–741. [Google Scholar] [PubMed]
  46. Thelen, P.; Burfeind, P.; Schweyer, S.; Scharf, J.G.; Wuttke, W.; Ringert, R.H. Molecular principles of alternative treatment approaches for hormone-refractory prostate cancer. Der. Urol. Ausg. A 2007, 46, 1271–1274. [Google Scholar] [CrossRef]
  47. Napora, J.K.; Short, R.G.; Muller, D.C.; Carlson, O.D.; Odetunde, J.O.; Xu, X.; Carducci, M.; Travison, T.G.; Maggio, M.; Egan, J.M.; et al. High-dose isoflavones do not improve metabolic and inflammatory parameters in androgen-deprived men with prostate cancer. J. Androl. 2011, 32, 40–48. [Google Scholar] [CrossRef] [PubMed]
  48. Albertazzi, P.; Pansini, F.; Bonaccorsi, G.; Zanotti, L.; Forini, E.; de Aloysio, D. The effect of dietary soy supplementation on hot flushes. Obstetrics Gynecol. 1998, 91, 6–11. [Google Scholar] [CrossRef]
  49. Charles, C.; Yuskavage, J.; Carlson, O.; John, M.; Tagalicud, A.S.; Maggio, M.; Muller, D.C.; Egan, J.; Basaria, S. Effects of high-dose isoflavones on metabolic and inflammatory markers in healthy postmenopausal women. Menopause 2009, 16, 395–400. [Google Scholar] [CrossRef] [PubMed]
  50. James, M.J.; Gibson, R.A.; Cleland, L.G. Dietary polyunsaturated fatty acids and inflammatory mediator production. Am. J. Clin. Nutr. 2000, 71, 343–348. [Google Scholar]
  51. Lippman, S.M.; Klein, E.A.; Goodman, P.J.; Lucia, M.S.; Thompson, I.M.; Ford, L.G.; Parnes, H.L.; Minasian, L.M.; Gaziano, J.M.; Hartline, J.A.; et al. Effect of selenium and vitamin E on risk of prostate cancer and other cancers: The Selenium and Vitamin E Cancer Prevention Trial (SELECT). Jama 2009, 301, 39–51. [Google Scholar] [CrossRef] [PubMed]
  52. Rubenstein, B.B. Vitamin E diminishes the vasomotor symptoms of menopause. Fed. Proc. 1948, 7, 106. [Google Scholar] [PubMed]
  53. Raffy, A. Menopause and vitamin E. Ther. Hung. 1955, 4, 12–14. [Google Scholar] [PubMed]
  54. Orwoll, E.; Nielson, C.M.; Marshall, L.M.; Lambert, L.; Holton, K.F.; Hoffman, A.R.; Barrett-Connor, E.; Shikany, J.M.; Dam, T.; Cauley, J.A. Vitamin D deficiency in older men. J. Clin. Endocrinol. Metab. 2009, 94, 1214–1222. [Google Scholar] [CrossRef] [PubMed]
  55. Liu, S.; Lee, I.M.; Song, Y.; Van Denburgh, M.; Cook, N.R.; Manson, J.E.; Buring, J.E. Vitamin E and risk of type 2 diabetes in the women’s health study randomized controlled trial. Diabetes 2006, 55, 2856–2862. [Google Scholar] [CrossRef] [PubMed]
  56. Klein, E.A.; Thompson, I.M., Jr.; Tangen, C.M.; Crowley, J.J.; Lucia, M.S.; Goodman, P.J.; Minasian, L.M.; Ford, L.G.; Parnes, H.L.; Gaziano, J.M.; et al. Vitamin E and the risk of prostate cancer: The Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA 2011, 306, 1549–1556. [Google Scholar] [CrossRef] [PubMed]
  57. Sesso, H.D.; Buring, J.E.; Christen, W.G.; Kurth, T.; Belanger, C.; MacFadyen, J.; Bubes, V.; Manson, J.E.; Glynn, R.J.; Gaziano, J.M. Vitamins E and C in the prevention of cardiovascular disease in men: The Physicians’ Health Study II randomized controlled trial. JAMA 2008, 300, 2123–2133. [Google Scholar] [CrossRef] [PubMed]
  58. Peters, U.; Littman, A.J.; Kristal, A.R.; Patterson, R.E.; Potter, J.D.; White, E. Vitamin E and selenium supplementation and risk of prostate cancer in the Vitamins and lifestyle (VITAL) study cohort. Cancer Causes Control 2008, 19, 75–87. [Google Scholar] [CrossRef] [PubMed]
  59. Seidlova-Wuttke, D.; Stecher, G.; Kammann, M.; Haunschild, J.; Eder, N.; Stahnke, V.; Wessels, J.; Wuttke, W. Osteoprotective effects of Cimicifuga racemosa and its triterpene-saponins are responsible for reduction of bone marrow fat. Phytomedicine 2012, 19, 855–860. [Google Scholar] [CrossRef] [PubMed]
  60. Kuhnle, G.G.; Ward, H.A.; Vogiatzoglou, A.; Luben, R.N.; Mulligan, A.; Wareham, N.J.; Forouhi, N.G.; Khaw, K.T. Association between dietary phyto-oestrogens and bone density in men and postmenopausal women. Br. J. Nutr. 2011, 106, 1063–1069. [Google Scholar] [CrossRef] [PubMed]
  61. Soung, D.Y.; Devareddy, L.; Khalil, D.A.; Hooshmand, S.; Patade, A.; Lucas, E.A.; Arjmandi, B.H. Soy affects trabecular microarchitecture and favorably alters select bone-specific gene expressions in a male rat model of osteoporosis. Calcif. Tissue Int. 2006, 78, 385–391. [Google Scholar] [CrossRef] [PubMed]
  62. Hogstrom, M.; Nordstrom, P.; Nordstrom, A. n-3 Fatty acids are positively associated with peak bone mineral density and bone accrual in healthy men: The NO2 Study. Am. J. Clin. Nutr. 2007, 85, 803–807. [Google Scholar] [PubMed]
  63. Watkins, B.A.; Li, Y.; Lippman, H.E.; Seifert, M.F. Omega-3 polyunsaturated fatty acids and skeletal health. Exp. Biol Med. (Maywood) 2001, 226, 485–497. [Google Scholar]
  64. Fernandes, G.; Bhattacharya, A.; Rahman, M.; Zaman, K.; Banu, J. Effects of n-3 fatty acids on autoimmunity and osteoporosis. Front. Biosci. 2008, 13, 4015–4020. [Google Scholar] [CrossRef] [PubMed]
  65. Shen, C.L.; Yeh, J.K.; Rasty, J.; Chyu, M.C.; Dunn, D.M.; Li, Y.; Watkins, B.A. Improvement of bone quality in gonad-intact middle-aged male rats by long-chain n-3 polyunsaturated fatty acid. Calcif. Tissue Int. 2007, 80, 286–293. [Google Scholar] [CrossRef] [PubMed]
  66. Rahman, M.M.; Bhattacharya, A.; Banu, J.; Fernandes, G. Conjugated linoleic acid protects against age-associated bone loss in C57BL/6 female mice. J. Nutr. Biochem. 2007, 18, 467–474. [Google Scholar] [CrossRef] [PubMed]
  67. Tang, B.M.; Eslick, G.D.; Nowson, C.; Smith, C.; Bensoussan, A. Use of calcium or calcium in combination with vitamin D supplementation to prevent fractures and bone loss in people aged 50 years and older: A meta-analysis. Lancet 2007, 370, 657–666. [Google Scholar] [CrossRef] [PubMed]
  68. Gates, B.J.; Das, S. Management of osteoporosis in elderly men. Maturitas 2011, 69, 113–119. [Google Scholar] [CrossRef] [PubMed]
  69. Gielen, E.; Boonen, S.; Vanderschueren, D.; Sinnesael, M.; Verstuyf, A.; Claessens, F.; Milisen, K.; Verschueren, S. Calcium and vitamin D supplementation in men. J. Osteoporos. 2011, 2011. [Google Scholar] [CrossRef]
  70. Verbrugge, F.H.; Gielen, E.; Milisen, K.; Boonen, S. Who should receive calcium and vitamin D supplementation? Age Ageing 2012, 41, 576–580. [Google Scholar] [CrossRef]
  71. Cranney, A.; Horsley, T.; O’Donnell, S.; Weiler, H.; Puil, L.; Ooi, D.; Atkinson, S.; Ward, L.; Moher, D.; Hanley, D.; et al. Effectiveness and safety of vitamin D in relation to bone health. Evid. Rep. Technol. Assess. 2007, 1–235. [Google Scholar]
  72. Moyad, M.A. Complementary therapies for reducing the risk of osteoporosis in patients receiving luteinizing hormone-releasing hormone treatment/orchiectomy for prostate cancer: A review and assessment of the need for more research. Urology 2002, 59, 34–40. [Google Scholar] [CrossRef] [PubMed]
  73. Stabler, S.N.; Tejani, A.M.; Huynh, F.; Fowkes, C. Garlic for the prevention of cardiovascular morbidity and mortality in hypertensive patients. Cochrane Database Syst. Rev. 2012, 8. [Google Scholar] [CrossRef]
  74. Shouk, R.; Abdou, A.; Shetty, K.; Sarkar, D.; Eid, A.H. Mechanisms underlying the antihypertensive effects of garlic bioactives. Nutr Res. 2014, 34, 106–115. [Google Scholar] [CrossRef] [PubMed]
  75. Clarkson, T.B. Soy, soy phytoestrogens and cardiovascular disease. J. Nutr. 2002, 132, 566–569. [Google Scholar]
  76. Atteritano, M.; Marini, H.; Minutoli, L.; Polito, F.; Bitto, A.; Altavilla, D.; Mazzaferro, S.; D’Anna, R.; Cannata, M.L.; Gaudio, A.; et al. Effects of the phytoestrogen genistein on some predictors of cardiovascular risk in osteopenic, postmenopausal women: A two-year randomized, double-blind, placebo-controlled study. J. Clin. Endocrinol. Metab. 2007, 92, 3068–3075. [Google Scholar] [CrossRef] [PubMed]
  77. Bloedon, L.T.; Balikai, S.; Chittams, J.; Cunnane, S.C.; Berlin, J.A.; Rader, D.J.; Szapary, P.O. Flaxseed and cardiovascular risk factors: Results from a double blind, randomized, controlled clinical trial. J. Am. Coll. Nutr. 2008, 27, 65–74. [Google Scholar] [CrossRef] [PubMed]
  78. Jacobson, T.A. Beyond lipids: The role of omega-3 fatty acids from fish oil in the prevention of coronary heart disease. Curr. Atheroscler. Rep. 2007, 9, 145–153. [Google Scholar] [CrossRef] [PubMed]
  79. Kris-Etherton, P.M.; Harris, W.S.; Appel, L.J. Fish consumption, fish oil, omega-3 fatty acids, and cardiovascular disease. Circulation 2002, 106, 2747–2757. [Google Scholar] [CrossRef] [PubMed]
  80. Simopoulos, A.P. The importance of the ratio of omega-6/omega-3 essential fatty acids. Biomed. Pharmacother. 2002, 56, 365–379. [Google Scholar] [CrossRef] [PubMed]
  81. Hjerkinn, E.M.; Seljeflot, I.; Ellingsen, I.; Berstad, P.; Hjermann, I.; Sandvik, L.; Arnesen, H. Influence of long-term intervention with dietary counseling, long-chain n-3 fatty acid supplements, or both on circulating markers of endothelial activation in men with long-standing hyperlipidemia. Am. J. Clin. Nutr. 2005, 81, 583–589. [Google Scholar] [PubMed]
  82. Dewell, A.; Marvasti, F.F.; Harris, W.S.; Tsao, P.; Gardner, C.D. Low- and high-dose plant and marine (n-3) fatty acids do not affect plasma inflammatory markers in adults with metabolic syndrome. J. Nutr. 2011, 141, 2166–2171. [Google Scholar] [CrossRef] [PubMed]
  83. Koyama, H.; Mutakin; Abdulah, R.; Yamazaki, C.; Kameo, S. Selenium supplementation trials for cancer prevention and the subsequent risk of type 2 diabetes mellitus. Nihon Eiseigaku Zasshi 2013, 68, 1–10. [Google Scholar] [CrossRef] [PubMed]
  84. Rayman, M.P.; Blundell-Pound, G.; Pastor-Barriuso, R.; Guallar, E.; Steinbrenner, H.; Stranges, S. A randomized trial of selenium supplementation and risk of type-2 diabetes, as assessed by plasma adiponectin. PLoS One 2012, 7, 45269. [Google Scholar] [CrossRef]
  85. Faghihi, T.; Radfar, M.; Barmal, M.; Amini, P.; Qorbani, M.; Abdollahi, M.; Larijani, B. A randomized, placebo-controlled trial of selenium supplementation in patients with type 2 diabetes: Effects on glucose homeostasis, oxidative stress, and lipid profile. Am. J. Ther. 2013. [Google Scholar] [CrossRef]
  86. Dennehy, C.; Tsourounis, C. A review of select vitamins and minerals used by postmenopausal women. Maturitas 2010, 66, 370–380. [Google Scholar] [CrossRef] [PubMed]
  87. Algotar, A.M.; Stratton, M.S.; Stratton, S.P.; Hsu, C.H.; Ahmann, F.R. No effect of selenium supplementation on serum glucose levels in men with prostate cancer. Am. J. Med. 2010, 123, 765–768. [Google Scholar] [CrossRef] [PubMed]
  88. Kristal, A.R.; Darke, A.K.; Morris, J.S.; Tangen, C.M.; Goodman, P.J.; Thompson, I.M.; Meyskens, F.L., Jr.; Goodman, G.E.; Minasian, L.M.; Parnes, H.L.; et al. Baseline selenium status and effects of selenium and vitamin E supplementation on prostate cancer risk. J. Nat. Cancer Inst. 2014, 106. [Google Scholar] [CrossRef] [PubMed]
  89. Algotar, A.M.; Stratton, M.S.; Ahmann, F.R.; Ranger-Moore, J.; Nagle, R.B.; Thompson, P.A.; Slate, E.; Hsu, C.H.; Dalkin, B.L.; Sindhwani, P.; et al. Phase 3 clinical trial investigating the effect of selenium supplementation in men at high-risk for prostate cancer. Prostate 2013, 73, 328–335. [Google Scholar] [CrossRef] [PubMed]
  90. b>Algotar, A.M.; Hsu, C.H.; Singh, P.; Stratton, S.P. Selenium supplementation has no effect on serum glucose levels in men at high risk for Prostate Cancer. J. Diabetes 2013, 5, 465–470. [Google Scholar] [CrossRef] [PubMed]
  91. Cook, N.R.; Albert, C.M.; Gaziano, J.M.; Zaharris, E.; MacFadyen, J.; Danielson, E.; Buring, J.E.; Manson, J.E. A randomized factorial trial of vitamins C and E and beta carotene in the secondary prevention of cardiovascular events in women: Results from the Women’s Antioxidant Cardiovascular Study. Arch. Internal Med. 2007, 167, 1610–1618. [Google Scholar] [CrossRef]
  92. Saremi, A.; Arora, R. Vitamin E and cardiovascular disease. Am. J. Ther. 2010, 17, 56–65. [Google Scholar] [CrossRef]
  93. Shahani, S.; Braga-Basaria, M.; Basaria, S. Androgen deprivation therapy in prostate cancer and metabolic risk for atherosclerosis. J. Clin. Endocrinol. Metab. 2008, 93, 2042–2049. [Google Scholar] [CrossRef] [PubMed]
  94. Smith, M.R.; Lee, H.; McGovern, F.; Fallon, M.A.; Goode, M.; Zietman, A.L.; Finkelstein, J.S. Metabolic changes during gonadotropin-releasing hormone agonist therapy for prostate cancer: Differences from the classic metabolic syndrome. Cancer 2008, 112, 2188–2194. [Google Scholar] [CrossRef] [PubMed]
  95. Braga-Basaria, M.; Dobs, A.S.; Muller, D.C.; Carducci, M.A.; John, M.; Egan, J.; Basaria, S. Metabolic syndrome in men with prostate cancer undergoing long-term androgen-deprivation therapy. J. Clin. Oncol. 2006, 24, 3979–3983. [Google Scholar] [CrossRef] [PubMed]
  96. Basaria, S.; Muller, D.C.; Carducci, M.A.; Egan, J.; Dobs, A.S. Hyperglycemia and insulin resistance in men with prostate carcinoma who receive androgen-deprivation therapy. Cancer 2006, 106, 581–588. [Google Scholar] [CrossRef] [PubMed]
  97. Laaksonen, D.E.; Niskanen, L.; Punnonen, K.; Nyyssonen, K.; Tuomainen, T.P.; Valkonen, V.P.; Salonen, R.; Salonen, J.T. Testosterone and sex hormone-binding globulin predict the metabolic syndrome and diabetes in middle-aged men. Diabetes Care 2004, 27, 1036–1041. [Google Scholar] [CrossRef] [PubMed]
  98. Tirabassi, G.; Gioia, A.; Giovannini, L.; Boscaro, M.; Corona, G.; Carpi, A.; Maggi, M.; Balercia, G. Testosterone and cardiovascular risk. Intern. Emerg. Med. 2013, 8, 65–69. [Google Scholar] [CrossRef] [PubMed]
  99. Stellato, R.K.; Feldman, H.A.; Hamdy, O.; Horton, E.S.; McKinlay, J.B. Testosterone, sex hormone-binding globulin, and the development of type 2 diabetes in middle-aged men: Prospective results from the Massachusetts male aging study. Diabetes Care 2000, 23, 490–494. [Google Scholar] [CrossRef] [PubMed]
  100. Eckel, R.H.; Alberti, K.G.; Grundy, S.M.; Zimmet, P.Z. The metabolic syndrome. Lancet 2010, 375, 181–183. [Google Scholar] [CrossRef] [PubMed]
  101. Fink, R.I.; Kolterman, O.G.; Griffin, J.; Olefsky, J.M. Mechanisms of insulin resistance in aging. J. Clin. Investig. 1983, 71, 1523–1535. [Google Scholar] [CrossRef] [PubMed]
  102. Lu-Yao, G.; Stukel, T.A.; Yao, S.L. Changing patterns in competing causes of death in men with prostate cancer: A population based study. J. Urol. 2004, 171, 2285–2290. [Google Scholar] [CrossRef] [PubMed]
  103. WHO Assessment of fracture risk and its application to screening for postmenopausal osteoporosis. Report of a WHO Study Group. World Health Organ. Technical Rep. Ser. 1994, 843, 1–129.
  104. Lee, R.J.; Saylor, P.J.; Smith, M.R. Treatment and prevention of bone complications from prostate cancer. Bone 2011, 48, 88–95. [Google Scholar] [CrossRef] [PubMed]
  105. Krupski, T.L.; Smith, M.R.; Lee, W.C.; Pashos, C.L.; Brandman, J.; Wang, Q.; Botteman, M.; Litwin, M.S. Natural history of bone complications in men with prostate carcinoma initiating androgen deprivation therapy. Cancer 2004, 101, 541–549. [Google Scholar] [CrossRef] [PubMed]
  106. Shahinian, V.B.; Kuo, Y.F.; Freeman, J.L.; Goodwin, J.S. Risk of fracture after androgen deprivation for prostate cancer. N. Engl. J. Med. 2005, 352, 154–164. [Google Scholar] [CrossRef] [PubMed]
  107. Oefelein, M.G.; Ricchiuti, V.; Conrad, W.; Resnick, M.I. Skeletal fractures negatively correlate with overall survival in men with prostate cancer. J. Urol. 2002, 168, 1005–1007. [Google Scholar] [CrossRef] [PubMed]
  108. Kiratli, B.J.; Srinivas, S.; Perkash, I.; Terris, M.K. Progressive decrease in bone density over 10 years of androgen deprivation therapy in patients with prostate cancer. Urology 2001, 57, 127–132. [Google Scholar] [CrossRef] [PubMed]
  109. Greenspan, S.L.; Coates, P.; Sereika, S.M.; Nelson, J.B.; Trump, D.L.; Resnick, N.M. Bone loss after initiation of androgen deprivation therapy in patients with prostate cancer. J. Clin. Endocrinol. Metab. 2005, 90, 6410–6417. [Google Scholar] [CrossRef] [PubMed]
  110. Morote, J.; Orsola, A.; Abascal, J.M.; Planas, J.; Trilla, E.; Raventos, C.X.; Cecchini, L.; Encabo, G.; Reventos, J. Bone mineral density changes in patients with prostate cancer during the first 2 years of androgen suppression. J. Urol. 2006, 175, 1679–1683. [Google Scholar] [CrossRef] [PubMed]
  111. Wadhwa, V.K.; Weston, R.; Mistry, R.; Parr, N.J. Long-term changes in bone mineral density and predicted fracture risk in patients receiving androgen-deprivation therapy for prostate cancer, with stratification of treatment based on presenting values. BJU Int. 2009, 104, 800–805. [Google Scholar] [CrossRef] [PubMed]
  112. Lloyd, K.B.; Hornsby, L.B. Complementary and alternative medications for women’s health issues. Nutr. Clin. Prac. 2009, 24, 589–608. [Google Scholar] [CrossRef]
  113. Dennehy, C.E. The use of herbs and dietary supplements in gynecology: An evidence-based review. J. Midwifery Women’s Health 2006, 51, 402–409. [Google Scholar] [CrossRef]
  114. Seidlova-Wuttke, D.; Eder, N.; Stahnke, V.; Kammann, M.; Stecher, G.; Haunschild, J.; Wessels, J.T.; Wuttke, W. Cimicifuga racemosa and its triterpene-saponins prevent the Metabolic Syndrome and deterioration of cartilage in the knee joint of ovariectomized rats by similar mechanisms. Phytomedicine 2012, 19, 846–853. [Google Scholar] [CrossRef]
  115. Mahon, S.M.; Kaplan, M. Placebo effect in hot flush research. Lancet Oncol. 2012, 13, 188. [Google Scholar] [CrossRef]
  116. Wuttke, W.; Gorkow, C.; Seidlova-Wuttke, D. Effects of black cohosh (Cimicifuga racemosa) on bone turnover, vaginal mucosa, and various blood parameters in postmenopausal women: A double-blind, placebo-controlled, and conjugated estrogens-controlled study. Menopause 2006, 13, 185–196. [Google Scholar] [CrossRef]
  117. Seidlova-Wuttke, D.; Jarry, H.; Pitzel, L.; Wuttke, W. Effects of estradiol-17beta, testosterone and a black cohosh preparation on bone and prostate in orchidectomized rats. Maturitas 2005, 51, 177–186. [Google Scholar] [CrossRef]
  118. Dueregger, A.; Guggenberger, F.; Barthelmes, J.; Stecher, G.; Schuh, M.; Intelmann, D.; Abel, G.; Haunschild, J.; Klocker, H.; Ramoner, R.; et al. Attenuation of nucleoside and anti-cancer nucleoside analog drug uptake in prostate cancer cells by Cimicifuga racemosa extract BNO-1055. Phytomedicine 2013, 20, 1306–1314. [Google Scholar] [CrossRef]
  119. Molina-Arcas, M.; Casado, F.J.; Pastor-Anglada, M. Nucleoside transporter proteins. Curr. Vasc. Pharmacol. 2009, 7, 426–434. [Google Scholar] [CrossRef]
  120. Cheema, D.; Coomarasamy, A.; El-Toukhy, T. Non-hormonal therapy of post-menopausal vasomotor symptoms: A structured evidence-based review. Arch. Gynecol. Obstet. 2007, 276, 463–469. [Google Scholar] [CrossRef]
  121. Wiklund, I.K.; Mattsson, L.A.; Lindgren, R.; Limoni, C. Effects of a standardized ginseng extract on quality of life and physiological parameters in symptomatic postmenopausal women: A double-blind, placebo-controlled trial. Swedish Alternative Medicine Group. Int. J. Clin. Pharmacol. Res. 1999, 19, 89–99. [Google Scholar]
  122. Nag, S.A.; Qin, J.J.; Wang, W.; Wang, M.H.; Wang, H.; Zhang, R. Ginsenosides as Anticancer Agents: In vitro and in vivo Activities, Structure-Activity Relationships, and Molecular Mechanisms of Action. Front. Pharmacol. 2012, 3, 25. [Google Scholar] [CrossRef]
  123. Wang, Z.; Wang, J.; Chan, P. Treating type 2 diabetes mellitus with traditional chinese and Indian medicinal herbs. Evid. Based Complement. Altern. Med. 2013, 2013. [Google Scholar] [CrossRef]
  124. Rahman, K.; Lowe, G.M. Garlic and cardiovascular disease: A critical review. J. Nutr. 2006, 136, 736–740. [Google Scholar]
  125. Rabito, M.J.; Kaye, A.D. Complementary and alternative medicine and cardiovascular disease: An evidence-based review. Evid. Based Complement. Altern. Med. 2013, 2013. [Google Scholar] [CrossRef]
  126. Smith, M.R.; Malkowicz, S.B.; Chu, F.; Forrest, J.; Sieber, P.; Barnette, K.G.; Rodriquez, D.; Steiner, M.S. Toremifene improves lipid profiles in men receiving androgen-deprivation therapy for prostate cancer: Interim analysis of a multicenter phase III study. J. Clin. Oncol. 2008, 26, 1824–1829. [Google Scholar] [CrossRef]
  127. Smith, M.R.; Morton, R.A.; Barnette, K.G.; Sieber, P.R.; Malkowicz, S.B.; Rodriguez, D.; Hancock, M.L.; Steiner, M.S. Toremifene to reduce fracture risk in men receiving androgen deprivation therapy for prostate cancer. J. Urol. 2013, 189, 45–50. [Google Scholar] [CrossRef]
  128. Kearns, A.E.; Northfelt, D.W.; Dueck, A.C.; Atherton, P.J.; Dakhil, S.R.; Rowland, K.M., Jr.; Fuloria, J.; Flynn, P.J.; Dentchev, T.; Loprinzi, C.L. Osteoporosis prevention in prostate cancer patients receiving androgen ablation therapy: Placebo-controlled double-blind study of estradiol and risedronate: N01C8. Support. Care Cancer 2010, 18, 321–328. [Google Scholar] [CrossRef]
  129. Wibowo, E.; Schellhammer, P.; Wassersug, R.J. Role of estrogen in normal male function: Clinical implications for patients with prostate cancer on androgen deprivation therapy. J. Urol. 2011, 185, 17–23. [Google Scholar] [CrossRef]
  130. Treatment and survival of patients with cancer of the prostate. The Veterans Administration Co-operative Urological Research Group. Surg. Gynecol. Obstet. 1967, 124, 1011–1017.
  131. Byar, D.P.; Corle, D.K. Hormone therapy for prostate cancer: Results of the Veterans Administration Cooperative Urological Research Group studies. NCI Monographs 1987, 7, 165–170. [Google Scholar]
  132. Murkies, A.L.; Wilcox, G.; Davis, S.R. Clinical review 92: Phytoestrogens. J. Clin. Endocrinol. Metab. 1998, 83, 297–303. [Google Scholar]
  133. Turner, J.V.; Agatonovic-Kustrin, S.; Glass, B.D. Molecular aspects of phytoestrogen selective binding at estrogen receptors. J. Pharm. Sci. 2007, 96, 1879–1885. [Google Scholar] [CrossRef]
  134. Dahlman-Wright, K.; Cavailles, V.; Fuqua, S.A.; Jordan, V.C.; Katzenellenbogen, J.A.; Korach, K.S.; Maggi, A.; Muramatsu, M.; Parker, M.G.; Gustafsson, J.A. International Union of Pharmacology. LXIV. Estrogen receptors. Pharmacol. Rev. 2006, 58, 773–781. [Google Scholar] [CrossRef]
  135. Khosla, S.; Melton, L.J.; Riggs, B.L. The unitary model for estrogen deficiency and the pathogenesis of osteoporosis: Is a revision needed? J. Bone Miner. Res. 2011, 26, 441–451. [Google Scholar] [CrossRef]
  136. De Kleijn, M.J.; van der Schouw, Y.T.; Wilson, P.W.; Adlercreutz, H.; Mazur, W.; Grobbee, D.E.; Jacques, P.F. Intake of dietary phytoestrogens is low in postmenopausal women in the United States: The Framingham study(1–4). J. Nutr. 2001, 131, 1826–1832. [Google Scholar]
  137. Jemal, A.; Siegel, R.; Ward, E.; Hao, Y.; Xu, J.; Murray, T.; Thun, M.J. Cancer statistics, 2008. Cancer J. Clin. 2008, 58, 71–96. [Google Scholar] [CrossRef]
  138. Villegas, R.; Gao, Y.T.; Yang, G.; Li, H.L.; Elasy, T.A.; Zheng, W.; Shu, X.O. Legume and soy food intake and the incidence of type 2 diabetes in the Shanghai Women’s Health Study. Am. J. Clin. Nutr. 2008, 87, 162–167. [Google Scholar]
  139. Huang, Y.; Cao, S.; Nagamani, M.; Anderson, K.E.; Grady, J.J.; Lu, L.J. Decreased circulating levels of tumor necrosis factor-alpha in postmenopausal women during consumption of soy-containing isoflavones. J. Clin. Endocrinol. Metab. 2005, 90, 3956–3962. [Google Scholar] [CrossRef]
  140. Sharma, P.; Wisniewski, A.; Braga-Basaria, M.; Xu, X.; Yep, M.; Denmeade, S.; Dobs, A.S.; DeWeese, T.; Carducci, M.; Basaria, S. Lack of an effect of high dose isoflavones in men with prostate cancer undergoing androgen deprivation therapy. J. Urol. 2009, 182, 2265–2272. [Google Scholar] [CrossRef]
  141. Fairweather-Tait, S.J.; Bao, Y.; Broadley, M.R.; Collings, R.; Ford, D.; Hesketh, J.E.; Hurst, R. Selenium in human health and disease. Antioxid. Redox signal. 2011, 14, 1337–1383. [Google Scholar] [CrossRef]
  142. Steinbrenner, H.; Sies, H. Protection against reactive oxygen species by selenoproteins. Biochim. Biophys. Acta 2009, 1790, 1478–1485. [Google Scholar] [CrossRef]
  143. Holvoet, P. Relations between metabolic syndrome, oxidative stress and inflammation and cardiovascular disease. Verhandelingen 2008, 70, 193–219. [Google Scholar]
  144. Yang, M.; Sytkowski, A.J. Differential expression and androgen regulation of the human selenium-binding protein gene hSP56 in prostate cancer cells. Cancer Res. 1998, 58, 3150–3153. [Google Scholar]
  145. Clark, L.C.; Dalkin, B.; Krongrad, A.; Combs G.F., Jr.; Turnbull, B.W.; Slate, E.H.; Witherington, R.; Herlong, J.H.; Janosko, E.; Carpenter, D.; et al. Decreased incidence of prostate cancer with selenium supplementation: Results of a double-blind cancer prevention trial. Br. J. Urol. 1998, 81, 730–734. [Google Scholar] [CrossRef]
  146. Yoshizawa, K.; Willett, W.C.; Morris, S.J.; Stampfer, M.J.; Spiegelman, D.; Rimm, E.B.; Giovannucci, E. Study of prediagnostic selenium level in toenails and the risk of advanced prostate cancer. J. Nat. Cancer Inst. 1998, 90, 1219–1224. [Google Scholar] [CrossRef]
  147. Duffield-Lillico, A.J.; Dalkin, B.L.; Reid, M.E.; Turnbull, B.W.; Slate, E.H.; Jacobs, E.T.; Marshall, J.R.; Clark, L.C. Selenium supplementation, baseline plasma selenium status and incidence of prostate cancer: An analysis of the complete treatment period of the Nutritional Prevention of Cancer Trial. BJU Int. 2003, 91, 608–612. [Google Scholar] [CrossRef]
  148. Rayman, M.P.; Stranges, S. Epidemiology of selenium and type 2 diabetes: Can we make sense of it? Free Radic. Biol. Med. 2013, 65, 1557–1564. [Google Scholar]
  149. Savini, I.; Catani, M.V.; Evangelista, D.; Gasperi, V.; Avigliano, L. Obesity-associated oxidative stress: Strategies finalized to improve redox state. Int J. Mol. Sci. 2013, 14, 10497–10538. [Google Scholar] [CrossRef]
  150. Hassanzadeh, M.; Faridhosseini, R.; Mahini, M.; Faridhosseini, F.; Ranjbar, A. Serum Levels of TNF-, IL-6, and Selenium in Patients with Acute and Chronic Coronary Artery Disease. Iran. J. Immunol. 2006, 3, 142–145. [Google Scholar]
  151. Zhang, Y.; Chen, X. Reducing selenoprotein P expression suppresses adipocyte differentiation as a result of increased preadipocyte inflammation. Am. J. Physiol. Endocrinol. Metab. 2011, 300, 77–85. [Google Scholar] [CrossRef]
  152. Steinbrenner, H.; Speckmann, B.; Pinto, A.; Sies, H. High selenium intake and increased diabetes risk: Experimental evidence for interplay between selenium and carbohydrate metabolism. J. Clin. Biochem. Nutr. 2011, 48, 40–45. [Google Scholar] [CrossRef]
  153. Steinbrenner, H. Interference of selenium and selenoproteins with the insulin-regulated carbohydrate and lipid metabolism. Free Radic. Biol. Med. 2013, 65, 1538–1547. [Google Scholar] [CrossRef]
  154. Quiner, T.E.; Nakken, H.L.; Mason, B.A.; Lephart, E.D.; Hancock, C.R.; Christensen, M.J. Soy content of basal diets determines the effects of supplemental selenium in male mice. J. Nutr. 2011, 141, 2159–2165. [Google Scholar] [CrossRef]
  155. Simopoulos, A.P. Essential fatty acids in health and chronic disease. Am. J. Clin. Nutr. 1999, 70, 560–569. [Google Scholar]
  156. Nettleton, J.A. Omega-3 Fatty Acids and Health; Springer US: New York, NY,  USA, 1995; pp. 64–76. [Google Scholar]
  157. Heinze, V.M.; Actis, A.B. Dietary conjugated linoleic acid and long-chain n-3 fatty acids in mammary and prostate cancer protection: A review. Int. J. Food Sci. Nutr. 2012, 63, 66–78. [Google Scholar] [CrossRef]
  158. Tziomalos, K.; Athyros, V.G.; Mikhailidis, D.P. Fish oils and vascular disease prevention: An update. Curr. Med. Chem. 2007, 14, 2622–2628. [Google Scholar] [CrossRef]
  159. Carpentier, Y.A.; Portois, L.; Malaisse, W.J. n-3 fatty acids and the metabolic syndrome. Am. J. Clin. Nutr. 2006, 83, 1499–1504. [Google Scholar]
  160. Barre, D.E. The role of consumption of alpha-linolenic, eicosapentaenoic and docosahexaenoic acids in human metabolic syndrome and type 2 diabetes--a mini-review. J. Oleo Sci. 2007, 56, 319–325. [Google Scholar] [CrossRef]
  161. Chen, Y.Q.; Edwards, I.J.; Kridel, S.J.; Thornburg, T.; Berquin, I.M. Dietary fat-gene interactions in cancer. Cancer Metastasis Rev. 2007, 26, 535–551. [Google Scholar] [CrossRef]
  162. Ballantyne, C.M.; Bays, H.E.; Kastelein, J.J.; Stein, E.; Isaacsohn, J.L.; Braeckman, R.A.; Soni, P.N. Efficacy and safety of eicosapentaenoic acid ethyl ester (AMR101) therapy in statin-treated patients with persistent high triglycerides (from the ANCHOR study). Am. J. Cardiol. 2012, 110, 984–992. [Google Scholar] [CrossRef]
  163. Bays, H.E.; Ballantyne, C.M.; Kastelein, J.J.; Isaacsohn, J.L.; Braeckman, R.A.; Soni, P.N. Eicosapentaenoic acid ethyl ester (AMR101) therapy in patients with very high triglyceride levels (from the Multi-center, plAcebo-controlled, Randomized, double-blINd, 12-week study with an open-label Extension [MARINE] trial). Am. J. Cardiol. 2011, 108, 682–690. [Google Scholar] [CrossRef]
  164. Connor, W.E. Importance of n-3 fatty acids in health and disease. Am. J. Clin. Nutr. 2000, 71, 171–175. [Google Scholar]
  165. Wang, S.; Wu, J.; Suburu, J.; Gu, Z.; Cai, J.; Axanova, L.S.; Cramer, S.D.; Thomas, M.J.; Perry, D.L.; Edwards, I.J.; et al. Effect of dietary polyunsaturated fatty acids on castration-resistant Pten-null prostate cancer. Carcinogenesis 2012, 33, 404–412. [Google Scholar] [CrossRef]
  166. Friedrichs, W.; Ruparel, S.B.; Marciniak, R.A.; deGraffenried, L. Omega-3 fatty acid inhibition of prostate cancer progression to hormone independence is associated with suppression of mTOR signaling and androgen receptor expression. Nutr. Cancer 2011, 63, 771–777. [Google Scholar] [CrossRef]
  167. Cavazos, D.A.; Price, R.S.; Apte, S.S.; deGraffenried, L.A. Docosahexaenoic acid selectively induces human prostate cancer cell sensitivity to oxidative stress through modulation of NF-kappaB. Prostate 2011, 71, 1420–1428. [Google Scholar] [CrossRef]
  168. McEntee, M.F.; Ziegler, C.; Reel, D.; Tomer, K.; Shoieb, A.; Ray, M.; Li, X.; Neilsen, N.; Lih, F.B.; O’Rourke, D.; Whelan, J. Dietary n-3 polyunsaturated fatty acids enhance hormone ablation therapy in androgen-dependent prostate cancer. Am. J. Pathol. 2008, 173, 229–241. [Google Scholar] [CrossRef]
  169. Epstein, M.M.; Kasperzyk, J.L.; Mucci, L.A.; Giovannucci, E.; Price, A.; Wolk, A.; Hakansson, N.; Fall, K.; Andersson, S.O.; Andren, O. Dietary fatty acid intake and prostate cancer survival in Orebro County, Sweden. Am. J. Epidemiol. 2012, 176, 240–252. [Google Scholar] [CrossRef]
  170. Aronson, W.J.; Kobayashi, N.; Barnard, R.J.; Henning, S.; Huang, M.; Jardack, P.M.; Liu, B.; Gray, A.; Wan, J.; Konijeti, R.; et al. Phase II prospective randomized trial of a low-fat diet with fish oil supplementation in men undergoing radical prostatectomy. Cancer Prev Res. (Phila) 2011, 4, 2062–2071. [Google Scholar] [CrossRef]
  171. Fradet, V.; Cheng, I.; Casey, G.; Witte, J.S. Dietary omega-3 fatty acids, cyclooxygenase-2 genetic variation, and aggressive prostate cancer risk. Clin. Cancer Res. 2009, 15, 2559–2566. [Google Scholar] [CrossRef]
  172. Berquin, I.M.; Edwards, I.J.; Chen, Y.Q. Multi-targeted therapy of cancer by omega-3 fatty acids. Cancer Lett. 2008, 269, 363–377. [Google Scholar] [CrossRef]
  173. Szymanski, K.M.; Wheeler, D.C.; Mucci, L.A. Fish consumption and prostate cancer risk: A review and meta-analysis. Am. J. Clin. Nutr. 2010, 92, 1223–1233. [Google Scholar] [CrossRef]
  174. Nelson, T.L.; Stevens, J.R.; Hickey, M.S. Inflammatory markers are not altered by an eight week dietary alpha-linolenic acid intervention in healthy abdominally obese adult males and females. Cytokine 2007, 38, 101–106. [Google Scholar] [CrossRef]
  175. Thies, F.; Miles, E.A.; Nebe-von-Caron, G.; Powell, J.R.; Hurst, T.L.; Newsholme, E.A.; Calder, P.C. Influence of dietary supplementation with long-chain n-3 or n-6 polyunsaturated fatty acids on blood inflammatory cell populations and functions and on plasma soluble adhesion molecules in healthy adults. Lipids 2001, 36, 1183–1193. [Google Scholar] [CrossRef]
  176. Zhao, G.; Etherton, T.D.; Martin, K.R.; West, S.G.; Gillies, P.J.; Kris-Etherton, P.M. Dietary alpha-linolenic acid reduces inflammatory and lipid cardiovascular risk factors in hypercholesterolemic men and women. J. Nutr. 2004, 134, 2991–2997. [Google Scholar]
  177. Pot, G.K.; Geelen, A.; Majsak-Newman, G.; Harvey, L.J.; Nagengast, F.M.; Witteman, B.J.; van de Meeberg, P.C.; Hart, A.R.; Schaafsma, G.; Lund, E.K.; et al. Increased consumption of fatty and lean fish reduces serum C-reactive protein concentrations but not inflammation markers in feces and in colonic biopsies. J. Nutr. 2010, 140, 371–376. [Google Scholar] [CrossRef]
  178. Micallef, M.A.; Garg, M.L. Anti-inflammatory and cardioprotective effects of n-3 polyunsaturated fatty acids and plant sterols in hyperlipidemic individuals. Atherosclerosis 2009, 204, 476–482. [Google Scholar] [CrossRef]
  179. Kelley, D.S.; Siegel, D.; Fedor, D.M.; Adkins, Y.; Mackey, B.E. DHA supplementation decreases serum C-reactive protein and other markers of inflammation in hypertriglyceridemic men. J. Nutr. 2009, 139, 495–501. [Google Scholar] [CrossRef]
  180. Geelen, A.; Brouwer, I.A.; Schouten, E.G.; Kluft, C.; Katan, M.B.; Zock, P.L. Intake of n-3 fatty acids from fish does not lower serum concentrations of C-reactive protein in healthy subjects. Eur. J. Clin. Nutr. 2004, 58, 1440–1442. [Google Scholar] [CrossRef]
  181. Vega-Lopez, S.; Kaul, N.; Devaraj, S.; Cai, R.Y.; German, B.; Jialal, I. Supplementation with omega3 polyunsaturated fatty acids and all-rac alpha-tocopherol alone and in combination failed to exert an anti-inflammatory effect in human volunteers. Metabolism 2004, 53, 236–240. [Google Scholar] [CrossRef]
  182. Damsgaard, C.T.; Frokiaer, H.; Andersen, A.D.; Lauritzen, L. Fish oil in combination with high or low intakes of linoleic acid lowers plasma triacylglycerols but does not affect other cardiovascular risk markers in healthy men. J. Nutr. 2008, 138, 1061–1066. [Google Scholar]
  183. Yusof, H.M.; Miles, E.A.; Calder, P. Influence of very long-chain n-3 fatty acids on plasma markers of inflammation in middle-aged men. Prostaglandins Leukot Essent Fatty Acids 2008, 78, 219–228. [Google Scholar] [CrossRef]
  184. Pot, G.K.; Brouwer, I.A.; Enneman, A.; Rijkers, G.T.; Kampman, E.; Geelen, A. No effect of fish oil supplementation on serum inflammatory markers and their interrelationships: A randomized controlled trial in healthy, middle-aged individuals. Eur. J. Clin. Nutr. 2009, 63, 1353–1359. [Google Scholar] [CrossRef]
  185. Fujioka, S.; Hamazaki, K.; Itomura, M.; Huan, M.; Nishizawa, H.; Sawazaki, S.; Kitajima, I.; Hamazaki, T. The effects of eicosapentaenoic acid-fortified food on inflammatory markers in healthy subjects—A randomized, placebo-controlled, double-blind study. J. Nutr. Sci. Vitaminol. 2006, 52, 261–265. [Google Scholar] [CrossRef]
  186. Carayol, M.; Grosclaude, P.; Delpierre, C. Prospective studies of dietary alpha-linolenic acid intake and prostate cancer risk: A meta-analysis. Cancer Causes Control 2010, 21, 347–355. [Google Scholar] [CrossRef]
  187. Ma, R.W.; Chapman, K. A systematic review of the effect of diet in prostate cancer prevention and treatment. J. Hum. Nutr Diet. 2009, 22, 187–199. [Google Scholar] [CrossRef]
  188. Chua, M.E.; Sio, M.C.; Sorongon, M.C.; Dy, J.S. Relationship of dietary intake of omega-3 and omega-6 Fatty acids with risk of prostate cancer development: A meta-analysis of prospective studies and review of literature. Prostate Cancer 2012, 2012. [Google Scholar] [CrossRef]
  189. Chua, M.E.; Sio, M.C.; Sorongon, M.C.; Morales, M.L., Jr. The relevance of serum levels of long chain omega-3 polyunsaturated fatty acids and prostate cancer risk: A meta-analysis. Can.Urol. Assoc. J. 2013, 7, 333–343. [Google Scholar]
  190. Larsson, S.C.; Kumlin, M.; Ingelman-Sundberg, M.; Wolk, A. Dietary long-chain n-3 fatty acids for the prevention of cancer: A review of potential mechanisms. Am. J. Clin. Nutr. 2004, 79, 935–945. [Google Scholar]
  191. Ngo, T.H.; Barnard, R.J.; Cohen, P.; Freedland, S.; Tran, C.; deGregorio, F.; Elshimali, Y.I.; Heber, D.; Aronson, W.J. Effect of isocaloric low-fat diet on human LAPC-4 prostate cancer xenografts in severe combined immunodeficient mice and the insulin-like growth factor axis. Clin. Cancer Res. 2003, 9, 2734–2743. [Google Scholar]
  192. Kobayashi, N.; Barnard, R.J.; Said, J.; Hong-Gonzalez, J.; Corman, D.M.; Ku, M.; Doan, N.B.; Gui, D.; Elashoff, D.; Cohen, P.; et al. Effect of low-fat diet on development of prostate cancer and Akt phosphorylation in the Hi-Myc transgenic mouse model. Cancer Res. 2008, 68, 3066–3073. [Google Scholar] [CrossRef]
  193. Berquin, I.M.; Min, Y.; Wu, R.; Wu, J.; Perry, D.; Cline, J.M.; Thomas, M.J.; Thornburg, T.; Kulik, G.; Smith, A.; et al. Modulation of prostate cancer genetic risk by omega-3 and omega-6 fatty acids. J. Clin. Investig. 2007, 117, 1866–1875. [Google Scholar] [CrossRef]
  194. De Marzo, A.M.; Platz, E.A.; Sutcliffe, S.; Xu, J.; Gronberg, H.; Drake, C.G.; Nakai, Y.; Isaacs, W.B.; Nelson, W.G. Inflammation in prostate carcinogenesis. Nat. Rev. Cancer 2007, 7, 256–269. [Google Scholar] [CrossRef]
  195. Chan, J.M.; Gann, P.H.; Giovannucci, E.L. Role of diet in prostate cancer development and progression. J. Clin. Oncol. 2005, 23, 8152–8160. [Google Scholar] [CrossRef]
  196. Datta, M.; Schwartz, G.G. Calcium and vitamin D supplementation during androgen deprivation therapy for prostate cancer: A critical review. Oncologist 2012, 17, 1171–1179. [Google Scholar] [CrossRef]
  197. Cosman, F.; de Beur, S.J.; LeBoff, M.S.; Lewiecki, E.M.; Tanner, B.; Randall, S.; Lindsay, R. Clinician’s Guide to Prevention and Treatment of Osteoporosis. Osteoporos. Int. 2014, 25, 2359–2381. [Google Scholar] [CrossRef]
  198. Planas, J.; Morote, J.; Orsola, A.; Salvador, C.; Trilla, E.; Cecchini, L.; Raventos, C.X. The relationship between daily calcium intake and bone mineral density in men with prostate cancer. BJU Int. 2007, 99, 812–816. [Google Scholar] [CrossRef]
  199. Ryan, C.W.; Huo, D.; Stallings, J.W.; Davis, R.L.; Beer, T.M.; McWhorter, L.T. Lifestyle factors and duration of androgen deprivation affect bone mineral density of patients with prostate cancer during first year of therapy. Urology 2007, 70, 122–126. [Google Scholar] [CrossRef]
  200. Mata-Granados, J.M.; Cuenca-Acebedo, R.; Luque de Castro, M.D.; Quesada Gomez, J.M. Lower vitamin E serum levels are associated with osteoporosis in early postmenopausal women: A cross-sectional study. J. Bone Miner. Metab. 2013, 31, 455–460. [Google Scholar] [CrossRef]
  201. Nieman, L.K. Management of surgically hypogonadal patients unable to take sex hormone replacement therapy. Endocrinol. Metab. Clin. N. Am. 2003, 32, 325–336. [Google Scholar] [CrossRef]
  202. Steinhubl, S.R. Why have antioxidants failed in clinical trials? Am. J. Cardiol. 2008, 101, 14–19. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Dueregger, A.; Heidegger, I.; Ofer, P.; Perktold, B.; Ramoner, R.; Klocker, H.; Eder, I.E. The Use of Dietary Supplements to Alleviate Androgen Deprivation Therapy Side Effects during Prostate Cancer Treatment. Nutrients 2014, 6, 4491-4519. https://doi.org/10.3390/nu6104491

AMA Style

Dueregger A, Heidegger I, Ofer P, Perktold B, Ramoner R, Klocker H, Eder IE. The Use of Dietary Supplements to Alleviate Androgen Deprivation Therapy Side Effects during Prostate Cancer Treatment. Nutrients. 2014; 6(10):4491-4519. https://doi.org/10.3390/nu6104491

Chicago/Turabian Style

Dueregger, Andrea, Isabel Heidegger, Philipp Ofer, Bernhard Perktold, Reinhold Ramoner, Helmut Klocker, and Iris E. Eder. 2014. "The Use of Dietary Supplements to Alleviate Androgen Deprivation Therapy Side Effects during Prostate Cancer Treatment" Nutrients 6, no. 10: 4491-4519. https://doi.org/10.3390/nu6104491

Article Metrics

Back to TopTop