World J Mens Health. 2022 Jul;40(3):412-424. English.
Published online Jan 01, 2022.
Copyright © 2022 Korean Society for Sexual Medicine and Andrology
Review

Evidence on Statins, Omega-3, and Prostate Cancer: A Narrative Review

Hwanik Kim,1 and Jung Kwon Kim1,2
    • 1Department of Urology, Seoul National University Bundang Hospital, Seongnam, Korea.
    • 2Department of Urology, Seoul National University College of Medicine, Seoul, Korea.
Received July 28, 2021; Revised September 07, 2021; Accepted September 23, 2021.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Dietary intake selections might play a crucial role in prostate cancer (PCa) occurrence and progression. Several studies have investigated whether statin use could reduce PCa risk but with conflicting results. Nevertheless, a significantly decreased incidence of advanced PCa has been consistently noted. Statins may also reduce the risk of biochemical recurrence (BCR) in men with PCa after receiving active treatment. However, the influence of statin usage on BCR and PCa progression in men with high prostate-specific antigen levels has been found to be insignificant. In contrast, the combined use of a statin and metformin was significantly related to the survival status of PCa patients. However, some studies have revealed that the intake of long-chain omega-3 fatty acid (ω-3) from fish or fish oil supplements may elevate PCa risk. Several meta-analyses on ω-3 consumption and PCa have shown controversial results for the relationship between PCa and ω-3 consumption. However, studies with positive results for various genotypes, fatty acid intake or levels, and PCA risk are emerging. This review highlights the association among statins, ω-3, and PCa. The findings summarized here may be helpful for clinicians counseling patients related to PCa.

Keywords
Omega-3 fatty acid; Prostate cancer; Recurrence; Risk; Statin; Survival

INTRODUCTION

Intriguing correlations between prostate cancer (PCa) and the usage of statin and omega-3 fatty acid (ω-3) have been reported. Statins are common drugs prescribed for dyslipidemia and ω-3s are fish oil supplements. One study [1] concluded the statin administration had a strong correlation with the decreased risk of PCa metastasis and mortality. Another study [2] investigated the effects of edible ω-3 on PCa progression in mice and resulted that its usage could suppress tumor cells’ growth. Nevertheless, these interesting associations remains controversial, warranting active research for further elucidation.

EFFECT OF STATINS ON PROSTATE CANCER

Growing evidence that supports the assumption that statin usage is related to a reduced risk of advanced PCa is being reported [3]. However, whether statin use in men with PCa could prevent biochemical recurrence (BCR) or disease progression remains unclear. Thus, the benefits of statins must be evaluated before we can recommend statins initiation for males at risk for PCa despite their cholesterol profile. Several clinical findings are summarized in Table 1.

Table 1
Clinical studies conducted for statins and PCa

1. Mechanism of statins and effects of statins on prostate cancer

The underlying mechanism of statins is by decreasing cholesterol synthesis via suppression of rate-limiting hepatic 3hydroxy3methylglutaryl coenzyme A (HMG-CoA) reductase. Statins are classified according to solubility as either lipophilic or hydrophilic. Lipophilic statins are presumed to have a larger impact on the prostate than hydrophilic agents [3]. However, this assumption has not been validated by observational investigations of PCa risk and statin usage, partly due to the low number of males using hydrophilic statins.

A positive relationship between PCa presence and cholesterol accumulation in prostatic tissues has already been reported [4]. Some mechanisms related to serve to dysregulation of cholesterol balance in PCa have since been revealed. Lee et al [5] reported that the ABCA1 promoter hypermethylation resulted in decreased encoded cholesterol efflux transporter expression, reduced cholesterol efflux rates, and increased intracellular cholesterol concentrations in PCa cell lines. They also showed that the existence of this epigenetic modification was related to high-grade PCa. Moreover, the mTOR pathway is essential in controlling sterol-regulatory-element-binding proteins (SREBPs), which are transcription factors controlling cholesterol and lipid balance [6]. Yue et al [7] showed that the intracellular aggregation of cholesteryl ester in lipid droplets was induced by the loss of tumor suppressor PTEN expression and further activation of the PI3K-AKT-mTOR signaling pathway. They also showed that the intracellular aggregation of cholesteryl ester was related to high-grade PCa [7]. One of the main cholesterol-mediated processes [3] by which statins suppress tumor growth involves particular cholesterol-abundant domains in the cell membrane referred to as lipid rafts [8]. These regions can ease membrane-initiated signaling episodes in cells via the compartmentalization of signaling routes, promoting tumor growth. Cell signaling routes involved PCa occurrence and progression, which could be impacted by lipid raft cholesterol distribution, include pathways related to the androgen receptor [9], the luteinizing hormone receptor [10], and the epidermal growth factor receptor (EGFR) [11]. Statins, via their effects on intracellular cholesterol balance, are considered to disturb lipid raft organization, hence interfering with these or other downstream intracellular signaling routes [12]. The treatment of PCa cells with cholesterol binders can interfere with EGFR signaling and disorganize lipid raft organization [11]. Other signaling routes involved in the occurrence of PCa and castration resistance, like IL 6 activated Janus kinase-signal transducer and activator of transcription 3 (JAK-STAT3) signaling, are influenced by lipid raft organization. Therefore, they are likely affected by lipid raft cholesterol levels [13]. As cholesterol is androgens’ precursor, cholesterol levels could also influence the development of PCa through the androgen signaling pathway. Decreasing cholesterol levels using statins might lower PCa growth by decreasing intratumoral or serum androgen levels. However, the effect of statins on serum androgen levels remains unsure. Several investigations have proposed that statins can decrease serum testosterone [14, 15, 16]. However, these decreases were minimal or induced by statin doses higher than are generally utilized in the real world. Emerging evidence has suggested that intratumoral androgen levels are persistently high even when castrated androgen levels are attained in the sera of PCa men conceivably due to novel androgen synthesis in tumor cells [17, 18, 19]. Therefore, statins could diminish intratumoral androgen levels by decreasing intratumoral cholesterol levels.

As for non-cholesterol mediated pathways, change from HMG-CoA to mevalonate could be suppressed by statins. Mevalonate belongs to a class of isoprenoids that includes geranyl pyrophosphate and farnesyl pyrophosphate. Geranyl pyrophosphate and farnesyl pyrophosphate can ease the recruitment of signaling proteins like G proteins of the Ras and Rho superfamilies by engaging their connection to plasma membranes, where their signaling actions could enhance PCa cell viability and proliferation [20, 21]. Therefore, statins might inhibit cancer cell proliferation by lowering mevalonate and downstream isoprenoids. Moreover, statins seem to directly promote apoptosis in tumor cells regardless of their effects on cholesterol levels [22]. For instance, statins were shown to suppress cyclin-dependent kinase 2 and induce cell cycle arrest [23] or trigger apoptosis-stimulating specific proteases [24]. Statins also possess direct antiangiogenic and anti-inflammatory features. They were also reported to suppress tumor growth and progression [22]. One study [25] of a group of patients receiving radical prostatectomy (RP) showed that statin takers were 69% less likely to display inflammation in their tumors than non-takers (p=0.047). Hoque et al [26] evaluated the action of statins on PCa cells and the elemental molecular mechanism of action. They found that antitumor function of statins was due to the promotion of growth arrest and cell apoptosis. The fundamental molecular mechanism of action of statins is arbitrated via RhoA inactivation, which successively promotes caspase enzymatic action with or without G1 cell cycle. Raittinen et al [27] elucidated atorvastatin’s impact on serum and prostatic tissue steroidomic profiles and exposed new pathways for lowering androgen levels in men with PCa. Most serum and prostatic steroids, including dihydrotestosterone and testosterone, were not related to atorvastatin usage. Yet, they concluded that atorvastatin use was associated with an adrenal androgen decrease in the serum and likely in the prostate.

2. Evidence on statin use and prostate cancer risk or prevention

Bonovas et al [28] performed a meta-analysis including several randomized clinical trials (RCTs) [29, 30, 31, 32, 33, 34]. They reported an insignificant association between statin usage and overall PCa among either RCTs (pooled relative risk [RR], 1.06; 95% confidence interval [CI], 0.93–1.20) or observational investigations (high heterogeneity detected) (pooled RR, 0.89; 95% CI, 0.65–1.24). Furthermore, longstanding statin usage did not significantly influence the total PCa risk (pooled RR, 0.93; 95% CI, 0.77–1.13). Nevertheless, the synthesis of applicable studies that particularly evaluated statin usage concerning advanced PCa indicated a protective relationship (pooled RR, 0.77; 95% CI, 0.64–0.93). Stranberg et al [32] investigated extended post-trial follow-up of an RCT of simvastatin or placebo in men with heart disease. They found that 227 and 248 incident cancers were reported in the simvastatin and placebo group, respectively (0.88; 95% CI, 0.73–1.05; p=0.150). The incidence of any malignancy type did not increase in the simvastatin group (55 and 51 PCas were diagnosed in the placebo and simvastatin groups, respectively). The Heart Protection Study Collaborative Group [33] performed an RCT including 20,536 patients aged 40 to 80 years with diabetes or vascular disease. The study found that the use of simvastatin did not reduce PCa (event ratio, 0.99; 95% CI, 0.79–1.25). Murtola et al [35] found that ever-usage of any statin was associated with a marginally elevated total PCa risk (odds ratio [OR], 1.07; 95% CI, 1.00–1.16). Nevertheless, none of these statins was related to the total PCa risk when analyzed individually. The risk of advanced PCa was reduced among the users of atorvastatin, simvastatin, and lovastatin (OR 0.61, 95% CI 0.37–0.98; OR 0.78, 95% CI 0.61–1.01; OR 0.61, 95% CI 0.43–0.85, individually). The risk was not affected in the users of other cholesterol medication groups. Kang et al [36] analyzed 143,870 males to evaluate the risk of kidney malignancy, bladder malignancy, and PCa developments, individually, during a 10-year follow-up (2004–2013). In their study, statins did not appear to be a predictive factor for the development of PCa (hazard ratio [HR], 1.01; 95% CI, 0.85–1.18; p=0.953). Kaye and Jick [37] studied the General Practice Research Database and reported that present statin usage was not related to a significantly modified PCa risk. However, untreated hyperlipidemia was related to a slightly elevated PCa risk (RR, 1.5; 95% CI, 1.1–2.0). Wang et al [38] reported that statin usage was associated with the decreased risks of both Gleason score (GS) low-grade PCa (<7) (HR, 0.85; 95% CI, 0.74–0.96) and high-grade PCa (≥7) (HR, 0.54; 95% CI, 0.42–0.69). This protective relationship was detected exclusively when statins were taken for a longer period (≥11 months) or a stronger dosage (≥121 daily doses). It was more prominent in men with PCa with a higher GS. Lipophilic agents (HR, 0.83; 95% CI, 0.72–0.95) might be more protective against PCa than hydrophilic agents (HR, 0.91; 95% CI, 0.63–1.33). Yu et al [39] analyzed 11,772 men with non-metastatic PCa (mean 4.4 years follow-up) and reported that post-diagnostic statin usage was associated with a lower possibility of PCa-specific death (HR, 0.76; 95% CI, 0.66–0.88) and overall death (HR, 0.86; 95% CI, 0.78–0.95). These reduced risks of PCa-specific death and overall death were more prominent in men who took statins before PCa diagnosis (HR 0.55, 95% CI 0.41–0.74; and HR 0.66; 95% CI 0.53–0.81, respectively), with lower impacts on men who started the therapy exclusively after PCa diagnosis (HR 0.82, 95% CI 0.71–0.96; and HR 0.91, 95% CI 0.82–1.01, respectively).

3. Evidence on statin use and active surveillance for prostate cancer

Nyame et al [40] retrospectively studied men with active surveillance (2005–2015) and evaluated disease reallocation, progression to decisive curative therapy, and surveillance failure (the occurrence of either biochemical failure (BF) after curative treatment, metastasis, or PCa-specific death) among statin and non-statin users. They found no significant difference in the BF rate among patients who proceeded to definitive treatment when stratified by statin usage (p=0.890). Statin use duration was inversely related to adverse RP pathology in both univariate (OR, 0.99; 95% CI, 0.98–0.99; p=0.030) and multivariate analysis (OR, 0.98; 95% CI, 0.97–0.99; p=0.020).

4. Evidence on statin use and biochemical recurrence after definitive local therapy (radical prostatectomy or radiation therapy) for prostate cancer

High variability in the definition of BCR following therapy for localized PCa exists. Following RP, it is recommended to define BCR as an initial serum prostate-specific antigen (PSA) of ≥0.2 ng/mL, with a second confirmatory PSA level of ≥0.2 ng/mL. Regarding radiation therapy (RT), BCR is defined as three consecutive rises in PSA after a nadir has been reached, with the failure date being the midpoint between the nadir and the first of three consecutive increases [41].

Allott et al [42] studied the influence of post-RP statin usage on BCR in men with PCa who had never taken statins before RP. They revealed that post-RP statin usage was significantly related to 36% decreased BCR risk (HR, 0.64; 95% CI, 0.47–0.87; p=0.004). After adjusting for preoperative serum cholesterol levels, post-RP statin usage was still related to a reduced BCR risk. In a secondary investigation, this protective relationship was pronounced in non-Afro-American patients (HR, 0.49; 95% CI, 0.32–0.75; p=0.001), but not in Afro-American patients (HR, 0.82; 95% CI, 0.53–1.28; p=0.384) after stratification by race. Jeong et al [43] investigated the efficacy of adjuvant atorvastatin in men with PCa after RP in a double-blind RCT. Among the PCa patients with high-risk pathologic features after RP, 1-year adjuvant atorvastatin usage was not associated with a lower risk of BCR compared to that for placebo (HR, 0.96; 95% CI, 0.58–1.60). Scosyrev et al [44] executed a meta-analysis (5 based on RP and 3 based on RT series). They reported that the pooled HR (pHR) estimates for BCR risk in statin takers vs. never-takers were 0.91 (95% CI, 0.72–1.13) for the overall investigations, 1.02 (95% CI, 0.80–1.29) for the RP studies, and 0.71 (95% CI, 0.44–1.16) for the RT studies. However, there were considerable disagreements in the reported findings and conclusions between the individual studies. Chao et al [45] analyzed 1,200 men with PCa who underwent RP for BCR with a follow-up for up to 5 years (a single PSA assessment of >0.2 ng/mL) and clinical progression (diagnosis of metastasis or PCa-specific mortality). Of these men, 37% and 56% used statins preoperatively and postoperatively, respectively. They reported that neither pre- nor postoperative statin usage was affiliated with disease progression (HR 0.63, 95% CI 0.31–1.27 and HR 1.20, 95% CI 0.63–2.30, respectively) or BCR (HR 1.00, 95% CI 0.72–1.39 and HR 1.05, 95% CI 0.76–1.46, respectively). An unclear dose-response relationship was revealed for the duration of usage. Meijer et al [46] investigated whether statin usage could decrease the incidence of advanced PCa in males with high PSA levels (≥4.0 ng/mL) and whether statin usage could reduce post-RP BCR risk. Of their subjects, 72% were confirmed to have PCa. At the time of PCa diagnosis, 23% had taken statins compared to 19% in the non-PCa men (p=0.100). There was an insignificant difference in statin use between the different risk groups. No association was proven between post-RP BCR risk and statin use in the total subjects (p=0.200), the intermediate-risk group (p=0.630), or the high-risk group (p=0.140). Raval et al [47] performed a meta-analysis and reported that statin usage was associated with a 21% decrease in BCR risk in patients who underwent RT (pHR, 0.79; 95% CI, 0.65–0.95; p=0.010; 10 studies; I2=54%). However, statin usage was not related to BCR among patients who underwent RP (pHR, 0.94; 95% CI, 0.81–1.09; p=0.430; 15studies; I2=65%). Otherwise, statin usage was associated with 22% decrease in metastasis possibility (pHR, 0.78; 95% CI, 0.68–0.87; p=0.001; 6 studies; I2=0%) and a 24% decrease in the risk of both cancer-specific death (pHR, 0.76; 95% CI, 0.64–0.89; p=0.0007; 5 studies; I2=40%) and overall death (pHR, 0.76; 95% CI, 0.63–0.91; p=0.004; 6 studies; I2=71%). Huynh et al [48] evaluated 1,581 patients undergoing RP or RT and the effect of statin usage on overall BF and the time to BF after initial treatment for localized PCa. When classified by statin usage, BF overall and within first, third, and fifth year did not differ significantly. A shorter time to BF was shown in men taking statins (1.8±1.9 y vs. 2.4±2.6 y; p=0.016). This finding remained in the multivariate investigation, wherein statin usage was not associated with BF, although it was related to a shorter time to BF.

5. Evidence for the effect of statin usage and clinical outcomes in advanced prostate cancer and the effect of statin use alone or combination with other drugs on prostate cancer

Platz et al [49] evaluated the relationship between statin usage and overall and advanced PCa in 34,989 American males who had no tumor in 1990 and were tracked until 2002. The age-standardized incidences of advanced PCa was 89 and 38 per 100,000 person-years in never or past-takers and current statin takers, respectively. The adjusted RR was 0.51 (95% CI, 0.30–0.86) for advanced disease and 0.39 (95% CI, 0.19–0.77) for lethal or metastatic disease among current statin users compared to non-current users. These significant associations were retained after adjusting for PSA screening record (advanced case: RR 0.57, 95% CI 0.30–1.11; lethal or metastatic case: RR 0.35, 95% CI 0.14–0.92). The risk of advanced PCa was lower with longer statin usage (p=0.003). Compared to never use, the RR was 0.60 for <5 years of use (95% CI, 0.35–1.03) and 0.26 for ≥5 years of use (95% CI, 0.08–0.83). No affiliation was shown between total PCa risk and statin usage (RR, 0.96; 95% CI, 0.85–1.09). Peltomaa et al [50] evaluated the effect of statins on PCa prognosis among patients managed with androgen deprivation therapy (ADT). Post-ADT statin usage was related to a reduced risk of PSA relapse (HR, 0.73; 95% CI, 0.65–0.82) and PCa-specific mortality (HR, 0.82; 95% CI, 0.69–0.96). However, statin usage with a one-year lag (HR, 0.89; 95% CI, 0.76–1.04), pre-ADT statin usage (HR, 1.12; 95% CI, 0.96–1.31), and stain usage in the first year of ADT (HR, 1.02; 95% CI, 0.85-1.24) were not associated with PCa mortality, indicating no dose dependency. Yang et al [51] investigated the impact of statin usage on the outcomes of men with advanced PCa treated with abiraterone/enzalutamide or ADT. They found that statin usage significantly lowered the risk of overall death (HR, 0.73; 95% CI, 0.64–0.83; p<0.00001) and the risk of PCa-specific mortality (HR, 0.64; 95% CI, 0.53–0.77; p<0.00001) in men with advanced PCa undergoing ADT. However, these findings provided no credible evidence for men with metastatic castration-resistant PCa (mCRPC) treated with enzalutamide/abiraterone since related investigations are scarce with inconsistent results.

Jiménez-Vacas et al [52] evaluated the presumed in vivo relationship between statins and/or metformin treatment and the core tumor and clinical factors. They also assessed the direct effects of different statins (atorvastatin, lovastatin, and simvastatin), biguanides (metformin, phenformin, and buformin), and their combination. The combination of statins and metformin in vivo was associated with a lower GS and longer BCR-free survival. Furthermore, statins and biguanides showed strong antitumor effects on PCa cells. These actions were mediated via the modulation of molecular mediators and key metabolic and oncogenic signaling pathways. Tan et al [53] have quantified individual and synergistic effects of statin and metformin usage among high-risk PCa patients. Statin-only or combined with metformin was significantly related to decreased PCa-specific death (HR 0.80, 95% CI 0.69–0.92 and HR 0.64, 95% CI 0.51–0.81, respectively) and overall death (HR 0.89, 95% CI 0.83–0.96 and HR 0.75, 95% CI 0.67–0.83, respectively). These impacts were more prominent in post-diagnostic takers, where the combined usage of statins and metformin was related to a 32% decrease in overall death (95% CI, 0.57–0.80) and a 54% decrease in PCa-specific death (95% CI, 0.30–0.69). However, an insignificant relationship between metformin usage only was shown with either PCa-specific death or all-cause death.

EFFECT OF OMEGA-3 ON PROSTATE CANCER

Omega-3s are polyunsaturated fatty acids (PUFAs) containing a double bond at carbon number 3 in the omega naming system [54]. They are proven to be anti-inflammatory substances against cardiovascular diseases and malignancies. They also play a nutrigenetic role by interacting with genes involved in inflammation and cancers [55]. Omega-3s include long-chain α-linolenic acid (ALA), docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA). Recent epidemiological investigations have proven that continuous oily fish intake was associated with decreased PCa risk. PUFAs, both ω-3 and omega-6 fatty acid (ω-6), are changed to eicosanoids (e.g., thromboxanes and prostaglandins) in the body. These compounds are commonly engaged in cell growth and differentiation, anti-inflammatory process, and immune modulation [56]. ALA is believed to be a fundamental fatty acid and must be obtained by the consumption of food or supplements because it cannot be synthesized by the body. Nevertheless, edible intake is not considered essential for health as DHA and EPA are made from ALA in the body [57]. Omega-3s can also be obtained from ALA containing plant oils such as walnuts, flaxseed, or canola [58]. Omega-3s are integrated into various parts of the body. For instance, DHA is a core element for all cell membranes [59]. EPA and DHA are metabolites, which can act as lipid mediators expected to be efficient in treating or preventing some disorders [60]. Several clinical findings are summarized in Table 2.

Table 2
Clinical studies conducted on ω-3 and PCa

1. Mechanism and effects of omega-3 on prostate cancer

The mechanism by which ω-3 prevents carcinogenesis involves the suppressive effect on arachidonic acid synthesized eicosanoids [61]. Eicosanoids extracted from arachidonic acid exert pro-inflammatory activity while ω-3 derived eicosanoids exert anti-inflammatory activity, thus preventing PCa. In eicosanoid synthesis, cyclooxygenase-2 (COX-2) acts as a core enzyme. Investigations have revealed that there is an overproduction of COX-2 in PCa [62, 63]. Eicosanoids derived from ω-3s have a suppressive effect on COX-2 overproduction, thus preventing PCa. Hedelin et al [64] found a meaningful relationship between the use of salmon-type fish abundant in omega-3 unsaturated fats and a genetic variation of COX-2 in establishing PCa risk. Among homozygotes or heterozygotes of the variant allele of +6365T/C SNP in COX-2, large salmon fish intake was associated with a prominent PCa risk decrease, although there was an insignificant association between fish intake and tumor potentiality in wild-type allele carriers [65].

It is possible that some metabolic statuses favoring the aggregation of long-chain ω-3 (LCn3) in plasma phospholipids has an additional promoting impact on PCa. For instance, estrogen function has been linked to an increased risk of advanced PCa, indicating a role of estrogen receptor-alpha in PCa progression [66, 67].

A recent study [68] reported the mechanisms underlying tumor lymph node (LN) metastasis. It was found that LN metastasis mandates tumor cells to go through a metabolic change toward fatty acid oxidation (FAO). In LN-metastatic tumors, selectively activated transcriptional coactivator yes-associated protein (YAP) leads to the upregulation of genes in the FAO signaling route. The genetic ablation of YAP or pharmacological suppression of FAO could inhibit LN metastasis in mice. These findings are consistent with our perspective on ω-3 discussed above.

2. Evidence on omega-3 use and prostate cancer risk or prevention

Circumstantial factors seem to be involved in the occurrence of hormone-dependent malignancies like PCa. The effect of dietary fat consumption on PCa has diversely been investigated. However, no precise conclusion has been reached yet. Preclinical studies have indicated the protective effect of ω-3s on tumor progression. From a comprehensive review of epidemiologic data, Terry et al [69] concluded that with a general paucity of research that includes essential measurements like consumed fish type and tissue ω-3 concentrations, there is scarce evidence to confirm that an association exists between human cancer and marine fatty acid consumption. Brasky et al [70] examined the relationships between the 7-year prevalence of PCa and phospholipid fatty acids and in a nested case-control investigation of subjects in the Prostate Cancer Prevention Trial. No fatty acids were related to low-grade PCa risk. DHA was positively related to high-grade PCa (OR, 2.50; 95% CI, 1.34–4.65). Trans-fatty acid (TFA) 18:1 and TFA 18:2 were inversely and linearly correlated with high-grade PCa risk (TFA 18:1: OR 0.55, 95% CI 0.30–0.98; TFA 18:2: OR 0.48, 95% CI 0.27–0.84). Another research study done by the same group [71] has evaluated relationships between PCa risk and plasma phospholipid fatty acids among subjects in the Selenium and Vitamin E Cancer Prevention Trial. Compared to subjects in the bottom quartiles of LCn3, the participants in the top quartile had higher possibilities of low-grade PCa (HR, 1.44; 95% CI, 1.08–1.93), high-grade PCa (HR, 1.71; 95% CI, 1.00–2.94), and overall PCa (HR, 1.43; 95% CI, 1.09–1.88). Elevated linoleic acid (ω-6) was related to decreased risks of low-grade PCa (HR, 0.75; 95% CI, 0.56–0.99) and overall PCa (HR, 0.77; 95% CI, 0.59–1.01). They also found that blood levels of DHA, but not EPA, were significantly related to an elevated possibility of low-grade PCa (RR, 1.20; 95% CI, 1.04–1.38), advanced PCa (RR, 1.48; 95% CI, 1.10–1.99), and total PCa (RR, 1.16; 95% CI, 1.03–1.31) comparing the lower and upper quantiles. From their analysis, they suggested the possibility that this relationship might be causal. The authors suggested that typical advice for raising LCn3 consumption should consider its possible harm.

Chua et al [72] systematically analyzed the association between PCa risk and serum level long chain ω-3 in human epidemiological studies. A significant inverse association was revealed between total PCa risk and high serum ω-3 levels including docosapentaenoic acid (DPA) (RR, 0.756; 95% CI, 0.599–0.955; p=0.019). Correspondingly, a positive relationship between high serum levels of EPA and DHA and high-grade PCa incidence (RR, 1.381; 95% CI, 1.050–1.817; p=0.021) was reported. Hanson et al [73] studied the relationships between LCn3, ALA, ω-6, and total PUFA intake with cancer risk. Increasing LCn3 was shown to slightly elevate PCa risk (low quality evidence). However, its impacts on PCa mortality were imprecise (very low-quality evidence, 5 mortalities). Seven studies (38,525 men, mean period 51 months, mean dose 1.2 g/d LCn3) showed 1,021 confirmed PCa cases, revealing an elevated PCa risk in males with elevated LCn3 intake (RR, 1.10; 95% CI, 0.97–1.24; I2=0%; 334 needed to treat to cause additional harm). This superficial increase in PCa risk was reliable in overall sensitivity analysis. However, the risk probability was not supported by the PSA data shown in a single large study (25% reduction, mean difference: -0.13 ng/mL; 95% CI, -0.25 to 0.01; 1,622 men). PSA elevation was reported in 12 of 62 subjects in another study (RR, 0.47; 95% CI, 0.16–1.40), also refuting the suggested LCn3 harm. Sorongon-Legaspi et al [74] showed a non-significant relationship of the overall effect estimates for total, high-grade, or advanced PCa. High serum ALA levels had a non-significant positive relationship with total PCa risk. High serum DPA levels had a significant negative relationship with total PCa risk. Particular ω-3s in fish oil, DHA, and EPA, were positively associated with high-grade PCa risk exclusively after adjusting for inter-study inconsistency. Torfadottir et al [75] explored fish consumption, particularly in adolescence and midlife, and evaluated the effect of smoked or salted fish and fish oil intake on PCa risk in a prospective cohort investigation. Large fish intake in early- and midlife was not related to total or advanced PCa risk. The large consumption of smoked or salted fish was related to a two-fold elevation in advanced PCa risk in early (95% CI, 1.08–3.62) and in later life (95% CI, 1.04–5.00). Males taking fish oil in later life had a lower risk of advanced PCa (HR, 0.43; 95% CI, 0.19–0.95). However, no such relationship was revealed for early- or midlife intake. Aucoin et al [76] evaluated the efficacy and safety of fish-derived ω-3 on the occurrence and progression of PCa through a systemic review of 44 studies. Interventional research using fish oil supplementations in PCa men revealed no impact on PSA concentrations. However, two studies reported decreases of inflammatory markers and other cancer markers. A small number of mild side effects have been reported. The results of case-control and cohort research evaluating the relationship between PCa risk and dietary fish consumption are equivocal. Cohort studies evaluating PCa-specific mortality have proposed a relationship between the reduced risk of PCa-specific death and higher fish consumption. Zuniga et al [77] implemented a questionnaire study of 7,989 Cancer of the Prostate Strategic Urologic Research Endeavor (CaPSURE) participants and reported that multivitamin and ω-3 use was common (in 40% and 24% of patients, respectively). The study discussed a vitamin D and ω-3 trial. It revealed no significant reduction in death from PCa in those treated with ω-3 (HR, 1.15; 95% CI, 0.94–1.39) or vitamin D (HR, 0.88; 95% CI, 0.72–1.07) for a median of 5.3 years compared to placebo. Several meta-analyses [78, 79, 80, 81, 82] on the consumption of ω-3 and PCa have been published. Some meta-analyses reported statistically meaningful results for the relationship between PCa and ω-3 consumption. One meta-analysis [79] revealed that LCn3 consumption increased PCa risk (RR, 1.14; 95% CI, 1.01–1.28), while the other two studies [79, 80] revealed a protective effect of ω-3 consumption on PCa. One study [81] reported a marginally insignificant relationship between PCa and high fish intake (p=0.05).

3. Evidence on omega-3 use and various stages of prostate cancer

Campbell et al [83] evaluated the effect of targeted serum vitamin-D levels and the omega-6:3 fatty-acid ratio on PSA concentration in PCa patients supervised with active surveillance that included nutritional intervention and vitamin supplements. They found that men with higher initial vitamin D concentrations were more likely to show a decreasing PSA tendency (OR, 2.04; 95% CI, 1.04–4.01; p=0.040). Fifty-five of 68 men with subsequent biopsies showed no disease progression. They concluded that nutritional intervention with ω-3 and vitamin D supplementation might benefit patients in active surveillance for PCa. Liang et al [84] reported that ω-3 intake combined with castration could cause greater MycCap tumor (grown subcutaneously in mice) regression than the ω-6 diet (p=0.003) and also significantly delay the time to CRPC (p=0.006). Similarly, ω-3 intake meaningfully slowed the progression of demonstrated castrate-resistant MycCaP tumors (p=0.003). Dietary ω-3 reduced CRPC occurrence and progression in an immunocompetent mouse model with suppressive effects on M2-like macrophage activity.

CONCLUSIONS

Albeit evidence that statin and ω-3 are effective in preventing PCa has been continuously accumulated, it has not reached the level of recommendations in guidelines. Nevertheless, it is expected that the time will soon come when we can confirm a clear effect on PCa prevention or PCa progression reduction through accumulating evidence. There will be abundant hopeful situations in which the use of statins or ω-3s can be commonly considered in real clinical practice consistent with the objectives in the era of precision medicine.

Notes

Conflict of Interest:The authors have nothing to disclose.

Funding:None.

Author Contribution:

  • Conceptualization: all authors.

  • Data curation: all authors.

  • Formal analysis: all authors.

  • Funding acquisition: JKK.

  • Investigation: all authors.

  • Methodology: all authors.

  • Project administration: all authors.

  • Resources: all authors.

  • Software: all authors.

  • Supervision: JKK.

  • Validation: all authors.

  • Visualization: all authors.

  • Writing — original draft: all authors.

  • Writing — review & editing: all authors.

References

    1. Allott EH, Freeman MR, Freedland SJ. Statin therapy to improve prostate cancer outcomes: who, when, and for how long? Eur Urol 2018;74:702–703.
    1. Gevariya N, Besançon M, Robitaille K, Picard V, Diabaté L, Alesawi A, et al. Omega-3 fatty acids decrease prostate cancer progression associated with an anti-tumor immune response in eugonadal and castrated mice. Prostate 2019;79:9–20.
    1. Alfaqih MA, Allott EH, Hamilton RJ, Freeman MR, Freedland SJ. The current evidence on statin use and prostate cancer prevention: are we there yet? Nat Rev Urol 2017;14:107–119.
    1. Schaffner CP. Prostatic cholesterol metabolism: regulation and alteration. Prog Clin Biol Res 1981;75A:279–324.
    1. Lee BH, Taylor MG, Robinet P, Smith JD, Schweitzer J, Sehayek E, et al. Dysregulation of cholesterol homeostasis in human prostate cancer through loss of ABCA1. Cancer Res 2013;73:1211–1218.
    1. Porstmann T, Santos CR, Griffiths B, Cully M, Wu M, Leevers S, et al. SREBP activity is regulated by mTORC1 and contributes to Akt-dependent cell growth. Cell Metab 2008;8:224–236.
    1. Yue S, Li J, Lee SY, Lee HJ, Shao T, Song B, et al. Cholesteryl ester accumulation induced by PTEN loss and PI3K/AKT activation underlies human prostate cancer aggressiveness. Cell Metab 2014;19:393–406.
    1. Simons K, Ikonen E. Functional rafts in cell membranes. Nature 1997;387:569–572.
    1. Freeman MR, Cinar B, Lu ML. Membrane rafts as potential sites of nongenomic hormonal signaling in prostate cancer. Trends Endocrinol Metab 2005;16:273–279.
    1. Smith SM, Lei Y, Liu J, Cahill ME, Hagen GM, Barisas BG, et al. Luteinizing hormone receptors translocate to plasma membrane microdomains after binding of human chorionic gonadotropin. Endocrinology 2006;147:1789–1795.
    1. Zhuang L, Lin J, Lu ML, Solomon KR, Freeman MR. Cholesterol-rich lipid rafts mediate akt-regulated survival in prostate cancer cells. Cancer Res 2002;62:2227–2231.
    1. Lawrence JC, Saslowsky DE, Edwardson JM, Henderson RM. Real-time analysis of the effects of cholesterol on lipid raft behavior using atomic force microscopy. Biophys J 2003;84:1827–1832.
    1. Horinaga M, Okita H, Nakashima J, Kanao K, Sakamoto M, Murai M. Clinical and pathologic significance of activation of signal transducer and activator of transcription 3 in prostate cancer. Urology 2005;66:671–675.
    1. Dobs AS, Schrott H, Davidson MH, Bays H, Stein EA, Kush D, et al. Effects of high-dose simvastatin on adrenal and gonadal steroidogenesis in men with hypercholesterolemia. Metabolism 2000;49:1234–1238.
    1. Hyyppä MT, Kronholm E, Virtanen A, Leino A, Jula A. Does simvastatin affect mood and steroid hormone levels in hypercholesterolemic men? A randomized double-blind trial. Psychoneuroendocrinology 2003;28:181–194.
    1. Smals AG, Weusten JJ, Benraad TJ, Kloppenborg PW. The HMG-CoA reductase inhibitor simvastatin suppresses human testicular testosterone synthesis in vitro by a selective inhibitory effect on 17-ketosteroid-oxidoreductase enzyme activity. J Steroid Biochem Mol Biol 1991;38:465–468.
    1. Mohler JL, Gregory CW, Ford OH 3rd, Kim D, Weaver CM, Petrusz P, et al. The androgen axis in recurrent prostate cancer. Clin Cancer Res 2004;10:440–448.
    1. Page ST, Lin DW, Mostaghel EA, Hess DL, True LD, Amory JK, et al. Persistent intraprostatic androgen concentrations after medical castration in healthy men. J Clin Endocrinol Metab 2006;91:3850–3856.
    1. Pelletier G, Luu-The V, El-Alfy M, Li S, Labrie F. Immunoelectron microscopic localization of 3beta-hydroxysteroid dehydrogenase and type 5 17beta-hydroxysteroid dehydrogenase in the human prostate and mammary gland. J Mol Endocrinol 2001;26:11–19.
    1. Graaf MR, Richel DJ, van Noorden CJ, Guchelaar HJ. Effects of statins and farnesyltransferase inhibitors on the development and progression of cancer. Cancer Treat Rev 2004;30:609–641.
    1. Weber MJ, Gioeli D. Ras signaling in prostate cancer progression. J Cell Biochem 2004;91:13–25.
    1. Demierre MF, Higgins PD, Gruber SB, Hawk E, Lippman SM. Statins and cancer prevention. Nat Rev Cancer 2005;5:930–942.
    1. Lee SJ, Ha MJ, Lee J, Nguyen P, Choi YH, Pirnia F, et al. Inhibition of the 3-hydroxy-3-methylglutaryl-coenzyme A reductase pathway induces p53-independent transcriptional regulation of p21(WAF1/CIP1) in human prostate carcinoma cells. J Biol Chem 1998;273:10618–10623.
    1. Marcelli M, Cunningham GR, Haidacher SJ, Padayatty SJ, Sturgis L, Kagan C, et al. Caspase-7 is activated during lovastatin-induced apoptosis of the prostate cancer cell line LNCaP. Cancer Res 1998;58:76–83.
    1. Bañez LL, Klink JC, Jayachandran J, Lark AL, Gerber L, Hamilton RJ, et al. Association between statins and prostate tumor inflammatory infiltrate in men undergoing radical prostatectomy. Cancer Epidemiol Biomarkers Prev 2010;19:722–728.
    1. Hoque A, Chen H, Xu XC. Statin induces apoptosis and cell growth arrest in prostate cancer cells. Cancer Epidemiol Biomarkers Prev 2008;17:88–94.
    1. Raittinen PVH, Syvälä H, Tammela TLJ, Häkkinen MR, Ilmonen P, Auriola S, et al. Atorvastatin induces adrenal androgen downshift in men with prostate cancer: a post hoc analysis of a pilot adaptive randomised clinical trial. EBioMedicine 2021;68:103432
    1. Bonovas S, Filioussi K, Sitaras NM. Statin use and the risk of prostate cancer: a metaanalysis of 6 randomized clinical trials and 13 observational studies. Int J Cancer 2008;123:899–904.
    1. Downs JR, Clearfield M, Weis S, Whitney E, Shapiro DR, Beere PA, et al. Primary prevention of acute coronary events with lovastatin in men and women with average cholesterol levels: results of AFCAPS/TexCAPS. Air Force/Texas coronary atherosclerosis prevention study. JAMA 1998;279:1615–1622.
    1. LIPID Study Group (Long-term Intervention with Pravastatin in Ischaemic Disease). Long-term effectiveness and safety of pravastatin in 9014 patients with coronary heart disease and average cholesterol concentrations: the LIPID trial follow-up. Lancet 2002;359:1379–1387.
    1. Serruys PW, de Feyter P, Macaya C, Kokott N, Puel J, Vrolix M, et al. Lescol Intervention Prevention Study (LIPS) Investigators. Fluvastatin for prevention of cardiac events following successful first percutaneous coronary intervention: a randomized controlled trial. JAMA 2002;287:3215–3222.
    1. Strandberg TE, Pyörälä K, Cook TJ, Wilhelmsen L, Faergeman O, Thorgeirsson G, et al. 4S Group. Mortality and incidence of cancer during 10-year follow-up of the Scandinavian Simvastatin Survival Study (4S). Lancet 2004;364:771–777.
    1. Heart Protection Study Collaborative Group. The effects of cholesterol lowering with simvastatin on cause-specific mortality and on cancer incidence in 20,536 high-risk people: a randomised placebo-controlled trial [ISRCTN48489393]. BMC Med 2005;3:6
    1. Ford I, Murray H, Packard CJ, Shepherd J, Macfarlane PW, Cobbe SM. West of Scotland Coronary Prevention Study Group. Long-term follow-up of the West of Scotland coronary prevention study. N Engl J Med 2007;357:1477–1486.
    1. Murtola TJ, Tammela TL, Lahtela J, Auvinen A. Cholesterol-lowering drugs and prostate cancer risk: a population-based case-control study. Cancer Epidemiol Biomarkers Prev 2007;16:2226–2232.
    1. Kang M, Ku JH, Kwak C, Kim HH, Jeong CW. Effects of aspirin, nonsteroidal anti-inflammatory drugs, statin, and COX2 inhibitor on the developments of urological malignancies: a population-based study with 10-year follow-up data in Korea. Cancer Res Treat 2018;50:984–991.
    1. Kaye JA, Jick H. Statin use and cancer risk in the General Practice Research Database. Br J Cancer 2004;90:635–637.
    1. Wang K, Gerke TA, Chen X, Prosperi M. Association of statin use with risk of Gleason score-specific prostate cancer: a hospital-based cohort study. Cancer Med 2019;8:7399–7407.
    1. Yu O, Eberg M, Benayoun S, Aprikian A, Batist G, Suissa S, et al. Use of statins and the risk of death in patients with prostate cancer. J Clin Oncol 2014;32:5–11.
    1. Nyame YA, Wilkins L, Greene DJ, Ganesan V, Dai C, Almassi N, et al. Assessing the relationship between statin use and oncologic outcomes among men electing active surveillance for localized prostate cancer. Prostate Cancer Prostatic Dis 2019;22:617–623.
    1. Cookson MS, Aus G, Burnett AL, Canby-Hagino ED, D’Amico AV, Dmochowski RR, et al. Variation in the definition of biochemical recurrence in patients treated for localized prostate cancer: the American Urological Association Prostate Guidelines for Localized Prostate Cancer Update Panel report and recommendations for a standard in the reporting of surgical outcomes. J Urol 2007;177:540–545.
    1. Allott EH, Howard LE, Cooperberg MR, Kane CJ, Aronson WJ, Terris MK, et al. Postoperative statin use and risk of biochemical recurrence following radical prostatectomy: results from the Shared Equal Access Regional Cancer Hospital (SEARCH) database. BJU Int 2014;114:661–666.
    1. Jeong IG, Lim B, Yun SC, Lim JH, Hong JH, Kim CS. Adjuvant low-dose statin use after radical prostatectomy: the PRO-STAT randomized clinical trial. Clin Cancer Res 2021;27:5004–5011.
    1. Scosyrev E, Tobis S, Donsky H, Wu G, Joseph J, Rashid H, et al. Statin use and the risk of biochemical recurrence of prostate cancer after definitive local therapy: a meta-analysis of eight cohort studies. BJU Int 2013;111(3 Pt B):E71–E77.
    1. Chao C, Jacobsen SJ, Xu L, Wallner LP, Porter KR, Williams SG. Use of statins and prostate cancer recurrence among patients treated with radical prostatectomy. BJU Int 2013;111:954–962.
    1. Meijer D, van Moorselaar RJA, Vis AN, Bijnsdorp IV. Prostate cancer development is not affected by statin use in patients with elevated PSA levels. Cancers (Basel) 2019;11:953
    1. Raval AD, Thakker D, Negi H, Vyas A, Kaur H, Salkini MW. Association between statins and clinical outcomes among men with prostate cancer: a systematic review and meta-analysis. Prostate Cancer Prostatic Dis 2016;19:151–162.
    1. Huynh LM, Keit E, Schuller AA, Carrillo RC, Huang E, Ahlering TE, et al. Impact of statin use on overall and time to biochemical failure following radical prostatectomy or radiation therapy. World J Urol 2021;39:3287–3293.
    1. Platz EA, Leitzmann MF, Visvanathan K, Rimm EB, Stampfer MJ, Willett WC, et al. Statin drugs and risk of advanced prostate cancer. J Natl Cancer Inst 2006;98:1819–1825.
    1. Peltomaa AI, Raittinen P, Talala K, Taari K, Tammela TLJ, Auvinen A, et al. Prostate cancer prognosis after initiation of androgen deprivation therapy among statin users. A population-based cohort study. Prostate Cancer Prostatic Dis 2021;24:917–924.
    1. Yang H, Pang L, Hu X, Wang W, Xu B, Zhang X, et al. The effect of statins on advanced prostate cancer patients with androgen deprivation therapy or abiraterone/enzalutamide: a systematic review and meta-analysis. J Clin Pharm Ther 2020;45:488–495.
    1. Jiménez-Vacas JM, Herrero-Aguayo V, Montero-Hidalgo AJ, Sáez-Martínez P, Gómez-Gómez E, León-González AJ, et al. Clinical, cellular, and molecular evidence of the additive antitumor effects of biguanides and statins in prostate cancer. J Clin Endocrinol Metab 2021;106:e696–e710.
    1. Tan XL, E JY, Lin Y, Rebbeck TR, Lu SE, Shang M, et al. Individual and joint effects of metformin and statins on mortality among patients with high-risk prostate cancer. Cancer Med 2020;9:2379–2389.
    1. Nasir A, Bullo MMH, Ahmed Z, Imtiaz A, Yaqoob E, Jadoon M, et al. Nutrigenomics: epigenetics and cancer prevention: a comprehensive review. Crit Rev Food Sci Nutr 2020;60:1375–1387.
    1. Deckelbaum RJ, Worgall TS, Seo T. n-3 fatty acids and gene expression. Am J Clin Nutr 2006;83 6 Suppl:1520S–1525S.
    1. Gu Z, Suburu J, Chen H, Chen YQ. Mechanisms of omega-3 polyunsaturated fatty acids in prostate cancer prevention. Biomed Res Int 2013;2013:824563
    1. Tur JA, Bibiloni MM, Sureda A, Pons A. Dietary sources of omega 3 fatty acids: public health risks and benefits. Br J Nutr 2012;107 Suppl 2:S23–S52.
    1. Kris-Etherton PM, Harris WS, Appel LJ. AHA Nutrition Committee; American Heart Association. Omega-3 fatty acids and cardiovascular disease: new recommendations from the American Heart Association. Arterioscler Thromb Vasc Biol 2003;23:151–152.
    1. Krauss-Etschmann S, Shadid R, Campoy C, Hoster E, Demmelmair H, Jiménez M, et al. Nutrition and Health Lifestyle (NUHEAL) Study Group. Effects of fish-oil and folate supplementation of pregnant women on maternal and fetal plasma concentrations of docosahexaenoic acid and eicosapentaenoic acid: a European randomized multicenter trial. Am J Clin Nutr 2007;85:1392–1400.
    1. Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol 2008;8:349–361.
    1. Wang W, Zhu J, Lyu F, Panigrahy D, Ferrara KW, Hammock B, et al. ω-3 polyunsaturated fatty acids-derived lipid metabolites on angiogenesis, inflammation and cancer. Prostaglandins Other Lipid Mediat 2014;113-115:13–20.
    1. Fradet V, Cheng I, Casey G, Witte JS. Dietary omega-3 fatty acids, cyclooxygenase-2 genetic variation, and aggressive prostate cancer risk. Clin Cancer Res 2009;15:2559–2566.
    1. Sikka S, Chen L, Sethi G, Kumar AP. Targeting PPARγ signaling cascade for the prevention and treatment of prostate cancer. PPAR Res 2012;2012:968040
    1. Hedelin M, Chang ET, Wiklund F, Bellocco R, Klint A, Adolfsson J, et al. Association of frequent consumption of fatty fish with prostate cancer risk is modified by COX-2 polymorphism. Int J Cancer 2007;120:398–405.
    1. Stefanska B, Rudnicka K, Bednarek A, Fabianowska-Majewska K. Hypomethylation and induction of retinoic acid receptor beta 2 by concurrent action of adenosine analogues and natural compounds in breast cancer cells. Eur J Pharmacol 2010;638:47–53.
    1. Salonia A, Gallina A, Briganti A, Suardi N, Capitanio U, Abdollah F, et al. Circulating estradiol, but not testosterone, is a significant predictor of high-grade prostate cancer in patients undergoing radical prostatectomy. Cancer 2011;117:5029–5038.
    1. Bonkhoff H, Berges R. The evolving role of oestrogens and their receptors in the development and progression of prostate cancer. Eur Urol 2009;55:533–542.
    1. Lee CK, Jeong SH, Jang C, Bae H, Kim YH, Park I, et al. Tumor metastasis to lymph nodes requires YAP-dependent metabolic adaptation. Science 2019;363:644–649.
    1. Terry PD, Rohan TE, Wolk A. Intakes of fish and marine fatty acids and the risks of cancers of the breast and prostate and of other hormone-related cancers: a review of the epidemiologic evidence. Am J Clin Nutr 2003;77:532–543.
    1. Brasky TM, Till C, White E, Neuhouser ML, Song X, Goodman P, et al. Serum phospholipid fatty acids and prostate cancer risk: results from the prostate cancer prevention trial. Am J Epidemiol 2011;173:1429–1439.
    1. Brasky TM, Darke AK, Song X, Tangen CM, Goodman PJ, Thompson IM, et al. Plasma phospholipid fatty acids and prostate cancer risk in the SELECT trial. J Natl Cancer Inst 2013;105:1132–1141.
    1. Chua ME, Sio MC, Sorongon MC, Morales ML 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:E333–E343.
    1. Hanson S, Thorpe G, Winstanley L, Abdelhamid AS, Hooper L. PUFAH group. Omega-3, omega-6 and total dietary polyunsaturated fat on cancer incidence: systematic review and meta-analysis of randomised trials. Br J Cancer 2020;122:1260–1270.
    1. Sorongon-Legaspi MK, Chua M, Sio MC, Morales M Jr. Blood level omega-3 fatty acids as risk determinant molecular biomarker for prostate cancer. Prostate Cancer 2013;2013:875615
    1. Torfadottir JE, Valdimarsdottir UA, Mucci LA, Kasperzyk JL, Fall K, Tryggvadottir L, et al. Consumption of fish products across the lifespan and prostate cancer risk. PLoS One 2013;8:e59799
    1. Aucoin M, Cooley K, Knee C, Fritz H, Balneaves LG, Breau R, et al. Fish-derived omega-3 fatty acids and prostate cancer: a systematic review. Integr Cancer Ther 2017;16:32–62.
    1. Zuniga KB, Zhao S, Kenfield SA, Cedars B, Cowan JE, Van Blarigan EL, et al. Trends in complementary and alternative medicine use among patients with prostate cancer. J Urol 2019;202:689–695.
    1. Alexander DD, Bassett JK, Weed DL, Barrett EC, Watson H, Harris W. Meta-analysis of long-chain omega-3 polyunsaturated fatty acids (LCω-3PUFA) and prostate cancer. Nutr Cancer 2015;67:543–554.
    1. Chua ME, Sio MC, Sorongon MC, Dy JS. 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;2012:826254
    1. Fu YQ, Zheng JS, Yang B, Li D. Effect of individual omega-3 fatty acids on the risk of prostate cancer: a systematic review and dose-response meta-analysis of prospective cohort studies. J Epidemiol 2015;25:261–274.
    1. Szymanski KM, Wheeler DC, Mucci LA. Fish consumption and prostate cancer risk: a review and meta-analysis. Am J Clin Nutr 2010;92:1223–1233.
    1. Lee KH, Seong HJ, Kim G, Jeong GH, Kim JY, Park H, et al. Consumption of fish and ω-3 fatty acids and cancer risk: an umbrella review of meta-analyses of observational studies. Adv Nutr 2020;11:1134–1149.
    1. Campbell RA, Li J, Malone L, Levy DA. Correlative analysis of vitamin D and omega-3 fatty acid intake in men on active surveillance for prostate cancer. Urology 2021;155:110–116.
    1. Liang P, Henning SM, Guan J, Grogan T, Elashoff D, Cohen P, et al. Effect of dietary omega-3 fatty acids on castrate-resistant prostate cancer and tumor-associated macrophages. Prostate Cancer Prostatic Dis 2020;23:127–135.

Metrics
Share
Tables

1 / 2

PERMALINK