Next Article in Journal
Exercise-Associated Hyponatremia During a Self-Paced Marathon Attempt in a 15-Year-Old Male Teenager
Previous Article in Journal
Distribution and Characteristics of Hypouricemia within the Japanese General Population: A Cross-Sectional Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Metabolic Alterations, Aggressive Hormone-Naïve Prostate Cancer and Cardiovascular Disease: A Complex Relationship

1
Department of Medical and Oral Sciences and Biotechnologies, G. D’Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
2
Department of Urological, Biomedical and Translational Sciences, Federiciana University, 87100 Cosenza, Italy
3
Institute of Molecular Genetics, National Research Council, Section of Chieti, 66100 Chieti, Italy
4
Urology and Andrology Section, Villa Immacolata Hospital, 01100 Viterbo, Italy
*
Author to whom correspondence should be addressed.
Medicina 2019, 55(3), 62; https://doi.org/10.3390/medicina55030062
Submission received: 5 October 2018 / Revised: 30 January 2019 / Accepted: 27 February 2019 / Published: 7 March 2019

Abstract

:
Background: Epidemiological studies suggest a possible relationship between metabolic alterations, cardiovascular disease and aggressive prostate cancer, however, no clear consensus has been reached. Objective: The aim of the study was to analyze the recent literature and summarize our experience on the association between metabolic disorders, aggressive hormone-naïve prostate cancer and cardiovascular disease. Method: We identified relevant papers by searching in electronic databases such as Scopus, Life Science Journals, and Index Medicus/Medline. Moreover, we showed our experience on the reciprocal relationship between metabolic alterations and aggressive prostate cancer, without the influence of hormone therapy, as well the role of coronary and carotid vasculopathy in advanced prostate carcinoma. Results: Prostate cancer cells have an altered metabolic homeostatic control linked to an increased aggressivity and cancer mortality. The absence of discrimination of risk factors as obesity, systemic arterial hypertension, diabetes mellitus, dyslipidemia and inaccurate selection of vascular diseases as coronary and carotid damage at initial diagnosis of prostate cancer could explain the opposite results in the literature. Systemic inflammation and oxidative stress associated with metabolic alterations and cardiovascular disease can also contribute to prostate cancer progression and increased tumor aggressivity. Conclusions: Metabolic alterations and cardiovascular disease influence aggressive and metastatic prostate cancer. Therefore, a careful evaluation of obesity, diabetes mellitus, dyslipidemia, systemic arterial hypertension, together with a careful evaluation of cardiovascular status, in particular coronary and carotid vascular disease, should be carried out after an initial diagnosis of prostatic carcinoma.

1. Introduction

Androgens play a key role in the development, growth and maintenance of prostate cells, as well as carcinogenesis and prostate cancer (PCa) progression [1,2]. Androgen dependence of PCa was first demonstrated in humans by Huggins in 1941 [3]. This assumption rationalized the “Historical myth” of androgen deprivation therapy for decades as a gold standard treatment in advanced and metastatic PCa [4,5]. After an average time of 12 to 33 months, despite the testosterone castration levels, PCa recovered and evolved towards a castration-resistant stage with limited effective treatment options [6]. Median survival of castration-resistant metastatic PCa was estimated not to exceed 25 months [7].
Metabolic alterations play a key role in manifesting this mechanism. Particularly PCa express profound metabolic reprogramming favoring biosynthesis processes and limiting catalytic mechanisms. Therefore, the metabolism of PCa represents a new therapeutic target and can offer new opportunities in the prevention and diagnosis of advanced PCa, independently from androgen ablation therapy [8,9,10,11].
Although numerous studies showed a possible association between metabolic disorders and aggressive PCa, the literature has often been limited and unambiguous.
Given that more and more epidemiological investigations showed that most patients with PCa died due to causes other than cancer, especially for cardiovascular events [12,13,14], the aim of the study was to analyze the recent literature and summarize our experience on the association between metabolic alterations and aggressive hormone-naïve PCa as well as the prevalence of cardiovascular damage, both at the initial diagnosis and in the advanced metastatic stage.

2. Materials and Methods

2.1. Identification of Studies

We identified significant recent papers in electronic databases as Scopus, Life Science Journals, and Index Medicus/Medline.
Studies were discovered using the following key words: prostate cancer, diabetes mellitus, dyslipidemia, obesity, systemic arterial hypertension, cardiovascular disease.

2.2. Inclusion Criteria

Working separately, reviewers selected all qualified studies in full text. To be included, articles had to (1) evaluate the association between aggressive PCa, metabolic alterations and/or cardiovascular disease; (2) contain an original data analysis and (3) from a peer reviewed journal. Articles were excluded if the clinical study (1) did not analyze a reciprocal relationship between PCa, metabolic alterations and/or cardiovascular disease (2) presented only as a case report or had an inappropriate design.

2.3. Our Experience

In the second part of article we finally focused and synthesized our personal experience regarding the relationship between aggressive hormone-naive PCa, metabolic disorders and cardiovascular damage.

3. Results and Discussion

3.1. Literature Review

3.1.1. Prostate Cancer and Diabetes Mellitus

The conversion of glucose to ATP, even in the presence of high levels of oxygen, occurs via the anaerobic effect (Warburg effect) in tumor cells due to the irreversible damage of the respiratory chain in the first phase of cancerogenesis. This results in ATP production at a rate 100 times faster than mitochondrial oxidation [8]. In the early stages of PCa, a widespread increase in lipogenesis directly coupled with glucose and glutamine metabolism is associated with PCa progression, worse prognosis and shorter survival [15,16].
Epidemiological studies show a conflicting association between Type 2 diabetes mellitus (DMII) and PCa. These inconsistent results are attributed to changes in insulin concentration during DMII development and progression. The risk of PCa, in particular, is associated with high insulin levels and insulin resistance. Consequently, it has hypothesized that the incidence rate of PCa increased in subjects with recent diagnosis of DMII, characterized by hyperglycemia and hyperinsulinaemia, and decreased with DMII progression [17,18,19]. Hyperinsulinaemia appears to be linked not only to a higher incidence of PCa, but also to an increased Gleason Score (GS) and cancer mortality, although not all studies are consistent [20,21,22,23,24,25,26].
The protect study showed that DMII was inversely associated only with differentiated PCa (Gleason Score 2–6), but not with poorly differentiated PCa [27]. Other studies, however, showed an inverse association between DMII and PCa for both high and low-grade tumors [22,28,29].
In the investigations in which an inverse relationship between DMII and PCa has been observed, this association appears to be more significant with the duration of DMII, but this does not seem to conform with all the studies that have examined the duration [28].The unanimity of the results was negatively affected by the absence of discrimination in diabetic status in clinical trials and mostly by inaccurate selection of comorbidities. This reflected a different impact of DMII on the incidence of PCa and in particular on tumor aggressivity [17,23,30,31,32]. Moreover, recent studies suggest that obesity, hypertension, DMII, hypercholesterolemia, in combination, can play a role in the development of high-grade PCa (GS ≥ 7) as well as in tumor progression [16,33]. Western diet with a high carbohydrate rate supports tumor progression in murine models and a high protein diet with a low-carbohydrate diet inhibits intratumoral androgen synthesis and castration-resistant cancer growth [34]. Therefore, it’s possible to hypothesize how a Mediterranean regimen characterized by a high consumption of vegetables, fruits, legumes, nuts, fish, can positively impact on a natural history of PCa and concomitant cardiovascular disease [35,36,37,38].
Moreover diet/genome interactions may influence the metabolic response to diet components and the susceptibility to PCa risk and progression, directly or indirectly altering gene expression or structure, while also modulating cell proliferation/apoptosis, DNA damage and repair, inflammation, oxidant/antioxidant balance and angiogenesis. Consequently, dietary interventions based on nutritional status, genotype analysis and “precision nutrition” can be used to establish specific nutritional guidelines to prevent or cure metabolic alterations and PCa [39,40,41].

3.1.2. Prostate Cancer and Dyslipidemia

Prostate cancer cells have an altered metabolic homeostatic control and increased endogenous cholesterol biosynthesis, with accumulation in tumor cells [42]. Cholesterol plays a key role in metastatic tumor progression by acting as a mediator in cell proliferation, inflammation and steroidogenesis [43,44].
Epidemiological and pre-clinical studies suggest that elevated levels of serum cholesterol could play a role in PCa progression through increased production of androgens by prostate cancer cells and activation of androgen receptors [42,45,46].
Cholesterol would then act as the precursor of intratumoral androgen biosynthesis [42,47]. Increased serum cholesterol levels were correlated with tumor volume, intratumoral testosterone levels and the expression of key steroid genes, such as CYP17A, suggesting that one of the intratumoral androgen sources would be represented by de novo steroid synthesis from circulating serum cholesterol [42]. Tumors in hypercholesterolemic environment also exhibits lower levels of apoptosis, increased activation of Akt kinase, related to aggressive PCa and increased vascularization [48,49]. Animal pre-clinical studies demonstrate that hypercholesterolemia was associated with increased PCa risk and metastatic progression [50]. In addition, increased cholesterol levels have been identified in PCa bone metastases, along with an increased expression of enzymes involved in steroidogenesis. In particular, the effects of CYP17A (which converts progestins into androgens), 17-keto-reductase (which converts androstenedione into testosterone) and 5α-reductase 1 (which converts testosterone into dihydrotestosterone) [51,52,53] can be seen in Figure 1.
Overall clinical trials reveal that hypercholesterolemia causes increased risk of PCa or aggressive disease, while statins reduce the risk of advanced disease [43,54,55,56].The mevalonate pathway was essential for the production of cholesterol and isoprenoids and had direct effects on the growth and progression of prostate cancer cells [57,58].The transcriptional regulation of the enzymes involved in this pathway occurs through the activation of SREBP-2, a regulator of androgenic synthesis which is itself regulated by androgens [59].The SREBP-2 expression increased during tumor progression, was significantly higher after castration and lost its inhibition feedback in tumor cells [55]. The drugs that most inhibited the Mevalonate pathway are nitrogen-containing bisphosphonates (N-BPs) and statins. Both have antitumor activity through effects on both bone tissue and cholesterol metabolism. In particular, observational studies show an inverse association between the use of statins and PCa progression, with a significant reduction in the risk of advanced disease with long-term use of statins, especially when used for more than 5 years and before tumor development [47,60,61,62,63,64]. These data suggest that an early approach based on the reduction of cholesterol levels before the onset of PCa could be an effective prevention and treatment strategy for PCa. In particular, prostate cancer cells increased the expression of Farnesil Pyrophosphate Synthetic Enzyme (FPPS), a key enzyme of the mevalonate pathway, detecting its correlation with PCa progression [65]. This enzyme is an important mediator of T cell activity essential for the elimination of tumor cells through the immune system [66]. FASN (Fatty Acid Synthase) catalyzed the synthesis of palmitate through the condensation of malonyl-CoA and acetyl-CoA. Its expression progressively increased during the natural history of PCa and reached maximum values in the androgen-independent PCa with bone metastases [67]. FASN inhibitors have been used to dysregulate lipid metabolism inhibiting PCa progression and causing cell death. FASN inhibition showed citotoxic and anti-proliferative activity in PCa cells, inducing apoptosis and reducing the lipid content in cancer cells in a concentration and time-dependent manner. FASN inhibitors induced degeneration and atrophy of prostatic tissue independently of the androgen status [68,69].

3.1.3. Prostate Cancer and Obesity

Adipose tissue is an active endocrine tissue capable of regulating prostate metabolic activity and influencing carcinogenesis and especially tumor progression [70,71,72,73,74,75].
The relationship between PCa and obesity, although subject to numerous studies, remains controversial at present. A possible explanation for this is due to the insufficient evaluation and control of possible concomitant metabolic factors associated with obesity such as systemic arterial hypertension (SAH), DMII, and dyslipidaemia, as well as a different design of the studies. Recent studies showed that the association between PCa and obesity could vary according to tumor grade, highlighting a direct relationship between adipose tissue and aggressive cancer and an inverse association with low-grade tumors [76,77,78,79,80,81]. However, not all studies were consistent and some clinical trials showed no relationship between obesity and tumor aggressiveness [82,83,84].
Leptin and adiponectin, adipokines produced by adipose tissue, regulated the action of Insulin on the glucose cellular uptake and reduced the metabolism of fatty acids. Leptin increased lipolysis, insulin sensitivity, inflammation and thromboembolism. In obesity in particular, leptin was increased and negatively affected cell differentiation, the risk of tumor progression, cell migration and prostate angiogenesis, by inducing pro-angiogenic factors [78,79,80,81,82,83,84,85,86].
Adiponectin, on the other hand, was reduced in obesity and adversely affected the histological grade and stage of PCa. In vitro studies showed that Adiponectin inhibited cell growth and proliferation in PCa and antagonized the proliferative effects of Leptin and IGF-1 in the androgen-independent PCa [87]. In addition to its effects on prostate cell proliferation, adipose tissue reduced the production of anti-inflammatory adipocytes such as adiponectin and increased the release of pro-inflammatory adipocytes such as leptin causing a chronic inflammatory state and prostatic tumor progression [6,74,78]. In particular, the expansion of adipose tissue caused the development of an inflammatory environment with increased secretion of cytokines such as IL-6, TNFα and MCP1. They in turn acted as attractants for further immune cells and created an environment that perpetuates immune infiltration and the production of inflammatory cytokines. Lipid inflammatory mediators such as arachidonic acid, eicosanoids, prostanoids and leukotrienes had also been increased [88]. Hormonal and inflammatory obesity-related alterations could therefore significantly contributed to prostate tumor growth and progression through the promotion of mitogenesis (Leptin, IGF-1, Insulin etc), angiogenesis (VEGF, IL6, IL8 etc) and tumor invasiveness (Leptin, IL6, etc.) [47,89].

3.1.4. Prostate Cancer and Systemic Arterial Hypertension

Only few studies have explored the association between SAH and PCa. Beebe-Dimmer et al. found a positive association between SAH and PCa [90]. Han et al. showed that diastolic pressure was positively associated with Prostate Specific Antigen (PSA) serum levels [91]. Subsequent studies, however, did not demonstrate a statistically significant association between SAH and PCa incidence [16]. On the other hand, a significant relationship between arterial SAH, advanced PCa and the risk of biochemical recurrence was demonstrated [92,93,94,95]. It has also been showed that patients with PCa with SAH and overweight had a significantly lower survival time than control subjects [94]. Preclinical evidence showed that the use of Beta Blockers could affect PCa progression; in particular, sympathetic nerve stimulation was able to induce metastases in prostate tumor models and the administration of beta blockers was capable to prevent the effect of adrenergic stimulation on the promotion of metastases [96,97]. The main mediator of these effects was the β-adrenergic receptor, which was also implicated in the tumor-immune system response [98]. Moreover, recent clinical trials confirmed these findings in humans by demonstrating that the use of Beta Blockers was associated with reduced mortality in patients with high-risk or metastatic PCa at the time of diagnosis [99,100].

3.2. Personal Experience

3.2.1. Obesity and Aggressive Prostate Cancer

In our experience, similar to the most recent literature, we showed that obesity was associated, both individually and in combination with cardiovascular risk factors, with aggressive PCa. A reverse association was instead observed between obesity and non-aggressive PCa, at initial diagnosis [101]. In particular, obese patients with early PCa had a higher risk of developing high-grade PCa compared to non-obese patients with PCa [101,102,103]. Several hypotheses could explain the association between obesity and high-grade PCa. The literature suggested that obesity could be associated with biological changes (e.g., inflammation, insulin resistance, angiogenesis, cell migration) and modification of adipokine levels related to aggressive tumor phenotype. It had also been reported that Adiponectin was involved in the regulation of metabolic homeostasis (in particular regulation of glucidic and lipid metabolism), inhibition of inflammation, atherogenesis, angiogenesis and cell migration [104,105,106].
Adiponectin levels were reduced in obesity, coronary heart disease and tumors such as PCa, with a negative association with histology and cancer stage [76,107]. In vitro, adiponectin inhibited cell growth and proliferation in the prostate, while in androgen-independent cancer tumor cells antagonized the proliferative effects of leptin and IGF-1 [85].
Leptin act directly on prostate cancer cells by affecting steroid activity, cell cycle regulation, and insulin activity [108]. In particular, Leptin increased lipolysis, insulin sensitivity, inflammation and thromboembolism [104]. It influenced cellular differentiation and cancer progression, cell migration, tumor angiogenesis and promoted advanced PCa through induction and activation of pro-angiogenic factors [78,85,86,109,110].
It was hypothesized that low adiponectin levels and high levels of leptin and resistin observed in obese patients could be involved in prostate cancer aggressiveness. Recent studies confirmed these hypotheses, showing that high levels of leptin could be correlated with a worse grade and tumor stage, while high levels of adiponectin were associated with a less aggressive tumor grade and stage [78,81,111]. This suggests that low levels of adiponectin and high levels of leptin observed in obese patients could be related with the development of high-grade PCa.
In vitro studies confirmed these results showing that specific genetic polymorphisms in leptin and adiponectin as their receptors were associated with PCa proliferation and progression, through increased inflammation, angiogenesis, worse pathological grade, metabolic syndrome, closely associated with aggressive PCa [111,112,113].

3.2.2. Obesity, Diabetes Mellitus and Aggressive Prostate Cancer

Obesity was a well-known risk factor for DMII. Since obesity was a factor involved both in the development of DMII and in the aggressive PCa, we analyzed the possibility of an association between DM and aggressive PCa hormone-naïve and demonstrated that DMII was associated with high-grade PCa, but only in obese subjects [101]. These observations agreed with the data reported in recent studies and suggested that DMII could be associated with aggressive PCa, but only in patients with concomitant obesity, whereas DMII wasn’t associated with high-grade PCa in non-obese patients [23,31,114]. Recent data from 119315 subjects with DM in order to examine the relationship between Metformin exposure and PCa risk indicate that there is no association between the use of Metformin and PCa respect to tumor grade [115]. However, the effects of antidiabetic agents in PCa development, aggressiveness and progression remain unclear. Specifically, the literature reports a conflicting relationship between the use of Metformin and PCa, demonstrating a reduced or increased risk or no association [116,117,118,119,120,121,122,123]. It is possible to hypothesize that a combination of biological factors may be responsible for the discrepant association between DMII and high-grade PCa. The link between obesity, DMII and PCa may be related to insulin resistance, hyperinsulinemia, reduced IGFBP (Insulin-like Growth Factor Binding Proteins), increased bioavailability of IGF-1 (Insulin-like Growth Factor-1), steroid hormones (reduced levels of androgens, increased estrogen Levels) and inflammatory markers [124,125,126]. In particular, hyperinsulinemia promoted prostatic cancer aggressiveness and progression [127]. In addition, insulin secretory capacity was higher in obese patients with DMII than non-obese patients with DMII [125]. DMII and obesity were associated with low serum testosterone levels and increased estrogen levels, correlated with high-grade PCa [23]. Some reports have suggested that low levels of testosterone were associated with aggressive PCa, inadequate treatment after radical prostatectomy, high-grade PCa (Gleason Score 8–10) and locally advanced prostate cancer (pT3 and pT4) [128,129,130,131]. These observations suggest a possible role of sex steroid hormones in the contribution of obesity and DMII in high-grade PCa.
Systemic inflammation can also contribute to increased tumor aggressivity in obesity and DMII. DMII and obesity are in fact inflammatory conditions associated with increased cytokine production such as Tumor necrosis factor (TNFα), Interleukin (IL) 6 and IL8 [132]. These cytokines stimulate the Nuclear Factor KappaB pathway (NF-kB), directly related to lymph node invasion and androgen-independent progression in PCa [133,134]. We have previously reported an important role of inflammation and the immune system as regulators of physiology and pathology of PCa [101,134]. Chronic inflammation and oxidative stress associated with both DMII and obesity could thus contribute to PCa development and progression [135,136].

3.2.3. Systemic Arterial Hypertension, Dyslipidemia and Aggressive Prostate Cancer

We analyzed the association between arterial hypertension and PCa in patients with PCa hormone naïve both at the initial diagnosis [103] and in the advanced metastatic phase [137]. We showed that the presence of arterial hypertension and obesity were significantly associated with aggressive PCa (OR 2.84, p < 0.05). In non-obese patients, however, no relationship was established between high blood pressure and PCa.
Previous studies have analyzed the association between arterial hypertension and PCa, but the results were conflicting, demonstrating either reduced risk or a positive association [93,138,139,140]. Similarly, the use of antihypertensive drugs as beta-blockers was linked to both decreased risk of PCa and cancer mortality as well as to an increased risk of PCa progression and death, showing inconclusive results [100,141,142,143].
Pathogenetic mechanisms that potentially link obesity and hypertension to PCa aggressivity are not known. It is possible to hypothesize that the link between obesity, arterial hypertension and aggressive PCa has a close relationship with common biological mechanisms such as insulin resistance, hyperinsulinemia and inflammation [144]. Chronic inflammation, reactive oxygen species and the oxidative stress associated with both hypertension and obesity could contribute to tumor progression [145,146,147]. In our experience, a statistically significant association between arterial hypertension and advanced metastatic PCa was demonstrated with an OR of 4.5. In the same study we also found that hypercholesterolemia, particularly high levels of total cholesterol and LDL-C, were significantly associated with aggressive metastatic PCa with an OR of 3.28 [137]. Recent studies indicated that hypertension, hypercholesterolemia, atherosclerosis, and a composite score of metabolic factors were associated with advanced PCa, biochemical recurrence and increased tumor mortality [28,93,139,148,149,150,151], as shown in Figure 2.
In our experience and consistent with these studies, we first highlighted a significant association between arterial hypertension (in particular systolic arterial hypertension), hypercholesterolaemia and coronary and carotid vascular disease in patients with advanced PCa and concomitant bone metastases [136].
If confirmed in larger studies, our research suggests that hypertension, hypercholesterolemia and atherosclerosis could be considered as “New Players” in advanced metastatic PCa and confirm common etiopathogenetic mechanisms, as shown in Figure 3. A study by Thysel et al., analyzed metabolites associated with PCa metastases and in particular identified high levels of cholesterol in bone metastases. The authors proposed that prostate cancer cells synthesize cholesterol de novo as well as the involvement of this metabolite through the surrounding environment [152], as shown in Figure 1.

3.2.4. Metabolic Syndrome and Aggressive Prostate Cancer

The metabolic syndrome (MetS) is characterized by a cluster of comorbidities as visceral obesity, hyperglycemia, systemic arterial hypertension and dyslipidaemia. Epidemiological Studies showed conflicting results on the association between MetS and aggressive PCa [153,154,155,156].
Overall a lower survival time, shorter time to castration-resistant Pca, increased prostate cancer-specific mortality, and biochemical recurrence were related to MetS [153,157].
Our group firstly investigate the association between MetS and high-grade PCa without the influence of endocrine therapy, showing a significant relationship between MetS and aggressive PCa at initial diagnosis (OR 1.87, p < 0.05) and a reduced risk of low-grade PCa (OR 0.53, p < 0.05). Moreover, fasting glucose, triglycerides, waist circumference, systemic arterial hypertension were significantly associated with high-grade PCa [158].
The metabolic alterations, chronic inflammation, oxidative stress associated to MetS may contribute to PCa progression causing a chronic inflammatory condition that predisposes to aggressive and metastatic PCa [145,146,147].

3.2.5. Hormone-naive Prostate Cancer and Vascular Damage

Epidemiological studies showed that cardiovascular disease is one of the major causes of death in patients with PCa (30%) [159,160,161]. In our experience we analyzed the relationship between PCa hormone-naive and carotid and coronary vascular disease, both at initial diagnosis of PCa [102,162] and metastatic cancer [136]. In particular, we showed a significant association between bone metastases and vascular pathology in advanced metastatic PCa with an OR of 3.8.
Consistent with our results, it was shown by Zoller et al. that the risk of coronary artery disease augmented in the first 6 months after the diagnosis of cancer and in the specific case of PCa this risk remained high for over 10 years. Furthermore, the risk of coronary artery disease was correlated with the presence of metastases and cardiovascular risk factors [163]. Moreover, we found that 23% of patients with hormone naïve localized PCa presented carotid vasculopathy (OR 2.43, p < 0.05), whereas 17% of patients had a history of symptomatic coronary artery disease (OR 1.88, P < 0.05). However, a limited number of studies explored the relationship between atherosclerotic vascular disease and PCa. Some studies showed no association between atherosclerotic disease and PCa; while others, similarly to our results, documented a possible relationship [164,165,166,167]. Consistent with our research in a large prospective study, a high prevalence of cardiovascular risk factors and cardiovascular disease (27%) has been demonstrated in PCa before ADT has begun. In particular, 19% and 8% of patients had cardiac ischemic disease and cerebrovascular disease, respectively [168]. Similarly, in the study REDUCES, the presence of coronary artery disease was associated with an increased risk of PCa by 35% [169]. Similar results were obtained from Stamatiou et al. in an autopsy study, as evidenced by the fact that patients with PCa had a greater risk of developing advanced coronary artery disease than those without PCa [166]. Although there are no other studies that have assessed the presence of carotid vascular damage in patients with PCa hormone naïve at initial diagnosis, the results of our research are also consistent with a prospective study of Pereira et al. which showed that myointimal carotid thickness was high at baseline in patients with PCa. In addition, another endothelial damage marker, represented by VCAM-1 levels, was reduced in PCa patients after radiotherapy.
The authors hypothesize that the inflammatory process associated with neoplasia may have been reduced with the treatment, despite the potential adverse effects of irradiation on vascularization.
It has been widely accepted in international literature that cancer cells can cause inflammatory conditions characterized by high levels of inflammatory biomarkers such as reactive protein C, prostaglandins, adhesion molecules, etc. [134,170,171]. If confirmed in other studies, the results of our research suggest that CAD and CVD may represent new risk factors in PCa and suggest common etiopathogenetic factors.
Atherosclerotic cardiovascular pathology and PCa could share common etiopathogenetic mechanisms. Therefore, we hypothesized that they could be present in patients with PCa from initial cancer diagnosis even in the absence of hormone therapy [162] and could be associated with subsequent phases of PCa progression to a more aggressive and metastatic phenotype [102,137]. Since inflammation was a factor that accompanies both atherosclerosis and activation of hemostasis and tumor pathology, a possible relationship between inflammation, PCa, hemostatic activation and vascular damage has been hypothesized. Based on this hypothesis, therefore, effective tumor treatment could reduce tumor volume, associated inflammation, hemostatic activation and hence the risk of vasculopathy [163].

4. Conclusions

Recent literature and our research showed that metabolic alterations and coronary and carotid vascular disease influenced aggressive and metastatic PCa. Therefore, a careful evaluation of cardiovascular risk factors should be carried out from the initial diagnosis of PCa, in particular focusing on risk factors such as obesity, arterial hypertension and dyslipidaemia, together with a careful evaluation of the concomitant presence of cardiovascular disease.
Overall, the results of our research add further motivation to control metabolic factors in PCa from initial cancer diagnosis, even in the absence of hormone therapy, in order to reduce cardiovascular risk and tumor progression. In addition, our results suggest that increased oxidative stress and a permanent inflammatory state may predispose to a more aggressive and metastatic tumor phenotype. Metabolic alterations can in fact create a favorable environment for tumor progression and cause an inflammatory state that predisposes to vascular disease and PCa.
On the other hand, chronic inflammation and oxidative stress associated with metabolic damage and atherosclerosis may also contribute to tumor progression, causing premature vascular and prostatic damage.
The metabolic and cardiovascular profile evaluation at initial diagnosis of PCa, particularly dyslipidemia, glucose intolerance, obesity, metabolic syndrome, might be used to discover new markers for PCa monitoring and more importantly to identify novel personalized integrative therapies targeting critical metabolic changes that sustain proliferation, migration, and invasion of PCa cells.

Author Contributions

Conception and design of the study, S.D.F.; acquisition and analysis of data S.D.F., I.R., M.C., G.G., D.F.; supervision and validation of Data A.M. and E.T.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Toivanen, R.; Shen, M.M. Prostate organogenesis: Tissue induction, hormonal regulation and cell type specification. Development 2017, 144, 1382–1398. [Google Scholar] [CrossRef] [PubMed]
  2. Izumi, K.; Shigehara, K.; Nohara, T.; Narimoto, K.; Kadono, Y.; Mizokami, A. Both High and Low Serum Total Testosterone Levels Indicate Poor Prognosis in Patients with Prostate Cancer. Anticancer Res. 2017, 37, 5559–5564. [Google Scholar] [PubMed]
  3. 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]
  4. Krakowsky, Y.; Morgentaler, A. Risk of Testosterone Flare in the Era of the Saturation Model: One More Historical Myth. Eur. Urol. Focus. 2019, 5, 81–89. [Google Scholar] [CrossRef] [PubMed]
  5. Morgentaler, A. Testosterone and prostate cancer: An historical perspective on a modern myth. Eur. Urol. 2006, 50, 935–939. [Google Scholar] [CrossRef] [PubMed]
  6. Gunter, J.H.; Sarkar, P.L.; Lubik, A.A.; Nelson, C.C. New players for advanced prostate cancer and the rationalisation of insulin-sensitising medication. Int. J. Cell Biol. 2013, 2013. [Google Scholar] [CrossRef] [PubMed]
  7. Ryan, C.J.; Smith, M.R.; de Bono, J.S.; Molina, A.; Logothetis, C.; de Souza, P.; Fizazi, K.; Mainwaring, P.; Piulats, J.M.; Ng, S.; et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N. Engl. J. Med. 2013, 368, 138–148. [Google Scholar] [CrossRef] [PubMed]
  8. Benedettini, E.; Nguyen, P.; Loda, M. The pathogenesis of prostate cancer: From molecular to metabolic alterations. Diagn. Histopathol. 2008, 14, 195–201. [Google Scholar] [CrossRef] [PubMed]
  9. Fritz, V.; Benfodda, Z.; Henriquet, C.; Hure, S.; Cristol, J.P.; Michel, F.; Carbonneau, M.A.; Casas, F.; Fajas, L. Metabolic intervention on lipid synthesis converging pathways abrogates prostate cancer growth. Oncogene 2013, 32, 5101–5110. [Google Scholar] [CrossRef] [PubMed]
  10. Itkonen, H.M.; Brown, M.; Urbanucci, A.; Tredwell, G.; Ho Lau, C.; Barfeld, S.; Hart, C.; Guldvik, I.J.; Takhar, M.; Heemers, H.V.; et al. Lipid degradation promotes prostate cancer cell survival. Oncotarget 2017, 8, 38264–38275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Sidaway, P. Prostate cancer: Targeting lipid metabolism. Nat. Rev. Urol. 2017, 14. [Google Scholar] [CrossRef] [PubMed]
  12. Ng, H.S.; Koczwara, B.; Order, D.; Vitry, A. Development of comorbidities in men with prostate cancer treated with androgen deprivation therapy: An Australian population-based cohort study. Prostate Cancer Prostatic Dis. 2018, 21, 403–410. [Google Scholar] [CrossRef] [PubMed]
  13. Wallis, C.J.; Mahar, A.L.; Satkunasivam, R.; Herschorn, S.; Kodama, R.T.; Lee, Y.; Kulkarni, G.S.; Narod, S.A.; Nam, R.K. Cardiovascular and Skeletal-related Events Following Localized Prostate Cancer Treatment: Role of Surgery, Radiotherapy, and Androgen Deprivation. Urology 2016, 97, 145–152. [Google Scholar] [CrossRef] [PubMed]
  14. Abdollah, F.; Sammon, J.D.; Reznor, G.; Sood, A.; Schmid, M.; Klett, D.E.; Sun, M.; Aizer, A.A.; Choueiri, T.K.; Hu, J.C.; et al. Medical androgen deprivation therapy and increased non-cancer mortality in non-metastatic prostate cancer patients aged ≥66 years. Eur. J. Surg. Oncol. 2015, 41, 1529–1539. [Google Scholar] [CrossRef] [PubMed]
  15. Menendez, J.A.; Lupu, R. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat. Rev. Cancer 2007, 7, 763–777. [Google Scholar] [CrossRef] [PubMed]
  16. Arthur, R.; Møller, H.; Garmo, H.; Holmberg, L.; Stattin, P.; Malmstrom, H.; Lambe, M.; Hammar, N.; Walldius, G.; Robinson, D.; et al. Association between baseline serum glucose, triglycerides and total cholesterol, and prostate cancer risk categories. Cancer Med. 2016, 5, 1307–1318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Bensimon, L.; Yin, H.; Suissa, S.; Pollak, M.N.; Azoulay, L. Type 2 diabetes and the risk of mortality among patients with prostate cancer. Cancer Causes Control 2014, 25, 329–338. [Google Scholar] [CrossRef] [PubMed]
  18. Rodriguez, C.; Patel, A.V.; Mondul, A.M.; Jacobs, E.J.; Thun, M.J.; Calle, E.E. Diabetes and risk of prostate cancer in a prospective cohort of US men. Am. J. Epidemiol. 2005, 161, 147–152. [Google Scholar] [CrossRef] [PubMed]
  19. Haring, A.; Murtola, T.J.; Talala, K.; Taari, K.; Tammela, T.L.; Auvinen, A. Antidiabetic drug use and prostate cancer risk in the Finnish Randomized Study of Screening for Prostate Cancer. Scand. J. Urol. 2017, 51, 5–12. [Google Scholar] [CrossRef] [PubMed]
  20. Barone, B.B.; Yeh, H.C.; Snyder, C.F.; Peairs, K.S.; Stein, K.B.; Derr, R.L.; Wolff, A.C.; Brancati, F.L. Long-term All-Cause Mortality in Cancer Patients with Preexisting Diabetes Mellitus: A Systematic Review and Meta-analysis. JAMA 2008, 300, 2754–2764. [Google Scholar] [CrossRef] [PubMed]
  21. Pierce, B.; Plymate, S.; Ostrander, E.; Stanford, J. Diabetes mellitus and prostate cancer risk. Prostate 2008, 68, 1126–1132. [Google Scholar] [CrossRef] [PubMed]
  22. Gong, Z.; Neuhouser, M.L.; Goodman, P.J.; Albanes, D.; Chi, C.; Hsing, A.W.; Lippman, S.M.; Platz, E.A.; Pollak, M.N.; Thompson, I.M.; et al. Obesity, Diabetes, and Risk of Prostate Cancer: Results from the Prostate Cancer Prevention Trial. Cancer Epidemiol. Prev. Biomark. 2006, 15, 1977–1983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Moreira, D.M.; Anderson, T.; Gerber, L.; Thomas, J.A.; Bañez, L.L.; McKeever, M.G.; Hoyo, C.; Grant, D.; Jayachandran, J.; Freedland, S.J. The association of diabetes mellitus and high-grade prostate cancer in a multiethnic biopsy series. Cancer Cause Control 2011, 22, 977–983. [Google Scholar] [CrossRef] [PubMed]
  24. Kang, J.; Chen, M.H.; Zhang, Y.; Moran, B.J.; Dosoretz, D.E.; Katin, M.J.; Braccioforte, M.H.; Salenius, S.A.; D’Amico, A.V. Type of diabetes mellitus and the odds of Gleason score 8 to 10 prostate cancer. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, 463–467. [Google Scholar] [CrossRef] [PubMed]
  25. Karlin, N.J.; Amin, S.B.; Verona, P.M.; Kosiorek, H.E.; Cook, C.B. Co-Existing Prostate Cancer and Diabetes Mellitus: Implications for Patient Outcomes and Care. Endocr. Pract. 2017, 23, 816–821. [Google Scholar] [CrossRef] [PubMed]
  26. Krönig, M.; Haverkamp, C.; Schulte, A.; Heinicke, L.; Schaal, K.; Drendel, V.; Werner, M.; Wetterauer, U.; Schultze-Seemann, W.; Jilg, C.A. Diabetes and beta-adrenergic blockage are risk factors for metastatic prostate cancer. World J. Surg. Oncol. 2017, 15. [Google Scholar] [CrossRef] [PubMed]
  27. Xu, H.; Jiang, H.W.; Ding, G.X.; Zhang, H.; Zhang, L.M.; Mao, S.H.; Ding, Q. Diabetes mellitus and prostate cancer risk of different grade or stage: A systematic review and meta-analysis. Diabetes Res. Clin. Pract. 2013, 99, 241–249. [Google Scholar] [CrossRef] [PubMed]
  28. Calton, B.A.; Chang, S.C.; Wright, M.E.; Kipnis, V.; Lawson, K.; Thompson, F.E.; Subar, A.F.; Mouw, T.; Campbell, D.S.; Hurwitz, P.; et al. History of diabetes mellitus and subsequent prostate cancer risk in the NIH-AARP diet and health study. Cancer Cause Control 2007, 18, 493–503. [Google Scholar] [CrossRef] [PubMed]
  29. Ozbek, E.; Otunctemur, A.; Dursun, M.; Sahin, S.; Besiroglu, H.; Koklu, I.; Erkoc, M.; Danis, E.; Bozkurt, M. Diabetes mellitus and HbA1c levels associated with high grade prostate cancer. Asian Pac. J. Cancer Prev. 2014, 15, 2555–2558. [Google Scholar] [CrossRef] [PubMed]
  30. Fukushima, H.; Masuda, H.; Kawakami, S.; Ito, M.; Sakura, M.; Numao, N.; Koga, F.; Saito, K.; Fujii, Y.; Yamamoto, S.; et al. Effect of diabetes mellitus on high-grade prostate cancer detection among Japanese obese patients with prostate-specific antigen less than 10 ng/mL. Urology 2012, 79, 1329–1334. [Google Scholar] [CrossRef] [PubMed]
  31. Daousi, C.; Casson, I.F.; Gill, G.V.; MacFarlane, I.A.; Wilding, J.P.H.; Pinkney, J.H. Prevalence of obesity in type 2 diabetes in secondary care: Association with cardiovascular risk factors. Postgrad. Med. J. 2006, 82, 280–284. [Google Scholar] [CrossRef] [PubMed]
  32. Wallner, L.P.; Morgenstern, H.; McGree, M.E.; Jacobson, D.J.; St Sauver, J.L.; Jacobsen, S.J.; Sarma, A.V. The effects of metabolic conditions on prostate cancer incidence over 15 years of follow-up: Results from the Olmsted County Study. BJU Int. 2011, 107, 929–935. [Google Scholar] [CrossRef] [PubMed]
  33. Fokidis, H.B.; Yieng Chin, M.; Ho, V.W.; Adomat, H.H.; Soma, K.K.; Fazli, L.; Nip, K.M.; Cox, M.; Krystal, G.; Zoubeidi, A.; et al. A low carbohydrate, high protein diet suppresses intratumoral androgen synthesis and slows castration-resistant prostate tumor growth in mice. J. Steroid Biochem. Mol. Biol. 2015, 150, 35–45. [Google Scholar] [CrossRef] [PubMed]
  34. Urquiza-Salvat, N.; Pascual-Geler, M.; Lopez-Guarnido, O.; Rodrigo, L.; Martinez-Burgos, A.; Cozar, J.M.; Ocaña-Peinado, F.M.; Álvarez-Cubero, M.J.; Rivas, A. Adherence to Mediterranean diet and risk of prostate cancer. Aging Male 2018, 15, 1–7. [Google Scholar] [CrossRef] [PubMed]
  35. Schwingshackl, L.; Schwedhelm, C.; Galbete, C.; Hoffmann, G. Adherence to mediterranean diet and risk of cancer: An updated systematic review and meta-analysis. Nutrients 2017, 9, 1063. [Google Scholar] [CrossRef] [PubMed]
  36. Mattioli, A.V.; Palmiero, P.; Manfrini, O.; Puddu, P.E.; Nodari, S.; Dei Cas, A.; Mercuro, G.; Scrutinio, D.; Palermo, P.; Sciomer, S.; et al. Mediterranean diet impact on cardiovascular diseases: A narrative review. J. Cardiovasc. Med. 2017, 18, 925–935. [Google Scholar] [CrossRef] [PubMed]
  37. Castelló, A.; Boldo, E.; Amiano, P.; Castaño-Vinyals, G.; Aragonés, N.; Gómez-Acebo, I.; Peiró, R.; Jimenez-Moleón, J.J.; Alguacil, J.; Tardón, A.L.; et al. MCC-Spain Researchers. Mediterranean Dietary Pattern is Associated with Low Risk of Aggressive Prostate Cancer: MCC-Spain Study. J. Urol. 2018, 199, 430–437. [Google Scholar] [CrossRef] [PubMed]
  38. Yuan, Y.; Ferguson, L.R. Nutrigenetics and prostate cancer: 2011 and beyond. Lifestyle Genom. 2011, 4, 121–136. [Google Scholar] [CrossRef] [PubMed]
  39. Bishop, K.S.; Erdrich, S.; Karunasinghe, N.; Han, D.Y.; Zhu, S.; Jesuthasan, A.; Ferguson, L.R. An investigation into the association between DNA damage and dietary fatty acid in men with prostate cancer. Nutrients 2015, 7, 405–422. [Google Scholar] [CrossRef] [PubMed]
  40. Ramos-Lopez, O.; Milagro, F.I.; Allayee, H.; Chmurzynska, A.; Choi, M.S.; Curi, R.; De Caterina, R.; Ferguson, L.R.; Goni, L.; Kang, J.X.; et al. Guide for Current Nutrigenetic, Nutrigenomic, and Nutriepigenetic Approaches for Precision Nutrition Involving the Prevention and Management of Chronic Diseases Associated with Obesity. Lifestyle Genom. 2017, 10, 43–62. [Google Scholar] [CrossRef] [PubMed]
  41. Mostaghel, E.A.; Solomon, K.R.; Pelton, K.; Freeman, M.R.; Montgomery, R.B. Impact of circulating cholesterol levels on growth and intratumoral androgen concentration of prostate tumors. PLoS ONE 2012, 7. [Google Scholar] [CrossRef] [PubMed]
  42. Pelton, K.; Freeman, M.R.; Solomon, K.R. Cholesterol and prostate cancer. Curr. Opin. Pharmacol. 2012, 12, 751–759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Galbraith, L.; Leung, H.Y.; Ahmad, I. Lipid pathway deregulation in advanced prostate cancer. Pharmacol. Res. 2018, 131, 177–184. [Google Scholar] [CrossRef] [PubMed]
  44. Platz, E.; Till, C.; Goodman, P.J.; Parnes, H.L.; Figg, W.D.; Albanes, D.; Neuhouser, M.L.; Klein, E.A.; Thompson, I.M., Jr.; Kristal, A.R. Men with low serum cholesterol have a lower risk of high-grade prostate cancer in the placebo arm of the prostate cancer prevention trial. Cancer Epidemiol. Biomark. Prev. 2009, 18, 2807–2813. [Google Scholar] [CrossRef] [PubMed]
  45. Leon, C.G.; Locke, J.A.; Adomat, H.H.; Etinger, S.L.; Twiddy, A.L.; Neumann, R.D.; Nelson, C.C.; Guns, E.S.; Wasan, K.M. Alterations in cholesterol regulation contribute to the production of intratumoral androgens during progression to castration-resistant prostate cancer in a mouse xenograft model. Prostate 2010, 70, 390–400. [Google Scholar] [CrossRef] [PubMed]
  46. Allott, E.H.; Masko, E.M.; Freedland, S.J. Obesity and prostate cancer: Weighing the evidence. Eur. Urol. 2013, 63, 800–809. [Google Scholar] [CrossRef] [PubMed]
  47. Todenhöfer, T.; Hennenlotter, J.; Kühs, U.; Gerber, V.; Gakis, G.; Vogel, U.; Aufderklamm, S.; Merseburger, A.; Knapp, J.; Stenzl, A.; et al. Altered expression of farnesyl pyrophosphate synthase in prostate cancer: Evidence for a role of the mevalonate pathway in disease progression? World J. Urol. 2013, 31, 345–350. [Google Scholar] [CrossRef] [PubMed]
  48. Shimizu, Y.; Segawa, T.; Inoue, T.; Shiraishi, T.; Yoshida, T.; Toda, Y.; Yamada, T.; Kinukawa, N.; Terada, N.; Kobayashi, T.; et al. Increased Akt and phosphorylated Akt expression are associated with malignant biological features of prostate cancer in Japanese men. BJU Int. 2007, 100, 685–690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Llaverias, G.; Danilo, C.; Wang, Y.; Witkiewicz, A.K.; Daumer, K.; Lisanti, M.P.; Frank, P.G. A Western-type diet accelerates tumor progression in an autochthonous mouse model of prostate cancer. Am. J. Pathol. 2010, 177, 3180–3191. [Google Scholar] [CrossRef] [PubMed]
  50. Locke, J.A.; Guns, E.S.; Lehman, M.L.; Ettinger, S.; Zoubeidi, A.; Lubik, A.; Margiotti, K.; Fazli, L.; Adomat, H.; Wasan, K.M.; et al. Arachidonic acid activation of intratumoral steroid synthesis during prostate cancer progression to castration resistance. Prostate 2010, 70, 239–251. [Google Scholar] [CrossRef] [PubMed]
  51. Montgomery, R.B.; Mostaghel, E.A.; Vessella, R.; Hess, D.L.; Kalhorn, T.F.; Higano, C.S.; True, L.D.; Nelson, P.S. Maintenance of intratumoral androgens in metastatic prostate cancer: A mechanism for castration-resistant tumor growth. Cancer Res. 2008, 68, 4447–4454. [Google Scholar] [CrossRef] [PubMed]
  52. Dillard, P.R.; Lin, M.F.; Khan, S.A. Androgen-independent prostate cancer cells acquire the complete steroidogenic potential of synthesizing testosterone from cholesterol. Mol. Cell Endocrinol. 2008, 295, 115–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Schnoeller, T.J.; Jentzmik, F.; Schrader, A.J.; Steinestel, J. Influence of serum cholesterol level and statin treatment on prostate cancer aggressiveness. Oncotarget 2017, 8, 47110–47120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Allott, E.H.; Arab, L.; Su, L.J.; Farnan, L.; Fontham, E.T.; Mohler, J.L.; Bensen, J.T.; Steck, S.E. Saturated fat intake and prostate cancer aggressiveness: Results from the population-based North Carolina-Louisiana Prostate Cancer Project. Prostate Cancer Prostatic Dis. 2017, 20, 48–54. [Google Scholar] [CrossRef] [PubMed]
  55. Allott, E.H.; Farnan, L.; Steck, S.E.; Arab, L.; Su, L.J.; Mishel, M.; Fontham, E.T.; Mohler, J.L.; Bensen, J.T. Statin Use and Prostate Cancer Aggressiveness: Results from the Population-Based North Carolina-Louisiana Prostate Cancer Project. Cancer Epidemiol. Biomark. Prev. 2016, 25, 670–677. [Google Scholar] [CrossRef] [PubMed]
  56. Roy, M.; Kung, H.J.; Ghosh, P.M. Statins and prostate cancer: Role of cholesterol inhibition versus prevention of small GTP binding proteins. Am. J. Cancer Res. 2011, 1, 542–561. [Google Scholar] [PubMed]
  57. Russell, R.G. Bisphosphonates: The first 40 years. Bone 2011, 49, 2–19. [Google Scholar] [CrossRef] [PubMed]
  58. Eberlé, D.; Hegarty, B.; Bossard, P.; Ferré, P.; Foufelle, F. SREBP transcription factors: Master regulators of lipid homeostasis. Biochimie 2004, 86, 839–848. [Google Scholar] [CrossRef] [PubMed]
  59. Ettinger, S.L.; Sobel, R.; Whitmore, T.G.; Akbari, M.; Bradley, D.R.; Gleave, M.E.; Nelson, C.C. Dysregulation of sterol response element-binding proteins and downstream effectors in prostate cancer during progression to androgen independence. Cancer Res. 2004, 64, 2212–2221. [Google Scholar] [CrossRef] [PubMed]
  60. Jespersen, C.G.; Nørgaard, M.; Friis, S.; Skriver, C.; Borre, M. Statin use and risk of prostate cancer: A Danish population-based case-control study, 1997–2010. Cancer Epidemiol. 2014, 38, 42–47. [Google Scholar] [CrossRef] [PubMed]
  61. Lustman, A.; Nakar, S.; Cohen, A.D.; Vinker, S. Statin use and incident prostate cancer risk: Does the statin brand matter? A population-based cohort study. Prostate Cancer Prostatic Dis. 2014, 17, 6–9. [Google Scholar] [CrossRef] [PubMed]
  62. Peng, X.; Li, W.; Yuan, L.; Mehta, R.G.; Kopelovich, L.; McCormick, D.L. Inhibition of proliferation and induction of autophagy by atorvastatin in PC3 prostate cancer cells correlate with downregulation of Bcl2 and upregulation of miR-182 and p21. PLoS ONE 2013, 8, e70442. [Google Scholar] [CrossRef] [PubMed]
  63. Bansal, D.; Undela, K.; D’Cruz, S.; Schifano, F. Statin use and risk of prostate cancer: A meta-analysis of observational studies. PLoS ONE 2012, 7, e46691. [Google Scholar] [CrossRef] [PubMed]
  64. Farwell, W.R.; D’Avolio, L.W.; Scranton, R.E.; Lawler, E.V.; Gaziano, J.M. Statins and prostate cancer diagnosis and grade in a veterans population. J. Natl. Cancer Inst. 2011, 103, 885–892. [Google Scholar] [CrossRef] [PubMed]
  65. Li, J.; Herold, M.J.; Kimmel, B.; Muller, I.; Rincon-Orozco, B.; Kunzmann, V.; Herrmann, T. Reduced expression of the mevalonate pathway enzyme farnesyl pyrophosphate synthase unveils recognition of tumor cells by Vc9Vd2 T cells. J. Immunol. 2009, 182, 8118–8124. [Google Scholar] [CrossRef] [PubMed]
  66. Rossi, S.; Graner, E.; Febbo, P.; Weinstein, L.; Bhattacharya, N.; Onody, T.; Bubley, G.; Balk, S.; Loda, M. Fatty acid synthase expression defines distinct molecular signatures in prostate cancer. Mol. Cancer Res. 2003, 1, 707–715. [Google Scholar] [PubMed]
  67. Hisanori, U.; Kobayashi, T.; Matsumoto, M.; Watanabe, S.; Yoneda, A.; Yoshimi, B. Adipose tissue: Critical contributor to the development of prostate cancer. J. Med. Investig. 2018, 65, 9–17. [Google Scholar]
  68. Wright, C.; Iyer, A.K.V.; Kaushik, V.; Azad, N. Anti-Tumorigenic Potential of a Novel Orlistat-AICAR Combination in Prostate Cancer Cells. J. Cell Biochem. 2017, 118, 3834–3845. [Google Scholar] [CrossRef] [PubMed]
  69. Sadowski, M.C.; Pouwer, R.H.; Gunter, J.H.; Lubik, A.A.; Quinn, R.J.; Nelson, C.C. The fatty acid synthase inhibitor triclosan: Repurposing an anti-microbial agent for targeting prostate cancer. Oncotarget 2014, 5, 9362–9381. [Google Scholar] [CrossRef] [PubMed]
  70. Flavin, R.; Zadra, G.; Loda, M. Metabolic alterations and targeted therapies in prostate cancer. J. Pathol. 2011, 223, 283–294. [Google Scholar] [CrossRef] [PubMed]
  71. Hu, M.B.; Xu, H.; Zhu, W.H.; Bai, P.D.; Hu, J.M.; Yang, T.; Jiang, H.W.; Ding, Q. High-fat diet-induced adipokine and cytokine alterations promote the progression of prostate cancer in vivo and in vitro. Oncol. Lett. 2018, 15, 1607–1615. [Google Scholar] [CrossRef] [PubMed]
  72. Figiel, S.; Pinault, M.; Domingo, I.; Guimaraes, C.; Guibon, R.; Besson, P.; Tavernier, E.; Blanchet, P.; Multigner, L.; Bruyère, F.; et al. Fatty acid profile in peri-prostatic adipose tissue and prostate cancer aggressiveness in African-Caribbean and Caucasian patients. Eur. J. Cancer 2018, 91, 107–115. [Google Scholar] [CrossRef] [PubMed]
  73. Xu, L.; Shen, M.; Chen, X.; Zhu, R.; Yang, D.R.; Tsai, Y.; Keng, P.C.; Chen, Y.; Lee, S.O. Adipocytes affect castration-resistant prostate cancer cells to develop the resistance to cytotoxic action of NK cells with alterations of PD-L1/NKG2D ligand levels in tumor cells. Prostate 2018, 78, 353–364. [Google Scholar] [CrossRef] [PubMed]
  74. Kershaw, E.E.; Flier, J.S. Adipose tissue as an endocrine organ. J. Clin. Endocrinol. Metab. 2004, 89, 2548–2556. [Google Scholar] [CrossRef] [PubMed]
  75. Park, J.; Cho, S.Y.; Lee, S.B.; Son, H.; Jeong, H. Obesity is associated with higher risk of prostate cancer detection in a Korean biopsy population. BJU Int. 2014, 114, 891–895. [Google Scholar] [CrossRef] [PubMed]
  76. Morote, J.; Celma, A.; Planas, J.; Placer, J.; Konstantinidis, C.; Iztueta, I.; de Torres, I.M.; Oliván, M.; Reventós, J.; Doll, A. Sedentarism and overweight as risk factors for the detection of prostate cancer and its aggressivenes. Actas Urológicas Españolas 2014, 38, 232–237. [Google Scholar] [CrossRef] [PubMed]
  77. Tewari, R.; Rajender, S.; Natu, S.M.; Goel, A.; Dalela, D.; Goel, M.M.; Tondon, P. Significance of obesity markers and adipocytokines in high grade and high stage prostate cancer in North Indian men—A cross-sectional study. Cytokine 2013, 63, 130–134. [Google Scholar] [CrossRef] [PubMed]
  78. Bai, P.D.; Hu, M.B.; Xu, H.; Zhu, W.H.; Hu, J.M.; Yang, T.; Jiang, H.W.; Ding, Q. Body mass index is associated with higher Gleason score and biochemical recurrence risk following radical prostatectomy in Chinese men: A retrospective cohort study and meta-analysis. World J. Surg. Oncol. 2015, 13, 311. [Google Scholar] [CrossRef] [PubMed]
  79. Khan, S.; Cai, J.; Nielsen, M.E.; Troester, M.A.; Mohler, J.L.; Fontham, E.T.; Hendrix, L.H.; Farnan, L.; Olshan, A.F.; Bensen, J.T. The association of diabetes and obesity with prostate cancer aggressiveness among Black Americans and White Americans in a population-based study. Cancer Causes Control 2016, 27, 1475–1485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Di Sebastiano, K.M.; Pinthus, J.H.; Duivenvoorden, W.C.; Patterson, L.; Dubin, J.A.; Mourtzakis, M. Elevated C-Peptides, Abdominal Obesity, and Abnormal Adipokine Profile are Associated With Higher Gleason Scores in Prostate Cancer. Prostate 2017, 77, 211–221. [Google Scholar] [CrossRef] [PubMed]
  81. Pruthi, R.S.; Swords, K.; Schultz, H.; Carson, C.C., 3rd; Wallen, E.M. The impact of obesity on the diagnosis of prostate cancer using a modern extended biopsy scheme. J. Urol. 2009, 181, 574–577. [Google Scholar] [CrossRef] [PubMed]
  82. Gallina, A.; Karakiewicz, P.I.; Hutterer, G.C.; Chun, F.K.; Briganti, A.; Walz, J.; Antebi, E.; Shariat, S.F.; Suardi, N.; Graefen, M.; et al. Obesity does not predispose to more aggressive prostate cancer either at biopsy or radical prostatectomy in European men. Int. J. Cancer 2007, 121, 791–795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Schiffmann, J.; Karakiewicz, P.I.; Rink, M.; Manka, L.; Salomon, G.; Tilki, D.; Budäus, L.; Pompe, R.; Leyh-Bannurah, S.R.; Haese, A.; et al. Obesity paradox in prostate cancer: Increased body mass index was associated with decreased risk of metastases after surgery in 13,667 patients. World J. Urol. 2018, 36, 1067–1072. [Google Scholar] [CrossRef] [PubMed]
  84. Hoda, M.R.; Popken, G. Mitogenic and anti-apoptotic actions of adipocyte-derived hormone leptin in prostate cancer cells. BJU Int. 2008, 102, 383–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Deo, D.D.; Rao, A.P.; Bose, S.S.; Ouhtit, A.; Baliga, S.B.; Rao, S.A.; Trock, B.J.; Thouta, R.; Raj, M.H.; Rao, P.N. Differential effects of leptin on the invasive potential of androgen dependent and independent prostate carcinoma cells. J. Biomed. Biotechnol. 2008, 2008, 163902. [Google Scholar] [CrossRef] [PubMed]
  86. Bub, J.D.; Miyazaki, T.; Iwamoto, Y. Adiponectin as a growth inhibitor in prostate cancer cells. Biochem. Biophys. Res. Commun. 2006, 340, 1158–1166. [Google Scholar] [CrossRef] [PubMed]
  87. Macciò, A.; Madeddu, C.; Massa, D.; Astara, G.; Farci, D.; Melis, G.B.; Mantovani, G. Interleukin-6 and leptin as markers of energy metabolic changes in advanced ovarian cancer patients. J. Cell Mol. Med. 2009, 13, 3951–3959. [Google Scholar] [CrossRef] [PubMed]
  88. Gilbert, C.A.; Slingerland, J.M. Cytokines, obesity, and cancer: New insights on mechanisms linking obesity to cancer risk and progression. Annu. Rev. Med. 2013, 64, 45–57. [Google Scholar] [CrossRef] [PubMed]
  89. Beebe-Dimmer, J.L.; Dunn, R.L.; Sarma, A.V.; Montie, J.E.; Cooney, K.A. Features of the metabolic syndrome and prostate cancer in African-American men. Cancer 2007, 109, 875–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Han, J.H.; Choi, N.Y.; Bang, S.H.; Kwon, O.J.; Jin, Y.W.; Myung, S.C.; Chang, I.H.; Kim, T.H.; Ahn, S.H. Relationship between serum prostate-specific antigen levels and components of metabolic syndrome in healthy men. Urology 2008, 72, 749–754. [Google Scholar] [CrossRef] [PubMed]
  91. Ohwaki, K.; Endo, F.; Hattori, K. Abdominal obesity, hypertension, antihypertensive medication use and biochemical recurrence of prostate cancer after radical prostatectomy. Eur. J. Cancer 2015, 51, 604–609. [Google Scholar] [CrossRef] [PubMed]
  92. Asmar, R.; Beebe-Dimmer, J.L.; Korgavkar, K.; Keele, G.R.; Cooney, K.A. Hypertension, obesity and prostate cancer biochemical recurrence after radical prostatectomy. Prostate Cancer Prostatic Dis. 2013, 16, 62–66. [Google Scholar] [CrossRef] [PubMed]
  93. Hammarsten, J.; Högstedt, B. Clinical haemodynamic, anthropometric, metabolic and insulin profile of men with high-stage and high-grade clinical prostate cancer. Blood Press. 2004, 13, 47–55. [Google Scholar] [CrossRef] [PubMed]
  94. Post, J.M.; Beebe-Dimmer, J.L.; Morgenstern, H.; Neslund-Dudas, C.; Bock, C.H.; Nock, N.; Rundle, A.; Jankowski, M.; Rybicki, B.A. The metabolic syndrome and biochemical recurrence following radical prostatectomy. Prostate Cancer 2012, 2011, 245642. [Google Scholar] [CrossRef] [PubMed]
  95. Xu, H.; Zhang, L.M.; Liu, J.; Ding, G.X.; Ding, Q.; Jiang, H.W. The association between overall survival of prostate cancer patients and hypertension, hyperglycemia, and overweight in Southern China: A prospective cohort study. J. Cancer Res. Clin. Oncol. 2013, 139, 943–951. [Google Scholar] [CrossRef] [PubMed]
  96. Palm, D.; Lang, K.; Niggemann, B.; Drell, T.L., 4th; Masur, K.; Zaenker, K.S.; Entschladen, F. The norepinephrine-driven metastasis development of PC-3 human prostate cancer cells in BALB/c nude mice is inhibited by beta-blockers. Int. J. Cancer 2006, 118, 2744–2749. [Google Scholar] [CrossRef] [PubMed]
  97. Fernández, E.V.; Price, D.K.; Figg, W.D. Prostate cancer progression attributed to autonomic nerve development: Potential for therapeutic prevention of localized and metastatic disease. Cancer Biol. Ther. 2013, 14, 1005–1006. [Google Scholar] [CrossRef] [PubMed]
  98. Benish, M.; Bartal, I.; Goldfarb, Y.; Levi, B.; Avraham, R.; Raz, A.; Ben-Eliyahu, S. Perioperative use of beta-blockers and COX-2 inhibitors may improve immune competence and reduce the risk of tumor metastasis. Ann. Surg. Oncol. 2008, 15, 2042–2052. [Google Scholar] [CrossRef] [PubMed]
  99. Grytli, H.H.; Fagerland, M.W.; Fosså, S.D.; Taskén, K.A. Association Between Use of β-Blockers and Prostate Cancer-Specific Survival: A Cohort Study of 3561 Prostate Cancer Patients with High-Risk or Metastatic Disease. Eur. Urol. 2014, 65, 635–641. [Google Scholar] [CrossRef] [PubMed]
  100. Lu, H.; Liu, X.; Guo, F.; Tan, S.; Wang, G.; Liu, H.; Wang, J.; He, X.; Mo, Y.; Shi, B. Impact of beta-blockers on prostate cancer mortality: A meta-analysis of 16,825 patients. OncoTargets Ther. 2015, 8, 985–990. [Google Scholar] [CrossRef] [PubMed]
  101. Di Francesco, S.; Tenaglia, R.L. Obesity, diabetes and aggressive prostate cancer hormone-naïve at initial diagnosis. Cent. Eur. J. Urol. 2013, 66, 423–427. [Google Scholar] [CrossRef] [PubMed]
  102. Di Francesco, S.; Tenaglia, R.L. Obesity, Diabetes Mellitus and Vascular Disease: A Complex Relationship with Prostate Cancer. J. Cancer Ther. 2014, 5, 442–447. [Google Scholar] [CrossRef]
  103. Di Francesco, S.; Tenaglia, R.L. The Association of Obesity and Sistemic Arterial Hypertension With High-Grade Prostate Cancer: Our Experience. J. Cancer Res. Updates 2014, 3, 191–195. [Google Scholar] [CrossRef]
  104. Piątkiewicz, P.; Czech, A. Glucose metabolism disorders and the risk of cancer. Arch. Immunol. Ther. Exp. 2011, 59, 215–230. [Google Scholar] [CrossRef] [PubMed]
  105. Ahima, R.S. Metabolic actions of adipocyte hormones: Focus on adiponectin. Obesity 2006, 14, 9S–15S. [Google Scholar] [CrossRef] [PubMed]
  106. Bråkenhielm, E.; Veitonmäki, N.; Cao, R.; Kihara, S.; Matsuzawa, Y.; Zhivotovsky, B.; Funahashi, T.; Cao, Y. Adiponectin-induced antiangiogenesis and antitumor activity involve caspase-mediated endothelial cell apoptosis. Proc. Natl. Acad. Sci. USA 2004, 101, 2476–2481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Tan, W.; Wang, L.; Ma, Q.; Qi, M.; Lu, N.; Zhang, L.; Han, B. Adiponectin as a potential tumor suppressor inhibiting epithelial-to-mesenchymal transition but frequently silenced in prostate cancer by promoter methylation. Prostate 2015, 75, 1197–1205. [Google Scholar] [CrossRef] [PubMed]
  108. Colli, S.; Cavalcante, F.S.; Martins, M.P.; Sampaio, F.J.; da Fonte Ramos, C. Leptin role in the rat prostate ventral lobe. Fertil. Steril. 2011, 95, 1490–1493. [Google Scholar] [CrossRef] [PubMed]
  109. Saglam, K.; Aydur, E.; Yilmaz, M.; Göktaş, S. Leptin influences cellular differentiation and progression in prostate cancer. J. Urol. 2003, 169, 1308–1311. [Google Scholar] [CrossRef] [PubMed]
  110. Garofalo, C.; Surmacz, E. Leptin and cancer. J. Cell Physiol. 2006, 207, 12–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Noda, T.; Kikugawa, T.; Tanji, N.; Miura, N.; Asai, S.; Higashiyama, S.; Yokoyama, M. Long-term exposure to leptin enhances the growth of prostate cancer cells. Int. J. Oncol. 2015, 46, 1535–1542. [Google Scholar] [CrossRef] [PubMed]
  112. Chang, S.; Hursting, S.D.; Contois, J.H.; Strom, S.S.; Yamamura, Y.; Babaian, R.J.; Troncoso, P.; Scardino, P.S.; Wheeler, T.M.; Amos, C.I.; et al. Leptin and prostate cancer. Prostate 2001, 46, 62–67. [Google Scholar] [CrossRef]
  113. Hu, M.B.; Xu, H.; Hu, J.M.; Zhu, W.H.; Yang, T.; Jiang, H.W.; Ding, Q. Genetic polymorphisms in leptin, adiponectin and their receptors affect risk and aggressiveness of prostate cancer: Evidence from a meta-analysis and pooled-review. Oncotarget 2016, 7, 81049–81061. [Google Scholar] [CrossRef] [PubMed]
  114. Wu, C.; Moreira, D.M.; Gerber, L.; Rittmaster, R.S.; Andriole, G.L.; Freedland, S.J. Diabetes and prostate cancer risk in the REDUCE trial. Prostate Cancer Prostatic Dis. 2011, 14, 326–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Margel, D.; Urbach, D.; Lipscombe, L.L.; Bell, C.M.; Kulkarni, G.; Austin, P.C.; Fleshner, N. Association Between Metformin Use and Risk of Prostate Cancer and Its Grade. J. Natl. Cancer Inst. 2013, 105, 1123–1131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Franciosi, M.; Lucisano, G.; Lapice, E.; Strippoli, G.F.; Pellegrini, F.; Nicolucci, A. Metformin therapy and risk of cancer in patients with type 2 diabetes: Systematic review. PLoS ONE 2013, 8, 71583. [Google Scholar]
  117. Zhang, P.; Li, H.; Tan, X.; Chen, L.; Wang, S. Association of metformin use with cancer incidence and mortality: A meta-analysis. Cancer Epidemiol. 2013, 37, 207–218. [Google Scholar] [CrossRef] [PubMed]
  118. Spratt, D.E.; Zhang, C.; Zumsteg, Z.S.; Pei, X.; Zhang, Z.; Zelefsky, M.J. Metformin and prostate cancer: Reduced development of castration resistant disease and prostate cancer mortality. Eur. Urol. 2013, 63, 709–716. [Google Scholar] [CrossRef] [PubMed]
  119. Wright, J.L.; Stanford, J.L. Metformin use and prostate cancer in Caucasian men: Results from a population-based case–control study. Cancer Causes Control 2009, 20, 1617–1622. [Google Scholar] [CrossRef] [PubMed]
  120. Murtola, T.J.; Tammela, T.L.; Lahtela, J.; Auvinen, A. Anti-diabetic medication and prostate cancer risk: A population-based case-control study. Am. J. Epidemiol. 2008, 168, 925–931. [Google Scholar] [CrossRef] [PubMed]
  121. Azoulay, L.; Dell’Aniello, S.; Gagnon, B.; Pollak, M.; Suissa, S. Metformin and the incidence of prostate cancer in patients with type 2 diabetes. Cancer Epidemiol. Biomark. Prev. 2011, 20, 337–344. [Google Scholar] [CrossRef] [PubMed]
  122. Mayer, M.J.; Klotz, L.H.; Venkateswaran, V. The Effect of Metformin Use during Docetaxel Chemotherapy on Prostate Cancer Specific and Overall Survival of Diabetic Patients with Castration Resistant Prostate Cancer. J. Urol. 2017, 197, 1068–1075. [Google Scholar] [CrossRef] [PubMed]
  123. Liu, Q.; Tong, D.; Liu, G.; Xu, J.; Do, K.; Geary, K.; Zhang, D.; Zhang, J.; Zhang, Y.; Li, Y.; et al. Metformin reverses prostate cancer resistance to enzalutamide by targeting TGF-β1/STAT3 axis-regulated EMT. Cell Death Dis. 2017, 8, e3007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Konijeti, R.; Koyama, S.; Gray, A.; Barnard, R.J.; Said, J.W.; Castor, B.; Elashoff, D.; Wan, J.; Beltran, P.J.; Calzone, F.J.; et al. Effect of a low-fat diet combined with IGF-1 receptor blockade on 22Rv1 prostate cancer xenografts. Mol. Cancer Ther. 2012, 11, 1539–1546. [Google Scholar] [CrossRef] [PubMed]
  125. Giovannucci, E.; Michaud, D. The role of obesity and related metabolic disturbances in cancers of the colon, prostate, and pancreas. Gastroenterology 2007, 132, 2208–2225. [Google Scholar] [CrossRef] [PubMed]
  126. Duman, B.S.; Turkoglu, C.; Gunay, D.; Cagatay, P.; Demiroglu, C.; Buyukdevrim, A.S. The interrelationship between insulin secretion and action in type 2 diabetes mellitus with different degrees of obesity: Evidence supporting central obesity. Diabetes Nutr. Metab. 2003, 16, 243–250. [Google Scholar] [PubMed]
  127. Prabhat, P.; Tewari, R.; Natu, S.M.; Dalela, D.; Goel, A.; Tandon, P.; Goel, M.M.; Singh, K. Is central obesity, hyperinsulinemia and dyslipidemia associated with high-grade prostate cancer? A descriptive cross-sectional study. Indian J. Urol. 2010, 26, 502–506. [Google Scholar] [PubMed]
  128. Xylinas, E.; Ploussard, G.; Durand, X.; Fabre, A.; Salomon, L.; Allory, Y.; Vordos, D.; Hoznek, A.; Abbou, C.C.; de la Taille, A. Low pretreatment total testosterone (3 ng/mL) predicts extraprostatic disease in prostatectomy specimens from patients with preoperative localized prostate cancer. BJU Int. 2011, 107, 1400–1403. [Google Scholar] [CrossRef] [PubMed]
  129. Yamamoto, S.; Yonese, J.; Kawakami, S.; Ohkubo, Y.; Tatokoro, M.; Komai, Y.; Takeshita, H.; Ishikawa, Y.; Fukui, I. Preoperative serum testosterone level as an independent predictor of treatment failure following radical prostatectomy. Eur. Urol. 2007, 52, 696–701. [Google Scholar] [CrossRef] [PubMed]
  130. Albuquerque, G.A.; Guglielmetti, G.B.; Barbosa, J.A.; Pontes, J., Jr.; Fazoli, A.J.; Cordeiro, M.D.; Coelho, R.F.; Carvalho, P.A.; Gallucci, F.P.; Padovani, G.P.; et al. Low serum testosterone is a predictor of high-grade disease in patients with prostate cancer. Rev. Assoc. Med. Bras. 2017, 63, 704–710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Llukani, E.; Katz, B.F.; Agalliu, I.; Lightfoot, A.; Yu, S.S.; Kathrins, M.; Lee, Z.; Su, Y.K.; Monahan Agnew, K.; McGill, A.; et al. Low levels of serum testosterone in middle-aged men impact pathological features of prostate cancer. Prostate Int. 2017, 5, 17–23. [Google Scholar] [CrossRef] [PubMed]
  132. Eltzschig, H.K.; Carmeliet, P. Hypoxia and inflammation. N. Engl. J. Med. 2011, 364, 656–665. [Google Scholar] [CrossRef] [PubMed]
  133. Jin, R.J.; Lho, Y.; Connelly, L.; Wang, Y.; Yu, X.; Saint Jean, L.; Case, T.C.; Ellwood-Yen, K.; Sawyers, C.L.; Bhowmick, N.A.; et al. The nuclear factor-kappaB pathway controls the progression of prostate cancer to androgen-independent growth. Cancer Res. 2008, 68, 6762–6769. [Google Scholar] [CrossRef] [PubMed]
  134. Di Francesco, S.; Castellan, P.; Manco, R.; Tenaglia, R.L. Reciprocal cross-talk between Prostaglandin E2 and bone in prostate cancer: A current Review. Cent. Eur. J. Urol. 2011, 4, 9–12. [Google Scholar] [CrossRef] [PubMed]
  135. Forte, V.; Pandey, A.; Abdelmessih, R.; Forte, G.; Whaley-Connell, A.; Sowers, J.R.; McFarlane, S.I. Obesity, Diabetes, the Cardiorenal Syndrome, and Risk for Cancer. Cardiorenal Med. 2012, 2, 143–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. 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] [CrossRef] [PubMed]
  137. Di Francesco, S.; Tenaglia, R.L. Metabolic alterations, vascular disease and advanced prostate cancer. New Players for Metastatic Advanced Prostate Cancer? J. Anal. Oncol. 2014, 3, 33–35. [Google Scholar] [CrossRef]
  138. Goyal, J.; Pond, G.R.; Galsky, M.D.; Hendricks, R.; Small, A.; Tsao, C.K.; Sonpavde, G. Association of the Charlson comorbidity index and hypertension with survival in men with metastatic castration-resistant prostate cancer. Urol. Oncol. 2014, 32, e27–e34. [Google Scholar] [CrossRef] [PubMed]
  139. Häggström, C.; Stocks, T.; Ulmert, D.; Bjørge, T.; Ulmer, H.; Hallmans, G.; Manjer, J.; Engeland, A.; Nagel, G.; Almqvist, M.; et al. Prospective study on metabolic factors and risk of prostate cancer. Cancer 2012, 118, 6199–6206. [Google Scholar] [CrossRef] [PubMed]
  140. Martini, L.A.; Wood, R.J. Milk intake and the risk of type 2 diabetes mellitus, hypertension and prostate cancer. Arq. Bras. Endocrinol. Metabol. 2009, 53, 688–694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Assayag, J.; Pollak, M.N.; Azoulay, L. Post-diagnostic use of beta-blockers and the risk of death in patients with prostate cancer. Eur. J. Cancer 2014, 50, 2838–2845. [Google Scholar] [CrossRef] [PubMed]
  142. Santala, E.E.; Rannikko, A.; Murtola, T.J. Antihypertensive drugs and prostate cancer survival after radical prostatectomy in Finland-A nationwide cohort study. Int. J. Cancer 2019, 144, 440–447. [Google Scholar] [CrossRef] [PubMed]
  143. Chang, P.Y.; Huang, W.Y.; Lin, C.L.; Huang, T.C.; Wu, Y.Y.; Chen, J.H.; Kao, C.H. Propranolol Reduces Cancer Risk: A Population-Based Cohort Study. Medicine 2015, 94, e1097. [Google Scholar] [CrossRef] [PubMed]
  144. Lim, K.; Burke, S.L.; Head, G.A. Obesity-related hypertension and the role of insulin and leptin in high-fat-fed rabbits. Hypertension 2013, 61, 628–634. [Google Scholar] [CrossRef] [PubMed]
  145. Touyz, R.M. Reactive oxygen species, vascular oxidative stress, and redox signaling in hypertension: What is the clinical significance? Hypertension 2004, 44, 248–252. [Google Scholar] [CrossRef] [PubMed]
  146. Khan, S.R. Is oxidative stress, a link between nephrolithiasis and obesity, hypertension, diabetes, chronic kidney disease, metabolic syndrome? Urol. Res. 2012, 40, 95–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Virdis, A.; Neves, M.F.; Duranti, E.; Bernini, G.; Taddei, S. Microvascular endothelial dysfunction in obesity and hypertension. Curr. Pharm. Des. 2013, 19, 2382–2389. [Google Scholar] [CrossRef] [PubMed]
  148. Sternberg, C.N.; Baskin-Bey, E.S.; Watson, M.; Worsfold, A.; Rider, A.; Tombal, B. Treatment patterns and characteristics of European patients with castration-resistant prostate cancer. BMC Urol. 2013, 13, 58. [Google Scholar] [CrossRef] [PubMed]
  149. Esposito, K.; Chiodini, P.; Capuano, A.; Bellastella, G.; Maiorino, M.I.; Parretta, E.; Lenzi, A.; Giugliano, D. Effect of metabolic syndrome and its components on prostate cancer risk: Meta-analysis. J. Endocrinol. Investig. 2013, 36, 132–139. [Google Scholar] [CrossRef] [PubMed]
  150. Kurata, S.; Tateishi, U.; Shizukuishi, K.; Yoneyama, T.; Hino, A.; Kaida, H.; Fujimoto, K.; Ishibashi, M.; Inoue, T. Assessment of atherosclerosis in oncologic patients using F-fluoride PET/CT. Ann. Nucl. Med. 2013, 27, 481–486. [Google Scholar] [CrossRef] [PubMed]
  151. Lee, B.H.; Taylor, M.G.; Robinet, P.; Smith, J.D.; Schweitzer, J.; Sehayek, E.; Falzarano, S.M.; Magi-Galluzzi, C.; Klein, E.A.; Ting, A.H. Dysregulation of cholesterol homeostasis in human prostate cancer through loss of ABCA1. Cancer Res. 2013, 733, 1211–1218. [Google Scholar] [CrossRef] [PubMed]
  152. Thysell, E.; Surowiec, I.; Hörnberg, E.; Crnalic, S.; Widmark, A.; Johansson, A.I.; Stattin, P.; Bergh, A.; Moritz, T.; Antti, H.; et al. Metabolomic characterization of human prostate cancer bone metastases reveals increased levels of cholesterol. PLoS ONE 2010, 5, e14175. [Google Scholar] [CrossRef] [PubMed]
  153. Jeon, K.P.; Jeong, T.Y.; Lee, S.Y.; Hwang, S.W.; Shin, J.H.; Kim, D.S. Prostate cancer in patients with metabolic syndrome is associated with low grade Gleason score when diagnosed on biopsy. Korean J. Urol. 2012, 53, 593–597. [Google Scholar] [CrossRef] [PubMed]
  154. Morote, J.; Ropero, J.; Planas, J.; Bastarós, J.M.; Delgado, G.; Placer, J.; Celma, A.; de Torres, I.M.; Carles, J.; Reventós, J.; et al. Metabolic syndrome increases the risk of aggressive prostate cancer detection. BJU Int. 2013, 111, 1031–1036. [Google Scholar] [CrossRef] [PubMed]
  155. De Nunzio, C.; Freedland, S.J.; Miano, R.; Trucchi, A.; Cantiani, A.; Carluccini, A.; Tubaro, A. Metabolic syndrome is associated with high grade gleason score when prostate cancer is diagnosed on biopsy. Prostate 2011, 71, 1492–1498. [Google Scholar] [CrossRef] [PubMed]
  156. Kheterpal, E.; Sammon, J.D.; Diaz, M.; Bhandari, A.; Trinh, Q.D.; Pokala, N.; Sharma, P.; Menon, M.; Agarwal, P.K. Effect of metabolic syndrome on pathologic features of prostate cancer. Urol. Oncol. 2013, 31, 1054–1059. [Google Scholar] [CrossRef] [PubMed]
  157. Flanagan, J.; Gray, P.K.; Hahn, N.; Hayes, J.; Myers, L.J.; Carney-Doebbeling, C.; Sweeney, C.J. Presence of the metabolic syndrome is associated with shorter time to castration-resistant prostate cancer. Ann. Oncol. 2011, 22, 801–807. [Google Scholar] [CrossRef] [PubMed]
  158. Di Francesco, S.; Tenaglia, R.L. Metabolic Syndrome and Aggressive Prostate Cancer at Initial Diagnosis. Horm. Metab. Res. 2017, 49, 507–509. [Google Scholar] [CrossRef] [PubMed]
  159. Satariano, W.A.; Ragland, K.E.; Van Den Eeden, S.K. Cause of death in men diagnosed with prostate carcinoma. Cancer 1998, 83, 1180–1188. [Google Scholar] [CrossRef] [Green Version]
  160. Fouad, M.N.; Mayo, C.P.; Funkhouser, E.M.; Hall, H.I.; Urban, D.A.; Kiefe, C.I. Comorbidity independently predicted death in older prostate cancer patients, more of whom died with than from their disease. J. Clin. Epidemiol. 2004, 57, 721–729. [Google Scholar] [CrossRef] [PubMed]
  161. Groome, P.A.; Rohland, S.L.; Siemens, D.R.; Brundage, M.D.; Heaton, J.; Mackillop, W.J. Assessing the impact of comorbid illnesses on death within 10 years in prostate cancer treatment candidates. Cancer 2011, 117, 3943–3952. [Google Scholar] [CrossRef] [PubMed]
  162. Di Francesco, S.; Tenaglia, R.l. Vascular Disease and Prostate Cancer: A Conflicting Association. J. Cancer Res. Updates 2014, 3, 81–84. [Google Scholar] [CrossRef]
  163. Zöller, B.; Ji, J.; Sundquist, J.; Sundquist, K. Risk of coronary heart disease in patients with cancer: A nationwide follow-up study from Sweden. Eur. J. Cancer 2012, 48, 121–128. [Google Scholar] [CrossRef] [PubMed]
  164. Omalu, B.I.; Hammers, J.L.; Parwani, A.V.; Balani, J.; Shakir, A.; Ness, R.B. Is there an association between coronary atherosclerosis and carcinoma of the prostate in men aged 50 years and older? An autopsy and coroner based post-mortem study. J. Clin. Pract. 2013, 16, 45–48. [Google Scholar] [CrossRef] [PubMed]
  165. Dreyer, L.; Olsen, J.H. Risk for non-smoking-related cancer in atherosclerotic patients. Cancer Epidemiol. Biomark. Prev. 1999, 8, 915–918. [Google Scholar]
  166. Stamatiou, K.N.; Alevizos, A.G.; Mihas, K.; Mariolis, A.D.; Michalodimitrakis, E.; Sofras, F. Associations between coronary heart disease, obesity and histological prostate cancer. Int. Urol. Nephrol. 2007, 39, 197–201. [Google Scholar] [CrossRef] [PubMed]
  167. Heron, M.; Hoyert, D.L.; Murphy, S.L.; Xu, J.; Kochanek, K.D.; Tejada-Vera, B. Deaths: Final data for 2006. Natl. Vital Stat. Rep. 2009, 57, 1–134. [Google Scholar] [PubMed]
  168. Cheung, A.S.; Pattison, D.; Bretherton, I.; Hoermann, R.; Lim Joon, D.; Ho, E.; Jenkins, T.; Hamilton, E.J.; Bate, K.; Chan, I.; et al. Cardiovascular risk and bone loss in men undergoing androgen deprivation therapy for non-metastatic prostate cancer: Implementation of standardized management guidelines. Andrology 2013, 1, 583–589. [Google Scholar] [CrossRef] [PubMed]
  169. Thomas, J.A.; Gerber, L.; Bañez, L.L.; Moreira, D.M.; Rittmaster, R.S.; Andriole, G.L.; Freedland, S.J. Prostate cancer risk in men with baseline history of coronary artery disease: Results from the REDUCE Study. Cancer Epidemiol. Biomark. Prev. 2012, 21, 576–581. [Google Scholar] [CrossRef] [PubMed]
  170. Pereira, L.M.N.; Biolo, A.; Foppa, M.; da Rosa, P.R.; Rohde, L.E.; Clausell, N. A prospective, comparative study on the early effects of local and remote radiation therapy on carotid intima-media thickness and vascular cellular adhesion molecule-1 in patients with head and neck and prostate tumors. Radiother. Oncol. 2011, 101, 449–453. [Google Scholar] [CrossRef] [PubMed]
  171. Touvier, M.; Fezeu, L.; Ahluwalia, N.; Julia, C.; Charnaux, N.; Sutton, A.; Méjean, C.; Latino-Martel, P.; Hercberg, S.; Galan, P.; et al. Association between prediagnostic biomarkers of inflammation and endothelial function and cancer risk: A nested case-control study. Am. J. Epidemiol. 2013, 177, 3–13. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Intratumoral de novo steroid synthesis from circulating serum cholesterol. Increased cholesterol and progesterone levels have been identified in prostate cancer along with an increased expression of enzymes involved in steroidogenesis, in particular, CYP17A, 17-keto-reductase and 5α-reductase 1.
Figure 1. Intratumoral de novo steroid synthesis from circulating serum cholesterol. Increased cholesterol and progesterone levels have been identified in prostate cancer along with an increased expression of enzymes involved in steroidogenesis, in particular, CYP17A, 17-keto-reductase and 5α-reductase 1.
Medicina 55 00062 g001
Figure 2. Reciprocal relationship between prostate cancer and cardiovascular disease. Common risk factors link prostate cancer and cardiovascular disease. It is possible to hypothesize that this relation has a direct association with common metabolic components as obesity, dyslipidaemia, systemic arterial hypertension, glucose intolerance, recent type II diabetes mellitus, metabolic syndrome, atherosclerosis. These factors are associated with Insulin Resistance, hyperinsulinemia, oxidative stress, inflammation and precocious vascular damage and are related to advanced prostate cancer, bone metastases, biochemical recurrence and increased tumor mortality.
Figure 2. Reciprocal relationship between prostate cancer and cardiovascular disease. Common risk factors link prostate cancer and cardiovascular disease. It is possible to hypothesize that this relation has a direct association with common metabolic components as obesity, dyslipidaemia, systemic arterial hypertension, glucose intolerance, recent type II diabetes mellitus, metabolic syndrome, atherosclerosis. These factors are associated with Insulin Resistance, hyperinsulinemia, oxidative stress, inflammation and precocious vascular damage and are related to advanced prostate cancer, bone metastases, biochemical recurrence and increased tumor mortality.
Medicina 55 00062 g002
Figure 3. “New Players” in advanced metastatic PCa. Hypertension, hypercholesterolemia and atherosclerosis could be considered common etiopathogenetic mechanisms in advanced metastatic prostate cancer, particularly in presence of bone metastases.
Figure 3. “New Players” in advanced metastatic PCa. Hypertension, hypercholesterolemia and atherosclerosis could be considered common etiopathogenetic mechanisms in advanced metastatic prostate cancer, particularly in presence of bone metastases.
Medicina 55 00062 g003

Share and Cite

MDPI and ACS Style

Di Francesco, S.; Robuffo, I.; Caruso, M.; Giambuzzi, G.; Ferri, D.; Militello, A.; Toniato, E. Metabolic Alterations, Aggressive Hormone-Naïve Prostate Cancer and Cardiovascular Disease: A Complex Relationship. Medicina 2019, 55, 62. https://doi.org/10.3390/medicina55030062

AMA Style

Di Francesco S, Robuffo I, Caruso M, Giambuzzi G, Ferri D, Militello A, Toniato E. Metabolic Alterations, Aggressive Hormone-Naïve Prostate Cancer and Cardiovascular Disease: A Complex Relationship. Medicina. 2019; 55(3):62. https://doi.org/10.3390/medicina55030062

Chicago/Turabian Style

Di Francesco, Simona, Iole Robuffo, Marika Caruso, Giulia Giambuzzi, Deborah Ferri, Andrea Militello, and Elena Toniato. 2019. "Metabolic Alterations, Aggressive Hormone-Naïve Prostate Cancer and Cardiovascular Disease: A Complex Relationship" Medicina 55, no. 3: 62. https://doi.org/10.3390/medicina55030062

Article Metrics

Back to TopTop