Human microbiome and prostate cancer development: current insights into the prevention and treatment

Solmaz Ohadian Moghadam , Seyed Ali Momeni

Front. Med. ›› 2021, Vol. 15 ›› Issue (1) : 11 -32.

PDF (790KB)
Front. Med. ›› 2021, Vol. 15 ›› Issue (1) : 11 -32. DOI: 10.1007/s11684-019-0731-7
REVIEW
REVIEW

Human microbiome and prostate cancer development: current insights into the prevention and treatment

Author information +
History +
PDF (790KB)

Abstract

The huge communities of microorganisms that symbiotically colonize humans are recognized as significant players in health and disease. The human microbiome may influence prostate cancer development. To date, several studies have focused on the effect of prostate infections as well as the composition of the human microbiome in relation to prostate cancer risk. Current studies suggest that the microbiota of men with prostate cancer significantly differs from that of healthy men, demonstrating that certain bacteria could be associated with cancer development as well as altered responses to treatment. In healthy individuals, the microbiome plays a crucial role in the maintenance of homeostasis of body metabolism. Dysbiosis may contribute to the emergence of health problems, including malignancy through affecting systemic immune responses and creating systemic inflammation, and changing serum hormone levels. In this review, we discuss recent data about how the microbes colonizing different parts of the human body including urinary tract, gastrointestinal tract, oral cavity, and skin might affect the risk of developing prostate cancer. Furthermore, we discuss strategies to target the microbiome for risk assessment, prevention, and treatment of prostate cancer.

Keywords

microbiome / prostate cancer / prevention / treatment / molecular pathological epidemiology (MPE) / biomarker

Cite this article

Download citation ▾
Solmaz Ohadian Moghadam, Seyed Ali Momeni. Human microbiome and prostate cancer development: current insights into the prevention and treatment. Front. Med., 2021, 15(1): 11-32 DOI:10.1007/s11684-019-0731-7

登录浏览全文

4963

注册一个新账户 忘记密码

Introduction

Prostate cancer has been reported as a worldwide important kind of cancer. Based on the reports of Cancer Research UK, this neoplasm is the most common form of non-skin cancer among men. It has been reported that 1 276 106 new cases were added to the prostate cancer patients all over the world [1]. The prevalence of prostate cancer is increasing worldwide, even in Asian countries including Iran [25].

Cancer is a complex multifactorial disease that involves multiple genetic, immunological, environmental and physiologic factors, leading to the complexity of the treatment. There are several risk factors considered for prostate cancer such as age, ethnicity, family history and environmental factors (diet and lifestyle), as well as microbial (viral and bacterial) infections and inflammation. Current reports suggest that prostate infections and bacterial communities within the host are associated with chronic inflammation and immunological responses, leading to prostate carcinogenesis [6,7].

There are 10 trillion to 100 trillion microbial cells in the human body including bacteria in the gut and the genes they harbor, which are called microbiota and microbiome, respectively [8]. The human-associated microbiota and its effect on the development of cancer is an interesting topic. The microbiome imposes an effect on the entire process of carcinogenesis from initiation to progression and even therapeutic consequences. This effect may be direct, such as the role of Helicobacter pylori in gastric cancer or indirect, through changes in metabolism and/or immune system settings. For example, a pathogenic shift in the intestinal microbial content may increase the risk of diseases, such as obesity, diabetes, and cancer via affecting the hormones [912].

In this review, we discuss recent data about how the microbes colonizing different parts of the human body including urinary tract, gastrointestinal tract, oral cavity, and skin might affect the risk of prostate cancer development. Furthermore, we discuss strategies to target the microbiome for risk assessment, prevention, and treatment of prostate cancer.

Microbes and genome instability

Specific microbes cause genome instability and have a serious effect on tumorigenesis through the production of tumor-promoting metabolites such as hydrogen sulfide and superoxide radicals and some microbial toxins including colibactin, Bacteroides fragilis toxin and cytolethal distending toxin (CDT). CDT is produced by numerous species associated with colorectal cancer, gastric cancer, and gallbladder cancer, such as Salmonella typhi, Escherichia coli, and H. pylori [13,14]. Moreover, some bacterial species including Campylobacter jejuni, Shigella dysenteriae, Hemophilus ducreyi, Helicobacter hepaticus, and Salmonella enterica can produce CDT, which has DNase activity and causes DNA double-strand breaks and apoptosis. The superoxide radicals produced by Enterococcus faecalis are also known to be involved in colorectal cancers by creating double-strand DNA breaks [1517].

Microbes and immune response

Infiltration of different immune cell populations, including neutrophils, macrophages, dendritic cells (DCs), adipose cells, T cells, B cells and natural killer (NK) cells is increased in tumor microenvironment. Leukocytes comprise more than 50% of tumor masses [18].

Microbes have been shown to regulate immunity processes. For example, Fusobacterium nucleatum and H. pylori can suppress the activity of T cells. The presence of T cells in cancerous tissues is associated with an increased overall survival among patients and a more effective response to treatment [19]. Microbes may cause tumor growth by changing the expression of growth hormones. For example, E. coli strains produce genotoxin colibactin which causes tumor growth by increasing production of growth factors [20]. Otherwise, some infectious agents such as the influenza A virus, Staphylococcus aureus, and Streptococcus Group A induce growth of tumor cells and metastases by utilizing transforming growth factor b (TGF-b) [2123]. TGF-b is a multifunctional regulatory cytokine controlling several aspects of cellular function, including differentiation, cellular proliferation, migration, apoptosis, angiogenesis, immune surveillance, and survival. It also has tumor suppressor role by inhibition of epithelial cell proliferation. However, loss of autocrine TGF-b activity may cause malignant progression of some epithelial cells, suggesting a prooncogenic role for TGF-b in addition to its tumor suppressor role [24].

The normal human prostate tissue contains various types of immune cells including lymphocytes, which are either stromal or intraepithelial. Immune effector cells such as DCs and macrophages are also present in the prostate. Other immune cells including basophils, neutrophils, and eosinophils are rare in healthy prostate tissue but increased in inflamed regions [2527]. Inflammatory changes in the prostate microenvironment accompanied by prostate infection cause epithelial barrier disruption and consequent progression of prostate cancer. It has also been shown that urinary tract microbiome alteration could lead to prostate infection. Several studies have looked at the role of inflammation and infection in the development of prostate cancer. Generally, the prostate tumor microenvironment is rich in inflammatory cells. It seems that with the progression of prostate tumors, number of anaerobic bacteria is increased due to the oxygen depletion [28,29].

Microbes and inflammation

Relationship between inflammation and cancer was hypothesized by Virchow over 150 years ago upon his discovery of leukocytes in cancerous tissues [30]. Up to 10%−20% of cancers are attributed to chronic inflammation involving microbes [31]. Inflammation can contribute to development of cancer in various organs, including liver, colon, bladder, lung, pancreas, and prostate [30]. Current molecular evidences from animal and human studies implicated the regulatory role of chronic inflammation in prostate cancer development and progression to advanced metastatic disease [3033].

Microbes colonize the human body shortly at birth [34] and are involved in homeostasis, immunity education, and host defense. Each organ favors the survival and growth of specific collection of microbes [32]. Microbiome have an important immunoregulatory role in healthy individuals. Activation of myelopoiesis in the bone marrow by commensal bacteria has been shown more than three decades ago, suggesting the role of microbiota in the induction of host immune system [35]. For instance, in vitro experiments have shown that the deficiency of bone marrow resident myeloid cell populations make them susceptible to Listeria infections. Re-colonization of germ-free mice with microbiota restores the immunity against Listeria [36].

Homeostasis depends on the integrity of the epithelial barrier colonized by the commensal microflora protecting the host. The balanced symbiotic relationship between the host and its microbiome could be distracted due to environmentally induced dysbiosis [37]. An altered microbiota, termed dysbiosis, could lead to loss of epithelial barrier integrity [38].

The extracellular matrix (ECM) is a three-dimensional network of extracellular macromolecules, such as laminin, collagen, fibronectin, and proteoglycans, that provides structural and biochemical support to adjacent cells. It has a critical role in regulation of cell survival in normal and tumorigenic growth through the regulation of cell communication, proliferation, differentiation, and survival [39].

Inflammatory cascade has an important effect on the tumor immune microenvironment. In chronic inflammation, several cytokines are produced by inflammatory cells, such as tumor necrosis factor (TNF), interleukin-7 (IL-7), interleukin-2 (IL-2), RANTES, and macrophage inflammatory protein-1b (MIP-1b). Moreover, chronic inflammatory cascade causes activation of growth factors including fibroblast growth factor (FGF) and TGF-b [40]. The release of soluble inflammatory mediators into the ECM results in activation of surrounding stromal cells and promotes formation of reactive stroma [41,42] (Fig. 1). The reactive stroma represents an inflammatory cytokine-rich microenvironment contributing to prostate tumor progression and is associated with poor outcome in clinically localized prostate cancer [42]. Generally, normal cells detached from the ECM are not able to survive and proliferate, and undergo a form of apoptosis termed anoikis. Anoikis is a variant of programmed cell death that occurs due to a lack of extracellular connections to the ECM and adjacent cells [43]. Following inflammation-mediated disruption of ECM, anoikis-resistant cells invade and migrate to distant sites and prostate cancer metastasis occurs [33].

Growth factors, DNA-damage-promoting agents, and a micro-environment rich in inflammatory cells are known as the other hallmarks of cancer. Host cells release several chemical signals in response to tissue damage, which stimulate activation and migration of leukocytes (neutrophils, monocytes, and eosinophils) to the damaged area [44]. Moreover, it was suggested that inflammation due to tissue injuries increases the proliferative capability of cells in the involved area, leading to carcinogenesis [45]. Neutrophils are the first leukocytes recruited to the sites of infection or tissue injury followed by monocytes [46,47]. Chemokines direct the recruitment of particular leukocyte effector cells, leading to the progression of the inflammatory response [44]. The dysregulation of cytokine/chemokine may result in chronic inflammation and subsequent subversion of cell death and/or repair programs and eventually, contributes to cancer pathogenesis [44]. Several chronic inflammatory diseases contributing to cancer, are due to either altered microbiome or the involvement of specific microbes [48]. Gut microbiota is capable of establishing a proinflammatory or antitumor milieu through the modulation of host physiology and functioning of immune cells [49]. Recent evidence suggests a role for inflammation and tissue microbes in prostate cancer [28]. Generally, intraprostatic inflammation is detected in prostate biopsies [50]. In addition, chronic inflammation in benign prostate tissue is associated with high-grade prostate cancer [51]. The high prevalence of chronic inflammatory infiltrates in histopathological examination of the prostate from radical prostatectomy (RP) specimens, prostate biopsy, and transurethral resection of the prostate (TURP) suggests a potential link between chronic inflammation and prostate cancer [52]. Prostatitis can be caused by a variety factors including microbial pathogens such as non-sexually transmitted organisms (E. coli and Propionibacterium acnes) [53,54], as well as sexual transmitted organisms (Neisseria gonorrhea and Chlamydia trachomatis) [55,56]. Apart from the causative factors for prostatitis (specific pathogen or environmental factor), inflammation can be considered as a ubiquitous factor associated with increased incidence of prostate cancer among patients with prostatitis [57]. Inflammatory cells result in enhanced vascularity, DNA damage, and ECM degradation to form a nurturing growth microenvironment [46]. Chronic inflammation of the prostate can be caused by persistent infection. Thereby, DNA damage occurs due to constant production of oxygen and nitrogen species produced by leukocytes [58].

Inflammation of prostate, is characterized by an increased number of inflammatory cells in the prostate tissue [59]. The tumor microenvironment in prostate tumor frequently contains inflammatory cells. It has been suggested that inflammatory changes in the microenvironment of prostate along with infection of prostate infection are associated with epithelial barrier disruption and stimulation of prostate cancer development [28]. Numerous signaling factors and biological events influenced inflammation in the prostate microenvironment, linking inflammation to prostate cancer progression and metastasis. These associations provide promising potential therapeutic targets. Currently, several researches have suggested the association of antiinflammatory agents and reduced prostate cancer risk [33].

Microbial-induced inflammation contributes to cancer progression by stimulation of cytokine and chemokine production that stimulates cell proliferation and/or apoptosis inhibition [60]. Numerous studies have shown the association of microbiome composition with modulation of tumor-promoting inflammatory cytokines [61]. A diverse microbial flora in healthy individuals results in production of inflammatory cytokines including TNF-α, IL-6, IL-1β, interferon g (IFNγ), IL-17, and IL-22 by myeloid and lymphoid cells [62]. Thereby, tumor progression is influenced by these cytokines through different mechanisms, including recruitment of suppressive immune cells into the tumor microenvironment via TNF-α, IL-6, IL-1β or tumor immune surveillance via IFNγ and IL-17 [61]. TNF is a cytokine involved in systemic inflammation [63]. It has been shown that TNF is a mediator of cancer-associated chronic inflammation and stimulates tumor growth and progression [63]. Nuclear factor k B (NF-κB) is a protein that regulates transcription of DNA, cytokine production, and cell survival. It induces the expression of various proinflammatory genes involved in cell growth, angiogenesis, and metastasis [64]. Increased expression of NF-κB have been found in prostate tumor cells [64]. It may stimulate cell proliferation in prostate tumor cells by regulating the expression of genes involved in cell cycle controlling (such as c-myc, cyclin D1, and IL-6) [65]. In addition, NF-κB transcription factors regulate the expression of angiogenic factors, including vascular endothelial growth factor (VEGF) and IL-8 [65]. Constitutive NF-kB activation in prostate cancer cells has been associated with invasive behavior of prostate cancer tumors [64].

In the tumor microenvironment, immune cells respond to lipopolysaccharide (LPS), flagellin, and other bacterial components and products by producing several cytokines such as IL-23 and IL-17 [66]. LPS is a characteristic component of Gram-negative bacteria. It is a ligand for TLR4 and stimulates the production of proinflammatory cytokines [66]. In addition, it has proangiogenic effects [67]. It also stimulates myeloid cells to produce reactive oxygen species (ROS) which result in DNA damage and mutation leading to cellular transformation [68]. Moreover, other bacterial components including flagellin and peptidoglycan affect systemic inflammation through the ligation of TLR5 and TLR2, respectively [66,69]. Microbial metabolites such as hydrogen sulfide (H2S], N-nitroso compounds, and polyamines have also direct effects on DNA including microsatellite instability [70]. On the other hand, the microbial products have the potential to suppress cancer progression. For instance, it has been reported that 3-ethylbutyrolactone, kynurenic acid, and 3-methyladenine from Lactobacillus johnsonii decrease the gene damage and inflammation that is beneficial in cancer prevention [71].

In summary, the complex relationship between inflammation, the microbiome, and cancer is yet to be elucidated. Precise defining the roles of microbes and inflammation in cancer can offer unique opportunities for improved management and prevention of cancer in the future.

Microbiome and prostate cancer

Gut microbiome and prostate cancer

The microbiome and its inflammatory and carcinogenic effects are organ specific [72]. Gut microbiome make up about 99% of the total microbial mass of the human body and has local and distal effects. The gut microbiome is better known than microbiome of other parts of the body [72]. The human is constantly exposed to microbiome as an environmental factor. Recent studies have shown the association between microbiome alteration and carcinogenesis in the colon, liver, and pancreas [38]. These carcinogenic alterations are due to mechanisms including stimulating inflammatory responses mediated by microorganism-associated molecular patterns (MAMPs) and genomic instability as described above. Inflammatory responses mediated by microbiota occur both locally and systemically via proinflammatory cytokines, such as IL-17, IL-23, TNF-α, and IFNγ [73].

The introduction of next generation sequencing (NGS) of bacterial 16S rRNA has led to better identification of gut microbiome particularly anaerobes that comprise the majority of the human gut microbiome. The human gut microbiome consist of 1013–1014 organisms and 3×106 genes [74]. The bacterial microbiome has several functions and its content is dependent on the individual’s diet, geography, initial infant colonization as well as the individual’s immune system [75].

There are four phyla of bacteria predominantly comprising human gut microbiome including Firmicutes, Bacteroidetes, Proteobacteria, and Actinomycetes, of which, the first two constitute about 90% of total bacteria. Firmicutes are mostly Gram-positive anaerobic clostridia, streptococci and enterococci, and Bacteroidetes is composed of Gram-negative bacilli. Bacteroides thetaiotamicron and Bacillus fragilis are recognized as examples of this phylum with the ability to digest complex carbohydrates. Actinobacteria represent Gram-positive bacteria with a high G+C content in their DNA with the example of Bifidobacteria. They are the first microbes colonizing the human gastrointestinal tract and are known as probiotic organisms. In addition, Proteobacteria represent a diverse group of Gram-negative bacteria including E. coli and Klebsiella species [75,76].

Estrogens are considered as important agents for treatment or prevention of prostate cancer. First, the association of androgens with prostate cancer was shown by Huggins and Hodges [77]. Then it was shown that estrogen has an indirect antiandrogenic effect by feedback inhibition of hypothalamic luteinizing hormone (LHRH), stimulating the release of luteinizing hormone (LH) from pituitary [78]. Studies have shown that exogenous estrogens such as the non-metabolized diethylstilbestrol (DES) can inhibit telomerase, resulting in inhibition of prostate cancer cell division [79,80].

It has been suggested that gut microbiome via a group of gut bacteria whose products can metabolize estrogens (defined as estrobolome) have major influence on level of circulating estrogens, and consequent risk of developing estrogen-driven cancers, including prostate cancer [8183]. Typically, circulating estrogens are hepatically conjugated via glucuronidation and producing glucuronides (conjugated estrogens), which do not bind to estrogen receptors (ERs). Dysbiosis of gut microbiome can promote deconjugation and recycling of estrogens via secretion of b-glucuronidase enzyme. Deconjugated estrogens (active forms) can bind to ERs, result in cell proliferation and tumor development [8386] (Fig. 1).

Therefore, it is rational that patients with prostate cancer may have altered gut microbiome in comparison to healthy individuals. In this respect, it has been demonstrated that there is a significant variation of microbiota among different patient populations. In a recent study, Bacteroides massiliensis was more prevalent among prostate cancer patients, and Faecalibacterium prausnitzii and Eubacterium rectale were more prevalent in benign controls. The differences in the gut microbiota of prostate cancer patients compared to benign controls suggest an important role in the pathobiology of prostate cancer and need further investigation (Table 1) [87]. B. massiliensis is a member of the Bacteroides species [87]. F. prausnitzii is a Gram-positive, rod-shaped, anaerobic bacterium and belongs to the phylum Firmicutes utilizing acetate to produce butyrate [88]. E. rectale is a member of Firmicutes phylum and produces butyrate [89].

Butyrate is a type of fatty acid and has important benefits, including antiinflammatory and antitumorigenic properties. Butyrate functions as an antitumor agent by inducing apoptosis and inhibiting cell proliferation [90]. An in vivo study has demonstrated that F. prausnitzii resulted in a reduction in proinflammatory cytokine secretion (TNF-α, IL-12) and increase in antiinflammatory cytokine secretion (IL-10) [91]. Moreover, Bacteroides and Faecalibacterium sp. are harbor genes encoding b-glucuronidase, which is essential for estrogen metabolism. However, the Eubacterium sp. do not have these genes [92]. β-glucuronidase function leads to increase the circulating levels of free estrogens and react with the host’s DNA and make mutations leading to prostate cancer [82] (Fig. 1).

Metabolic pathways of gut microbiome and prostate cancer

Life style is considered as one of the important factors associated with prostate cancer, which greatly affects the microbiota of the intestine. Reduction of the intestinal microbial diversity would make an increase in the number of the bacteria causing systemic inflammation and may lead to increased risk of tumorigenesis [93,94] (Fig. 1). Intestinal bacteria are well recognized for their important role in production of active biological agents including folate, riboflavin (vitamin B12), biotin (vitamin B7), and arginine, contributing in development and progression of cancer. Association of folate and the risk of prostate cancer is the most widely studied of these to date, followed by biotin and arginine [95,96].

Fecal microbiome is a reflection of the intestinal microbiome. Some studies focused on the systemic effects of fecal microbiome, in order to examine their metabolic pathways and to achieve an appropriate diet for prostate cancer patients [28,29,97] (Table 1). Evaluation of microbiome metabolic pathways can provide an insight into the modifiable risk factors of prostate cancer. A study examined various bacterial components in fecal microbiome to consider the metabolic pathways associated with prostate cancer [98]. The analysis showed high frequency of bacteria associated with carbohydrate metabolism pathways and lack of bacteria producing B vitamins. Epidemiological studies have shown the protective effect of folate against cancers including intestinal and cervix cancers [98101].

Folate plays a significant role in the synthesis of nucleotides and DNA methylation. In vivo studies have shown that folate deficiency causes substitution of uracil with thymine in DNA, the instability of DNA, and the higher mutation frequency [100]. According to a research report, folate producer microbiota is more abundant in non-cancer than in cancer patients [98]. It seems that folate supplements source increases the risk of prostate cancer, but natural sources of folate have protective effects. Therefore, it has been suggested that men with high-grade prostate cancer use probiotics instead of supplements. Furthermore, although riboflavin has received less attention than folate, it is a cofactor of methylenetetrahydrofolate reductase and there is a synergy between the protective function of folate and riboflavin [98,102]. Mammalian cells are not capable of producing biotin and are dependent to the gut microbiome. The deficiency of this vitamin has been reported in men with prostate cancer. LASSO analysis has shown a lack of biotin-producing microbiome among patients with prostate cancer that can indicate the importance of this vitamin [102]. In vitro studies have shown that biotin supplementation enhances mRNA encoding cytochrome P450 1B1 (CYP1B1) in human lymphocytes, and may have similar effects on non-lymphoid cells [103]. Therefore, it should be noted that, like other B group vitamins, supplementation with a highly concentrated biotin can have adverse effects [103].

Oral microbiome and prostate cancer

There are more than 700 different bacterial species in the human oral cavity recognized as oral microbiome [104]. Recent studies have shown that inadequate oral hygiene has led to a destruction of the oral microbial population, resulting in an increase in the number of oral pathogens [105] (Fig. 1). The association of oral pathogens with various diseases including cardiovascular diseases [106,107], preterm birth [108], as well as pancreatic cancer [105] has been shown previously. The important question is that how oral bacteria get to the prostate. And how can they contribute to inflammation or carcinogenesis? As mentioned, chronic inflammation could be a risk factor for prostate cancer. In addition, prostatitis is an inflammatory status for the prostate. Several studies have shown the spread of pathogenic bacteria from the oral cavity to other parts of the body in a variety of diseases, including prostatitis. Etiopathogenesis is similar in chronic prostatitis and periodontitis. Periodontitis is a chronic inflammation and is caused by oral pathogens and causes the loss of soft tissue attached to the teeth [109,110]. Some prostatitis categories, as well as periodontitis, are due to Gram-negative bacteria, increased proinflammatory cytokines, and decreased antiinflammatory cytokine [111,112]. Several systemic diseases are associated with periodontitis, including blood disorders and atherosclerosis [113,114]. The relationship between periodontitis and systemic diseases might be due to the dissemination of bacteria and their toxins throughout the body or immune deficiency [10]. For example, the presence of oral bacteria was shown in synovial fluid of patients with rheumatoid arthritis. Moreover, several studies have shown the association of periodontitis with increased prostate-specific antigen (PSA) [115,116]. There is also a correlation between PSA levels and coexistence of chronic prostatitis and periodontitis [115]. Higher levels of serum PSA may also be associated with aggressiveness of prostate cancer [117,118].

Oral infectious diseases can cause inflammation throughout the body by increasing C-reactive protein (CRP) as well as proinflammatory cytokines such as TNF, IL-1, and IL-6, which may accordingly cause prostate inflammation [119]. In addition, inflammatory responses may destroy the integrity of the prostate epithelium and result in release of PSA to the blood stream [115]. Detection of oral bacterial DNA in prostatic secretions of men with both prostatitis and periodontitis conditions, indicates an association between these diseases and the potential role of inflammatory processes [120]. Thus, elimination of the oral infection foci is an essential prostate health priority [116]. Porphyromonas gingivalis was prevalently isolated from prostatic secretions. This bacterium has the ability to produce Arg-gingipain to destroy collagen [121]. This bacterium can enter into epithelial cells through binding to erythrocytes [122]. The association of P. gingivalis with pancreatic cancer has already been shown [109,123]. P. gingivalis has also the potential to invade the human immune system via destroying signaling pathways [124,125].

To date, no study have addressed the relationship between oral microbiome and prostate cancer risk that warrants further research. However, oral microbiome might be associated with prostate cancer, or at least inflammatory condition which increases the risk of prostate cancer.

Urinary microbiome and prostate cancer

For a long time it was thought that the urinary tract is sterile [126]. Recent studies indicated a typical urinary microbiome, which is distinctive from the gut microbiome [127129]. One of the concerns about urinary microbiome was the possibility of sampling contamination. To rule out this concern, highly advanced molecular techniques such as 16S rDNA sequencing were used [127].

The recent discovery of the existence of urinary tract microbiome has highlighted the role of the microbiome in prostate cancer [126]. Since the urinary tract is very close to the prostate and can contaminate it, urinary microbial studies are important in identifying prostate diseases [130,131]. Several studies have reported the isolation of various microbial strains, including Corynebacterium, Streptococcus, Veillonella, Prevotella, Anaerococcus, Propionibacterium, Finegoldia, and Staphylococcus, from the urine of adult males [132134]. P. acnes is one of the most commonly isolated bacteria from male urine, and is a proinflammatory bacterium. Association of P. acnes with prostatitis in animal models as well as human prostate cancer has been studied previously [29,53,135139]. Chronic prostatitis is most commonly caused by uropathogenic strains of E. coli and enterococci [140]. Prostatitis due to E. coli and P. acne strains may cause morphologic changes and hyperplasia. These changes have also been associated with decreasing the expression of tumor suppressor NKX 3.1 in the prostate [53]. According to studies, proinflammatory bacteria such as Streptococcus anginosus, Anaerococcus lactolyticus, Varibaculum cambriense, and Propionimicrobium lymphophilum have been found to be more common in patients with cancer [141,142], which suggests that inflammatory bacteria are likely to cause prostate inflammation for the development or progression of prostate cancer [28].

Using advanced molecular techniques such as polymerase chain reaction (PCR) and 16S rRNA sequencing, it has been suggested that some unchangeable host factors such as the expression of specific receptors, and the mother-to-child bacterial transmission during the first few months of life are contributed to the colonization and survival of these bacteria in urinary tract without causing infection [143146]. Although studies are different in terms of sample collection, inclusion criteria, methodology, etc., all of them have shown that human urinary microbiome vary according to age, sex, and disease [128,129,134]. The male urinary microbiome is mainly composed of the genus Corynebacterium, Staphylococcus, and Streptococcus [29,129,147]. Urinary microbiome alteration may occur for various reasons, including puberty, type of sexual behavior, urinary incontinence, and antimicrobial agents of prostatic secretions [145,146]. Dysbiosis affects immune molecules as well as response to treatment of urinary tract infections [148,149]. In this regard, changes in the frequency and diversity of microbiome have been shown in individuals with neurogenic bladder dysfunction (NBD), interstitial cystitis (IC), urinary incontinence, and sexually transmitted infections (STIs) [132134,144,148,150]. It is also possible that difference between urinary microbiome of men and women is responsible for the difference in the incidence and survival rate of bladder cancer between the sexes [151].

Inter-individual variation in urinary microbiome composition affects susceptibility to infection with STIs such as C. trachomatis and N. gonorrhoeae [133]. Studies also have shown the elevated PSA levels associated with STIs, which could be a sign of prostate involvement [152,153]. The history of inflammatory STIs can increase the risk of developing prostate cancer [57]. Moreover, the association between male urinary microbiome and prostate problems including prostatitis, benign prostatic hyperplasia (BPH) as well as prostate cancer has been explored previously [29,154,155]. Additional studies are needed to investigate the effect of urinary microbiome composition on progression of prostate cancer.

Prostate microbiome

There are many studies on the microbial composition of various parts of the body [156159], but little studies have been done on microbiome of healthy tissue of prostate as well as prostate tumor tissues [136,160]. It has not yet been elucidated whether there is prostate microbiome. Using the 16S rDNA PCR has not shown any bacteria in a healthy prostate. Some studies have shown the presence of bacteria in prostate cancer tissues [97,136,160162]. One of the concerns about the presence of microbiome in the prostate is treatment of false positive results due to contamination, which must be minimized by using aseptic methods, as well as negative controls [163,164].

In the study of Sfanos et al., in which negative control was also used, bacterial DNA of various species has been isolated from prostatectomy tissues. In this study, tissue cores of prostate were negative for bacterial DNA. Hence, it was concluded that there is no flora in the prostate and microorganisms in the focal regions are likely to be associated with prostate inflammation and are bacterial remnants within the macrophages. Using 16S sequencing, most of the strains were related to the normal flora of urethra (Acinetobacter spp., Prevotella spp., Actinomyces spp., Streptococcus spp., Pseudomonas spp.) or urinary tract infections (UTIs) (Escherichia spp., Pseudomonas spp., and Enterococci spp.) that highlighted the theory of colonization of the prostate by urine flora [136].

As mentioned above, inflammation plays an important role as stimulant for carcinogenesis. A change in bacterial populations, may lead to an increase of cancer risk by increasing inflammatory responses [53,135,165]. Some bacteria are known to have potential for creating inflammation in the prostate, including Enterobacteriaceae, such as E. coli and Pseudomonas, as well as the bacteria causing STIs [166,167].

In another study using ultradeep pyrosequencing (UDPS), the microbiome associated with the nontumor, peri-tumor, and tumor tissue of prostate was evaluated [97]. In that study, existence of prostate specific microbiome was reported and the dominant isolated bacterium was P. acne. Moreover, nontumor regions of prostate were considered as healthy prostate samples, and thus there were no confounding factors such as diet and lifestyle. High prevalence of Lactobacillales in nontumor regions as a normal microbiome of prostate was reported, which may play a role in maintaining the health of the prostate [97].

In summary, considering the antibacterial properties of prostatic fluid [168,169], and the impact of the urinary microbiome as well as skin and gut microbiome on the prostate, and finally according to all studies that have been done so far, there is no evidence to prove the definite existence of prostate microbiome. In all the studies that have been done so far, there are limitations that make it difficult to interpret their results. Further studies are needed in order to gain insight into characterization of the prostate tumor microenvironment.

Skin microbiome and prostate cancer

P. acnes is an anaerobic Gram-positive bacterium found in sebaceous follicles of the human skin, and its strains cause inflammatory diseases through their hemolytic, cytotoxic, and immunostimulatory activities. This bacterium was abundantly isolated from patients with prostate cancer [135,170]. It has been reported that P. acnes strains involved in prostate cancer were different in surface properties from strains associated with skin disorders. Studies have shown a powerful inflammatory activity of P. acnes in prostate tissue. In vitro studies on RWPE1 cells infected with P. acnes showed activation of NF-κB, the IL-6-Stat3, and the COX2-PGE2 pathways and increased transcriptional activation of IL-8, VEGF genes [171]. P. acnes infection results in the continuous degradation of IκBα (major NF-κB inhibitor) and subsequent activation of NF-κB, leading to positive regulation of genes involved in development and progression of prostate cancer [171174]. Furthermore, increased level of IL-6 in the serum of prostate cancer patients is associated with advanced metastases. IL-6 activates the JAK/STAT signaling pathway. Persistent activation of Stat3 transcription factor contribute to increase tumor growth and proliferation. VEGF and COX-2 are also important molecules in angiogenesis [172174].

Molecular pathological epidemiology (MPE) in the context of prostate cancer

MPE is a discipline combining epidemiology and pathology. The goal of pathology and epidemiology is clarifying etiology of disease, and MPE intends to achieve this aim at molecular, individual, and population levels. Generally, MPE employs tissue pathology resources and data available in epidemiological studies. Molecular epidemiology includes MPE and conventional molecular epidemiology as well as traditional disease designation systems [175]. In MPE, interrelationships between exposures including environmental, dietary, lifestyle and genetic factors; cellular or extracellular molecules alterations (molecular signature of disease); and evolution and progression of disease are evaluated [176].

Application of molecular signatures to improve the value of standard clinical-pathological parameters has affected clinical practice in several cancer types including prostate cancer. The molecular signatures indicative of disease grade and predictive of subsequent behavior could expedite the optimal treatment choice for prostate cancer. In this context, genome, immunity, and microbiome can be analyzed to identify new biomarkers for potential clinical utilities. The concept of MPE in clinical medicine is equivalent to precision medicine and personalized medicine [177]. Epidemiology provides analytical frameworks to evaluate the association of exposure (endogenous or exogenous factor) and incidence of a disease or its outcome [178]. It aims to identify patterns and determinants of health and disease conditions [178]. Conventional epidemiological researches comprise quantitative and qualitative study designs and examine association of an exposure and a disease entity in population-based cohorts. For example, genome-wide association studies (GWAS) implicate scanning markers across the genomes of numerous people to find genetic variations associated with a specific disease [178]. Currently, due to the emerging of molecular diagnostic tests in the field of infectious disease, molecular pathology has become a major subfield of pathology. Therefore, MPE has been emerged as an interdisciplinary integrative scientific discipline which analyses complex interplay between molecular pathological signatures, environment, lifestyle factors, disease occurrence and progression, by using large populations [178180]. MPE can be considered as the next step of GWAS, termed “GWAS-MPE approach.” It has demonstrated to be a promising approach to identify biomarkers for precision medicine [179,180].

As mentioned before, dysbiosis can have carcinogenic effects. Therefore, analyses of the microbiome in various body sites as well as pathologically transformed tissue (tumor) provide a basis for better understanding the cancer etiologies and their population impact. These analyses should be integrated into MPE, which is recognized as microbiology-MPE [178,181]. Microbiology-MPE affords a valuable approach to evaluate the interpersonal heterogeneity of the carcinogenic process related to the dysbiosis and to provide evidence for the role of microbiome in the processes of tumor development and progression [181].

In epidemiology, the term “exposure” designates any factor that may (or may not) associate with cause, prevent, or influence an outcome of interest [178]. In microbiology-MPE study, the microbial profile can be considered as an exposure or outcome variable. For instance, in cancer research, the microbial profile in non-cancerous tissue or biospecimen obtained before cancer diagnosis can be evaluated as an exposure associated with disease incidence as an outcome [178]. However, the detection of a certain microorganism may show either causal association with the tumor or can occur as a result of tumor development [178]. There are microbiology-MPE studies that have evaluated microbiome profile in prostate cancer patients and provided new insights into prostate cancer etiologies (See previous sections). Several studies have reported the association of microbial agents with prostate tumors (Table 1). In these studies, the composition of microbiome of different parts of human body, their interactions with the host and effects on host health have been widely considered. These studies have been carried out using advanced molecular techniques such as PCR-based targeted detection, sequencing and qPCR, and pyrosequencing. Their findings indicated differences in the diversity and abundance of microbes in healthy individuals and cancer patients [132,150,155,182,183]. Studies related to microbiome of different parts of the body associated with prostate cancer are discussed in their respective sections in this article (Table 1).

A current study used an array-based metagenomic analysis to define the prostate tumors microbiome in comparison with non-cancerous prostate tissues [184]. They defined the microbiome signatures associated with prostate cancer. They also suggested increased recombination activity in the tumor showing the viral and bacterial sequences integration into the somatic chromosomes of tumor cells. These integrations may affect host genes related to oncogenic activities in tumor cells [184]. Moreover, they compared the microbiome of cancerous prostate tissue with tissue from BPH patients and reported diverse and distinct prostate tumor microbiome in comparison with that of the controls [184]. Viral signatures including oncogenic human papillomavirus 18 (HPV18), Epstein-Barr virus (EBV), human cytomegalovirus (HCMV), and JC virus (JCV) in prostate cancers, were also reported previously [182,185,186].

Target the microbiome for risk assessment, prevention and treatment of prostate cancer

Cancer is a complex disease influenced by an interplay between host genetic diversity, immunology, and environmental factors. Despite genetic factors being involved in cancer, several studies have shown the influence of microorganisms on cancer biology [187]. In addition to metabolism-related genetic profiles, gut microbiome and its related metabolic properties also can potentially be important in cancer risk including prostate cancer [188]. There are some comprehensive review articles available on interactions between intake and species of gut microbes [189,190].

A biomarker is indicative of the severity or the presence of a particular disease, or pharmacologic responses to a therapeutic intervention [191]. A cancer biomarker refers to either a biological molecule secreted by a tumor or it can be a particular response of the body that is a sign of the presence of cancer. Cancer biomarkers are used as diagnostic markers to detect the presence of cancer, and prognostic markers to anticipate how well the body responds to a treatment [192]. Some of these markers have created paradigm shifts in personalized treatment of cancer [192]. Theranostics is a newly defined field of study intended to combine particular targeted therapy based on specific targeted diagnostic tests. It provides an evolution from conventional medicine to a contemporary personalized and accurate medicine approach. Today’s biomarker will be tomorrow’s theranostics for risk-stratification and treatment personalization [193]. Current researches are focused on the human microbiome as a potential early detection biomarker for diseases [194]. Some metagenomic markers have also potential for cancer diagnosis. Current evidences suggest that carcinogenesis proceeds by microbial factors. Therefore, several studies have examined the probability of profiling microbiome for cancer diagnosis [29,97,98,138,155, 195–198]. These studies have brought much hope in the possibility of microbiome manipulation to improve treatment efficacy and to reduce side effects (Table 1). Despite promising preliminary data, clinical use of these metagenomic markers needs to be supported by further studies [192].

Several factors including aging, lifestyle changes (in diet, exercise, nutrition, and stress), medications, and gastrointestinal pathogens can affect the microbiome composition with consequent change of inflammatory and pathophysiological states of various organs and tissues [199]. Lifestyle factors may influence prostate cancer susceptibility [200]. Consumption of meat [201], dairy products [202], eggs [203], and fish oil [204] has been studied in relation to prostate cancer risk. However, few studies have been performed on the interactions between how intake and metabolic factors influence cancer risk [188]. The performed studies are mostly limited to the characterization of genetic variation effect on metabolism or intake [205207]. For instance, previous studies examined the effect of a single-nucleotide polymorphism (rs4988235) on the lactase (LCT) gene and the effect of arachidonic acid metabolism gene polymorphisms on prostate cancer risk [206,207].

Microbiome composition exists in a precise balance that, if disrupted, dysbiosis occurs which has been considered as etiology of several cancers, including prostate cancer [208]. During dysbiosis, a decrease of Lentisphaerae, Bacteroides, and Parabacteroides has been seen. This condition can cause an increase in the serum levels of TNF-α, IL-8, IL-1β, and CRP [209].

Antibiotics cause the death of pathogens as well as the death of commensal bacteria that modulate gut microbiota composition [210]. The microbiome alteration depends on the antibiotic class, dose, and duration of antimicrobial exposure, their pharmacological action, and targeted bacteria [210]. There are some evidences that antibiotic-caused dysbiosis can increase the frequency of some cancers including prostate cancer which is increased by using penicillin, suggesting a link between the microbiome and carcinogenesis [211].

Moreover, gut microbiome metabolism impacts on digestion of dairy product, affects the composition of bioactive fatty acids in adipose tissue of host [212], and contributes to the production of carcinogenic metabolites and inflammation [213]. There are several metabolic pathways encoded among various microbes. These pathways may encode some rare functions including specific nutrient exploitation, antibiotic resistance, heavy metal utilization/resistance and the production of hormones, such as androgens [214,215]. Since our diet contains atypical compounds, the microbiome capable of utilizing them are rapidly enriched [216]. This microbial composition shift could have significant consequences for the host; for instance, intended medications may be modified or inactivated by the microbiome or may become toxic to the host [216].

Gut microbiome affects the host’s digestive process by enzymatic actions [188]. Some gut microbes contribute to the digestion of phenolic compounds from tea, coffee, and other plant-based diet sources into biologically active metabolites. Resveratrol belongs to a class of polyphenolic compounds and has potential antiinflammatory effects by modification of eicosanoid synthesis and inhibiting cytokines such as PTGS2, IL6, and TNF [217]. Hepatically conjugated estrogens excreted into the intestinal tract by bile can be deconjugated by bacterial β-glucuronidases, which leads to their reabsorption into the circulation [218,219]. A previous study showed that the intestinal microbial richness and alpha diversity affects the total urinary estrogen levels as well as risk for estrogen-related conditions [218]. Therefore, it was suggested that the gut microbiome should be considered as a biodynamic system interacting with its living environment and associates with disease risk [220].

In addition, gut microbiome can impact prostate cancer risk due to the presence of isoflavone-metabolizing, equol-producing bacteria. Equol, a nonsteroidal estrogen, may influence prostate cancer susceptibility. Slackia sp. strain NATTS, classified in the Coriobacteriacea family, belonging to the Slackia genus, is a newly identified human intestinal bacterium with a high equol-producing activity. It is capable of degrading one of the daidzeins in soy isoflavones into equol with high efficiency [221]. These can emphasize the role of microbiome in prostate cancer risk [221]. The first step to determine the influential role of gut microbiome in prostate cancer development is to evaluate key differences in the microbial profiles of men who do and do not progress aggressive disease [188]. Chemoprevention of prostate cancer has gained great interest in recent years. It has been suggested that incapability to convert daidzein to equol in the intestine due to lack of equol-converting bacteria in the intestinal environment can be considered as a risk factor for prostate cancer susceptibility. Therefore, using the NATTS strain bacteria to improve the intestinal environment as well as administration of S-equol-containing supplement can be considered as preventive options for prostate cancer [220].

Nowadays, immunotherapy is one of the successful treatment options for metastatic diseases. The main focus of cancer immunotherapy is on the modulation of T cells that are present in normal and cancerous prostate tissues [222,223]. Evidences suggest that the gut microbiome composition can have a significant effect on modulation of immunotherapy response and toxicity. Therefore, re-educating and/or diversifying the gut microbiome through using probiotics and prebiotics before or in combination with immunotherapies can lead to a better response rate [224226].

Current studies have reported that some bacteria might increase the effect of some traditional anticancer drugs and immunotherapy drugs. Local and systemic effects of gut microbiome on cancer is through various mechanisms that involve the innate and adaptive immunity, endocrine and neural pathways, bacterial products and toxins, modulation of the systemic inflammation, and the oxidative stress [199]. Since the extensive effect of microbiome on human health, microbiome composition differences between patients can be considered as a factor to decide who would benefit from a specific treatment modality [187]. In addition, the presence or absence of particular bacteria as well as their metabolites may affect the prevalence, severity, and treatment of a tumor and can serve as prognostic biomarkers [187]. In the foreseeable future, gut microbiome may be used as a biomarker to segregate healthy and cancer patients. Currently, cellular targets for improving chemotherapies and targeted therapies are the matters of interest. Therefore, microbial drug targets have the potential to improve the harmful side effects of chemotherapy as well [187].

Studies on animal models have shown that the gut microbiome has effect on antitumor activity of agents, such as cyclophosphamide [224] as well as anti-PDL1 [226] and CTLA-4 blockade immunotherapies [141]. Furthermore, animal model studies have shown that apoptotic activity of platinum-based antineoplastic compounds in the tumor cells is decreased in the absence of commensal microbes [224,227].

Human microbiome can affect a patient’s response to immune checkpoint inhibitors (ICIs) that mainly target the immune system rather than the patient’s tumor [216]. ICIs, such as programmed cell death protein (PD-1), have been reported effective in patients with advanced melanoma, non-small cell lung cancer (NSCLC) and renal cell carcinoma (RCC). The use of ICIs, such as monoclonal antibodies targeting PD-1 and its ligand (PD-L), promotes the memory T lymphocyte-mediated immune responses via suppressing the interaction of T inhibitory receptors with cognate ligand on tumor cells [228,229]. PD-1/PD-L1 ligand belongs to the superfamily of CD28/B7 and PD-1/PD-L1 signaling pathway inhibits the T cell mediated immune responses [230,231]. Studies have shown that the agents targeting PD-1 are useful in men who have received androgen deprivation therapy (ADT) with androgen receptor antagonist (enzalutamide) [232]. One study reported that antibiotic treatment before anti-PD-1 therapy had reduced the survival of patients compared to those who had not received antibiotics [28]. Antibiotic administration can also impair therapeutic outcomes of some chemotherapy drugs such as cyclophosphamide, highlighting the importance of the microbiome in patients’ response to these immune modulating agents [233].

A study on metastatic melanoma patients showed that the patients who responded to anti-PD-1 therapy had more diverse gut microbiome than non-responders [189]. Fecal microbiota transplantation (FMT) from healthy donors with a diverse microbiome profile could improve patient’s response to anti-PD-1 therapy. Moreover, antibiotic treatment for common infections including dental, pulmonary, and urinary has been shown to reduce significantly progression-free survival (PFS) and overall survival in patients treated with ICIs [228].

Microbes play an important role in pharmacokinetics, toxicity, and mechanism of action of chemotherapy as well as local and systemic immune responses. For example, Mycoplasma hyorhinis converts Gemcitabine into its deaminated inactive form [234]. Currently there is no approved second-line therapy for metastatic hormone-refractory prostate cancer (MHRPC) after docetaxel failure [235]. Metronomic administration of the cyclophosphamide has been shown preclinically to be an effective inhibitor of angiogenesis [236], and induces a decrease in circulating regulatory T cells, leading to the restoration of NK cell cytotoxicity, and proliferation of peripheral T cells [237]. Human clinical trials (Phase II) showed that cyclophosphamide administered in metronomic schedule has significant activity in HRPC [238240]. In addition, data from a study that assessed the efficacy and tolerance of metronomic cyclophosphamide administration in HRPC, suggested that metronomic cyclophosphamide/prednisolone chemotherapy might be advantageous for patients with docetaxel-resistant HRPC [235].

Cisplatin is a platinum (II) compound that represents clinical activity against several solid tumors, and was shown to have potential in management of castration-resistant prostate cancer (CRPC) [241]. The efficacy of alkylating antineoplastic agents (cyclophosphamide and cisplatin) can be affected by the microbiome. The efficiency of these compounds has been shown a great decrease in germ-free mice [242]. This efficacy reduction was also shown in other animal models receiving antibiotics to eradicate bacteria in the gut [243]. Moreover, chemotherapy can damage the diversity and health of gut microbiota by reducing the abundance of advantageous bacteria (e.g., Lactobacilli and Bifidobacteria), while increasing pathogenic bacteria (e.g., Clostridia and Enterobacteriaceae) [226]. Diversity is identified as an important factor for a healthy microbiome. Studies have shown that gut microbiome could be changed by castration or pelvic radiotherapy [183,244].

Abiraterone acetate (trade name: Zytiga) and enzalutamide (trade name: Xtandi) are antiandrogen medications used in the treatment of prostate cancer. Abiraterone acetate targets the biosynthesis of testosterone and enzalutamide directly binds to the androgen receptor (AR). Both of them are administered orally and have poor solubility, so they are in exposure of gut microbiome for a considerable time. This maximizes the probability of microbial modification. The acetate portion of abiraterone acetate can be a source of carbon for microorganisms [216].

There are two important pathways of drug metabolism. First, the drug may be modified into more active form or into inactive form by microbiome. Second, the microbiome may produce metabolites to decrease the drug’s efficacy, such as the regulation of testosterone in prostate cancer. In the absence of endogenous production of testosterone, this may act as a harmful mechanism in the context of prostate cancer patients. A previous study showed that a human gut bacterial species (Clostridium scindens) converts glucocorticoids into androgens. The implication is that not only the host endocrine system but also gut microbiome, may be the source of androgens [214].

Although the most microbiome studies have been done on the gut, chemotherapeutic agents may also affect microbial population of other sites of the human body [220].

Soy bean-derived products are considered as factors for reducing prostate cancer risk. Among these products, soy isoflavones and their interaction with gut microbiome has gained more attention [220]. Animal studies suggest that the gut microbiome is affected by circulating androgen levels and castration [244]. A previous study assessed the compositional profile of the gut microbiome in men with and without prostate cancer and with and without treatment with androgen receptor axis-targeted therapies (ATT) [29]. They reported that alpha diversity of the gut microbiome in men without prostate cancer is greater than the men with a prostate cancer diagnosis. They also found that men taking oral ATT had a different gut microbiome composition than men taking GNRH agonists/antagonists alone [29]. Studies of rodent models have shown that chemotherapy- and immunotherapy-caused dysbiosis of gut microbiome [225,245] could subsequently affect the local inflammatory environment in the intestinal tract, systemic inflammatory effects, and/or the administered cancer therapies efficacy [246]. Steroid biosynthesis takes place in prokaryotes [247] and some bacterial species are able to metabolize estrogen and androgen precursors and to catabolize them, so affect systemic levels of these hormones [214]. On the other hand, hormone levels can affect the microbiome [244]. The species capable of steroid/hormone biosynthesis were significantly more abundant in the gut flora of men taking oral ATT [246]. This finding may show an alternative mechanism for the production of steroid metabolites that could affect treatment response to oral ATT [246]. Moreover, overrepresentation of certain types of bacteria including Ruminococcaceae and particularly Akkermansia muciniphila was seen in the fecal microbiome of men taking oral ATT [246]. A series of human studies in melanoma patients have indicated the association between the presence of these same species (Ruminococcaceae, Bifidobacteriaceae, and Akkermansia muciniphila) and positive response to anti-PD-1 immunotherapy [229,248]. These studies may show that the gut microbiome has an important role both for therapeutic efficacy and as a target that could be modulated to improve treatment response. This influence may contribute to the variation in the effectiveness of immunotherapies [246].

Collectively, regarding the importance of gut microbiome content for the effectiveness of treatment as well as increasing the treatment response, therefore it is possible to reduce the toxicity and increase efficacy of chemotherapy by using symbiotics (combination of prebiotics and probiotics). The introduction of various useful bacteria as probiotics with anticancer properties forms the subject of many current studies [249].

Both probiotics and prebiotics have important roles in maintaining microbiota composition. Probiotics are microbial food supplements that may improve the gut microbiome balance and can increase the host’s immune response through numerous mechanisms [250]. They can stimulate the immunity by increasing the mucosal barrier function, enhancing the mucosal antibody production, and increasing the epithelial integrity and direct antagonism of pathogenic microorganisms [251]. One study has shown a relationship between the efficacy of CTLA-4 blockade and microbiota composition (B. fragilis and/or B. thetaiotaomicron and Burkholderiales) [252]. A previous in vivo study demonstrated the role of commensal Bifidobacterium in increasing antitumor immunity. They reported alteration of innate immune function and improved antitumor activity through an antigen-independent fashion. Oral administration of the Bifidobacteria alone or in combination with anti-PD-L1 immunotherapy in melanoma murine models led to a decrease in tumor growth [226]. Additionally, the antitumor activity of cisplatin could be restored by Lactobacillus acidophilus. Also this bacterium reduces the side effect of platins including nephrotoxicity [249].

Probiotics such as lactobacilli are found in dairy products [253]. A previous study showed that the production of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) was induced by lactobacilli in human immune cells [254]. Treatment with lactobacilli also have facilitated NK activity through TRAIL production against prostate cancer cells. TRAIL is an endogenous cytokine that induces apoptosis in tumor cells [254]. L. acidophilus and L. salivarius are commonly used for probiotic supplementation. L. acidophilus may help digesting lactose and L. salivarius can help kill Listeria [255,256].

Prebiotics are a type of fibers that are nonviable and indigestible compounds inducing the quantity and activity of specific gut microbiota including Bifidobacteria and Lactobacilli [257]. They are considered as a potential treatment option and have also been proposed as a supplement to repair chemotherapy-induced gut dysbiosis [258]. Prebiotics have a positive effect in rebalancing the gut microbiota and can stimulate the host to respond appropriately to immunotherapies [259]. Some of the prebiotics such as phytoestrogens downregulate the COX-2 mediated inflammation and have been shown to have preventive roles in cancer [199].

Fiber presents in many fruits and vegetables and is fermented by gut bacteria into short-chain fatty acids such as butyrate which has tumor suppressive properties [258]. A fiber-poor diet and rich of fat alters the microbiome-associated metabolites, such as vitamins B7 and B12 correlating with enhanced inflammatory state. Prolonged calorie restriction diets leads to an increase in Lactobacillus species [199]. In addition, the role of short-chain fatty acids produced by bacterial fermentation of fibers has been shown in protection against cancer development [187]. Therefore, using fiber-rich foods and prebiotics may help to decrease global cancer burden in the long run [187].

Conclusions and future perspectives

Recently, the role of the human microbiome in health and disease is highly regarded. There are several studies exploring the potential role of the microbiome that inhabit the human body (including gut, urinary, and skin) and prostate cancer. However, it is still blurred whether the human microbiome is causative or contributory to prostate cancer. It is noteworthy that the data at this time suggest the probable role of microbiome on this disease. The exact mechanisms of microbial involvement in prostate cancer development described in this review remain to be fully understood. The studies published so far utilizing molecular approaches to characterizing microbial diversity have radically changed our view of the human microbiome, consequently raising many questions about the human microbiota relationship and its relevance to cancer. Metagenomic analysis of whole microbiota composition and function could provide insight into these questions.

Advanced molecular analysis suggests significant differences in the gut microbiome of men with prostate cancer in comparison to benign controls, which may associate in the pathobiology of prostate cancer.

Overall, microbiome is linked to prostate health and disease and future investigations are needed to discover whether the human microbiota and/or their metabolites can be considered as novel biomarkers and therapeutic targets for prostate cancer. The mechanisms by which the human microbiome modulates carcinogenesis, including systemic inflammatory state, ability to affect systemic hormone levels, metabolism and genotoxicity, can provide opportunities to target the microbiome for diagnostic, preventive, and therapeutic strategies. Moreover, microbiology-MPE evaluating microbiome profile can provide new insights into the tumor-immune-microbiome interaction from human tissue and population-based data, so suggesting targeted microbiome-modulating strategies for prevention and treatment of prostate cancer.

References

[1]

Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin 2018; 68(1): 7–30

[2]

Center MM, Jemal A, Lortet-Tieulent J, Ward E, Ferlay J, Brawley O, Bray F. International variation in prostate cancer incidence and mortality rates. Eur Urol 2012; 61(6): 1079–1092

[3]

Kimura T, Egawa S. Epidemiology of prostate cancer in Asian countries. Int J Urol 2018; 25: 524–531

[4]

Nowroozi MR, Momeni SA, Ohadian Moghadam S, Ayati E, Mortazavi A, Arfae S, Jamshidian H, Taherimahmoudi M, Ayati M. Prostate-specific antigen density and gleason score predict adverse pathologic features in patients with clinically localized prostate cancer. Nephrourol Mon Nephrourol Mon 2016; 8(6): e39984

[5]

Moradpour F, Fatemi Z. Estimation of the projections of the incidence rates, mortality and prevalence due to common cancer site in Isfahan, Iran. Asian Pac J Cancer Prev 2013; 14(6): 3581–3585

[6]

Sfanos KS, Isaacs WB, De Marzo AM. Infections and inflammation in prostate cancer. Am J Clin Exp Urol 2013; 1(1): 3–11

[7]

Peisch SF, Van Blarigan EL, Chan JM, Stampfer MJ, Kenfield SA. Prostate cancer progression and mortality: a review of diet and lifestyle factors. World J Urol 2017; 35(6): 867–874

[8]

Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature 2007; 449(7164): 804–810

[9]

Uemura N, Okamoto S, Yamamoto S, Matsumura N, Yamaguchi S, Yamakido M, Taniyama K, Sasaki N, Schlemper RJ. Helicobacter pylori infection and the development of gastric cancer. N Engl J Med 2001; 345(11): 784–789

[10]

Sheh A, Fox JG. The role of the gastrointestinal microbiome in Helicobacter pylori pathogenesis. Gut Microbes 2013; 4(6): 505–531

[11]

DeGruttola AK, Low D, Mizoguchi A, Mizoguchi E. Current understanding of dysbiosis in disease in human and animal models. Inflamm Bowel Dis 2016; 22(5): 1137–1150

[12]

Hartstra AV, Bouter KE, Bäckhed F, Nieuwdorp M. Insights into the role of the microbiome in obesity and type 2 diabetes. Diabetes Care 2015; 38(1): 159–165

[13]

Carbonero F, Benefiel AC, Alizadeh-Ghamsari AH, Gaskins HR. Microbial pathways in colonic sulfur metabolism and links with health and disease. Front Physiol 2012; 3: 448

[14]

Huycke MM, Gaskins HR. Commensal bacteria, redox stress, and colorectal cancer: mechanisms and models. Exp Biol Med (Maywood) 2004; 229(7): 586–597

[15]

Wang X, Huycke MM. Extracellular superoxide production by Enterococcus faecalis promotes chromosomal instability in mammalian cells. Gastroenterology 2007; 132(2): 551–561

[16]

Wang X, Yang Y, Moore DR, Nimmo SL, Lightfoot SA, Huycke MM. 4-hydroxy-2-nonenal mediates genotoxicity and bystander effects caused by enterococcus faecalis-infected macrophages. Gastroenterology 2012; 142: 543–551

[17]

Nesić D, Hsu Y, Stebbins CE. Assembly and function of a bacterial genotoxin. Nature 2004; 429: 429–433

[18]

Liang W, Ferrara N. The complex role of neutrophils in tumor angiogenesis and metastasis. Cancer Immunol Res 2016; 4: 83–91

[19]

Mima K, Cao Y, Chan AT, Qian ZR, Nowak JA, Masugi Y, . Fusobacterium nucleatum in colorectal carcinoma tissue according to tumor location. Clin Transl Gastroenterol 2016; 7: e200

[20]

Cougnoux A, Dalmasso G, Martinez R, Buc E, Delmas J, Gibold L, . Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut 2014; 63: 1932–1942

[21]

Zhang Q, Yu N, Lee C. Mysteries of TGF-β paradox in benign and malignant cells. Front Oncol 2014; 4: 94

[22]

Menzies BE. The role of fibronectin binding proteins in the pathogenesis of Staphylococcus aureus infections. Curr Opin Infect Dis 2003; 16: 225–229

[23]

Li N, Ren A, Wang X, Fan X, Zhao Y, Gao GF, Cleary P, Wang B. Influenza viral neuraminidase primes bacterial coinfection through TGF-β-mediated expression of host cell receptors. Proc Natl Acad Sci USA 2015; 112(1): 238–243

[24]

Jakowlew SB. Transforming growth factor-β in cancer and metastasis. Cancer Metastasis Rev 2006; 25(3): 435–457

[25]

Bostwick DG, de la Roza G, Dundore P, Corica FA, Iczkowski KA. Intraepithelial and stromal lymphocytes in the normal human prostate. Prostate 2003; 55: 187–193

[26]

Dikov D, Bachurska S, Staikov D, Sarafian V. Intraepithelial lymphocytes in relation to NIH category IV prostatitis in autopsy prostate. Prostate 2015; 75(10): 1074–1084

[27]

Fujii T, Shimada K, Asai O, Tanaka N, Fujimoto K, Hirao K, Konishi N. Immunohistochemical analysis of inflammatory cells in benign and precancerous lesions and carcinoma of the prostate. Pathobiology 2013; 80(3): 119–126

[28]

Sfanos KS, Yegnasubramanian S, Nelson WG, De Marzo AM. The inflammatory microenvironment and microbiome in prostate cancer development. Nat Rev Urol 2018; 15: 11–24

[29]

Shrestha E, White JR, Yu SH, Kulac I, Ertunc O, De Marzo AM, Yegnasubramanian S, Mangold LA, Partin AW, Sfanos KS. Profiling the urinary microbiome in men with positive versus negative biopsies for prostate cancer. J Urol 2018; 199(1): 161–171

[30]

Virchow R. An address on the value of pathological experiments. Br Med J 1881; 2(1075): 198–203

[31]

Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell 2010; 140(6): 883–899

[32]

Delongchamps NB, de la Roza G, Chandan V, Jones R, Sunheimer R, Threatte G, . Evaluation of prostatitis in autopsied prostates—is chronic inflammation more associated with benign prostatic hyperplasia or cancer? J Urol 2008; 179(5): 1736–1740

[33]

Stark T, Livas L, Kyprianou N. Inflammation in prostate cancer progression and therapeutic targeting. Transl Androl Urol 2015; 4(4): 455–463

[34]

Maynard CL, Elson CO, Hatton RD, Weaver CT. Reciprocal interactions of the intestinal microbiota and immune system. Nature 2012; 489(7415): 231–241

[35]

Goris H, de Boer F, van der Waaij D. Myelopoiesis in experimentally contaminated specific-pathogen-free and germfree mice during oral administration of polymyxin. Infect Immun 1985; 50(2): 437–441

[36]

Khosravi A, Yáñez A, Price JG, Chow A, Merad M, Goodridge HS, . Gut microbiota promote hematopoiesis to control bacterial infection. Cell Host Microbe 2014; 15(3): 374–381

[37]

Schwabe RF, Jobin C. The microbiome and cancer. Nat Rev Cancer 2013; 13(11): 800–812

[38]

Sheflin AM, Whitney AK, Weir TL. Cancer-promoting effects of microbial dysbiosis. Curr Oncol Rep 2014; 16(10): 406

[39]

Horbinski C, Mojesky C, Kyprianou N. Live free or die: tales of homeless (cells) in cancer. Am J Pathol 2010; 177: 1044–1052

[40]

Galdiero MR, Bonavita E, Barajon I, Garlanda C, Mantovani A, Jaillon S. Tumor associated macrophages and neutrophils in cancer. Immunobiology 2013; 218(11): 1402–1410

[41]

Puhr M, De Marzo A, Isaacs W, Lucia MS, Sfanos K, Yegnasubramanian S, . Inflammation, microbiota, and prostate cancer. Eur Urol Focus 2016; 2(4): 374–382

[42]

Barron DA, Rowley DR. The reactive stroma microenvironment and prostate cancer progression. Endocr Relat Cancer 2012; 19(6): R187–204

[43]

Frisch SM, Screaton RA. Anoikis mechanisms. Curr Opin Cell Biol 2001; 13(5): 555–562

[44]

Armstrong H, Bording-Jorgensen M, Dijk S, Wine E. The complex interplay between chronic inflammation, the microbiome, and cancer: understanding disease progression and what we can do to prevent it. Cancers (Basel) 2018; 10(3): 83

[45]

Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet 2001; 357: 539–545

[46]

Coussens LM, Werb Z. Inflammation and cancer. Nature 2002; 420(6917): 860–867

[47]

Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 2009; 30(7): 1073–1081

[48]

Francescone R, Hou V, Grivennikov SI. Microbiome, inflammation, and cancer. Cancer J 2014; 20(3): 181–189

[49]

Dzutsev A, Badger JH, Perez-Chanona E, Roy S, Salcedo R, Smith CK, Trinchieri G. Microbes and cancer. Annu Rev Immunol 2017; 35(1): 199–228

[50]

Schatteman PH, Hoekx L, Wyndaele JJ, Jeuris W, Van Marck E. Inflammation in prostate biopsies of men without prostatic malignancy or clinical prostatitis: correlation with total serum PSA and PSA density. Eur Urol 2000; 37(4): 404–412

[51]

Gurel B, Lucia MS, Thompson IM Jr, Goodman PJ, Tangen CM, Kristal AR, . Chronic inflammation in benign prostate tissue is associated with high-grade prostate cancer in the placebo arm of the prostate cancer prevention trial. Cancer Epidemiol Biomarkers Prev 2014; 23(5): 847–856

[52]

De Nunzio C, Kramer G, Marberger M, Montironi R, Nelson W, Schröder F, . The controversial relationship between benign prostatic hyperplasia and prostate cancer: the role of inflammation. Eur Urol 2011; 60(1): 106–117

[53]

Shinohara DB, Vaghasia AM, Yu SH, Mak TN, Brüggemann H, Nelson WG, . A mouse model of chronic prostatic inflammation using a human prostate cancer-derived isolate of Propionibacterium acnes. Prostate 2013; 73(9): 1007–1015

[54]

Elkahwaji JE, Zhong W, Hopkins WJ, Bushman W. Chronic bacterial infection and inflammation incite reactive hyperplasia in a mouse model of chronic prostatitis. Prostate 2007; 67(1): 14–21

[55]

Pelouze PS. Gonorrhea in the male and female: a book for practitioners. Philadelphia: W. B. Saunders Company, 1935

[56]

Poletti F, Medici MC, Alinovi A, Menozzi MG, Sacchini P, Stagni G, . Isolation of Chlamydia trachomatis from the prostatic cells in patients affected by nonacute abacterial prostatitis. J Urol 1985; 134(4): 691–693

[57]

Hayes RB, Pottern LM, Strickler H, Rabkin C, Pope V, Swanson GM, . Sexual behavior, STDs and risks for prostate cancer. Br J Cancer 2000; 82(3): 718–725

[58]

Maeda H, Akaike T. Nitric oxide and oxygen radicals in infection, inflammation, and cancer. Biochemistry (Mosc) 1998; 63(7): 854–865

[59]

De Marzo AM, Platz EA, Sutcliffe S, Xu J, Grönberg H, Drake CG, Nakai Y, Isaacs WB, Nelson WG. Inflammation in prostate carcinogenesis. Nat Rev Cancer 2007; 7(4): 256–269

[60]

Bultman SJ. Emerging roles of the microbiome in cancer. Carcinogenesis 2014; 35(2): 249–255

[61]

Buchta Rosean CM, Rutkowski MR. The influence of the commensal microbiota on distal tumor-promoting inflammation. Semin Immunol 2017; 32: 62–73

[62]

Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, . Linking the human gut microbiome to inflammatory cytokine production capacity. Cell 2016; 167(4): 1125–1136.e8

[63]

Chu WM. Tumor necrosis factor. Cancer Lett 2013; 328: 222–225

[64]

Suh J, Rabson AB. NF-κB activation in human prostate cancer: important mediator or epiphenomenon? J Cell Biochem 2004; 91: 100–117

[65]

Lee CH, Jeon YT, Kim SH, Song YS. NF-κB as a potential molecular target for cancer therapy. Biofactors 2007; 29(1): 19–35

[66]

Ohadian Moghadam S, Nowroozi MR. Toll-like receptors: the role in bladder cancer development, progression and immunotherapy. Scand J Immunol 2019: e12818

[67]

Harmey JH, Bucana CD, Lu W, Byrne AM, McDonnell S, Lynch C, . Lipopolysaccharide-induced metastatic growth is associated with increased angiogenesis, vascular permeability and tumor cell invasion. Int J Cancer 2002; 101(5): 415–422

[68]

Simon F, Fernández R. Early lipopolysaccharide-induced reactive oxygen species production evokes necrotic cell death in human umbilical vein endothelial cells. J Hypertens. 2009; 27(6): 1202–1216

[69]

Xu XL, Lee RT, Fang HM, Wang YM, Li R, Zou H, Zhu Y, Wang Y. Bacterial peptidoglycan triggers Candida albicans hyphal growth by directly activating the adenylyl cyclase Cyr1p. Cell Host Microbe 2008; 4(1): 28–39

[70]

Maneval ML, Eckert KA. Effects of oxidative and alkylating damage on microsatellite instability in nontumorigenic human cells. Mutat Res 2004; 546: 29–38

[71]

Cheema AK, Maier I, Dowdy T, Wang Y, Singh R, Ruegger PM, . Chemopreventive metabolites are correlated with a change in intestinal microbiota measured in A-T mice and decreased carcinogenesis. PLoS One 2016; 11: e0151190

[72]

Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012; 486(7402): 207–214

[73]

van der Meulen TA, Harmsen H, Bootsma H, Spijkervet F, Kroese F, Vissink A. The microbiome-systemic diseases connection. Oral Dis 2016; 22(8): 719–734

[74]

Pabst O. Correlation, consequence, and functionality in microbiome-immune interplay. Immunol Rev 2017; 279(1): 4–7

[75]

Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, . Enterotypes of the human gut microbiome. Nature 2011; 473: 174–180

[76]

Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano GAD, Gasbarrini A, . What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms 2019; 7(1): 14

[77]

Huggins C, Hodges CV. Studies on prostatic cancer. I. The effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. 1941. J Urol 2002; 167(2 Pt 2): 948–952

[78]

Strobl FJ, Levine JE. Estrogen inhibits luteinizing hormone (LH), but not follicle-stimulating hormone secretion in hypophysectomized pituitary-grafted rats receiving pulsatile LH-releasing hormone infusions. Endocrinology 1988; 123(1): 622–630

[79]

Geier R, Adler S, Rashid G, Klein A. The synthetic estrogen diethylstilbestrol (DES) inhibits the telomerase activity and gene expression of prostate cancer cells. Prostate 2010; 70(12): 1307–1312

[80]

Thelen P, Wuttke W, Jarry H, Grzmil M, Ringert RH. Inhibition of telomerase activity and secretion of prostate specific antigen by silibinin in prostate cancer cells. J Urol 2004; 171(5): 1934–1938

[81]

Plottel CS, Blaser MJ. Microbiome and malignancy. Cell Host Microbe 2011; 10: 324–335

[82]

Cavalieri E, Chakravarti D, Guttenplan J, Hart E, Ingle J, Jankowiak R, Muti P, Rogan E, Russo J, Santen R, Sutter T. Catechol estrogen quinones as initiators of breast and other human cancers: implications for biomarkers of susceptibility and cancer prevention. Biochim Biophys Acta 2006; 1766(1): 63–78

[83]

Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen-gut microbiome axis: physiological and clinical implications. Maturitas 2017; 103: 45–53

[84]

Nelles JL, Hu WY, Prins GS. Estrogen action and prostate cancer. Expert Rev Endocrinol Metab 2011; 6: 437–451

[85]

Gadelle D, Raibaud P, Sacquet E. β-glucuronidase activities of intestinal bacteria determined both in vitro and in vivo in gnotobiotic rats. Appl Environ Microbiol 1985; 49(3): 682–685

[86]

Gloux K, Berteau O, Oumami H, Beguet F, Leclerc M, Dore J. A metagenomic β-glucuronidase uncovers a core adaptive function of the human intestinal microbiome. Proc Natl Acad Sci USA 2011; 108: 4539–4546

[87]

Golombos DM, Ayangbesan A, O’Malley P, Lewicki P, Barlow L, Barbieri CE, Chan C, DuLong C, Abu-Ali G, Huttenhower C, Scherr DS. The role of gut microbiome in the pathogenesis of prostate cancer: a prospective, pilot study. Urology 2018; 111: 122–128

[88]

Duncan SH, Barcenilla A, Stewart CS, Pryde SE, Flint HJ. Acetate utilization and butyryl coenzyme A (CoA):acetate-CoA transferase in butyrate-producing bacteria from the human large intestine. Appl Environ Microbiol 2002; 68(10): 5186–5190

[89]

Cockburn DW, Orlovsky NI, Foley MH, Kwiatkowski KJ, Bahr CM, Maynard M, Demeler B, Koropatkin NM. Molecular details of a starch utilization pathway in the human gut symbiont Eubacterium rectale. Mol Microbiol 2015; 95(2): 209–230

[90]

Hamer HM, Jonkers D, Venema K, Vanhoutvin S, Troost FJ, Brummer RJ. The role of butyrate on colonic function. Aliment Pharmacol Ther 2008; 27: 104–119

[91]

Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermúdez-Humarán LG, Gratadoux JJ, Blugeon S, Bridonneau C, Furet JP, Corthier G, Grangette C, Vasquez N, Pochart P, Trugnan G, Thomas G, Blottière HM, Dorv J, Marteau P, Seksik P, Langella P. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci USA 2008; 105(43): 16731–16736

[92]

Kwa M, Plottel CS, Blaser MJ, Adams S. The intestinal microbiome and estrogen receptor-positive female breast cancer. J Natl Cancer Inst 2016; 22: 108

[93]

Hullar MA, Burnett-Hartman AN, Lampe JW. Gut microbes, diet, and cancer. Cancer Treat Res 2014; 159: 377–399

[94]

Schwabe RF, Jobin C. The microbiome and cancer. Nat Rev Cancer 2013; 13(11): 800–812

[95]

Price AJ, Travis RC, Appleby PN, Albanes D, Barricarte Gurrea A, Bjørge T, . Circulating folate and vitamin B12 and risk of prostate cancer: a collaborative analysis of individual participant data from six cohorts including 6875 cases and 8104 controls. Eur Urol 2016; 70: 941–951

[96]

Wang R, Zheng Y, Huang JY, Zhang AQ, Zhou YH, Wang JN. Folate intake, serum folate levels, and prostate cancer risk: a meta-analysis of prospective studies. BMC Public Health 2014; 14: 1326

[97]

Cavarretta I, Ferrarese R, Cazzaniga W, Saita D, Lucianò R, Ceresola ER, Locatelli I, Visconti L, Lavorgna G, Briganti A, Nebuloni M, Doglioni C, Clementi M, Montorsi F, Canducci F, Salonia A. The microbiome of the prostate tumor microenvironment. Eur Urol 2017; 72(4): 625–631

[98]

Liss MA, White JR, Goros M, Gelfond J, Leach R, Johnson-Pais T, Lai Z, Rourke E, Basler J, Ankerst D, Shah DP. Metabolic biosynthesis pathways identified from fecal microbiome associated with prostate cancer. Eur Urol 2018; 74(5): 575–582

[99]

James SJ, Basnakian AG, Miller BJ. In vitro folate deficiency induces deoxynucleotide pool imbalance, apoptosis, and mutagenesis in Chinese hamster ovary cells. Cancer Res 1994; 54(19): 5075–5080

[100]

Wickramasinghe SN, Fida S. Misincorporation of uracil into the DNA of folate- and B12-deficient HL60 cells. Eur J Haematol 1993; 50(3): 127–132

[101]

Duthie SJ, Hawdon A. DNA instability (strand breakage, uracil misincorporation, and defective repair) is increased by folic acid depletion in human lymphocytes in vitro. FASEB J 1998; 12(14): 1491–1497

[102]

Pompei A, Cordisco L, Amaretti A, Zanoni S, Matteuzzi D, Rossi M. Folate production by bifidobacteria as a potential probiotic property. Appl Environ Microbiol 2007; 73(1): 179–185

[103]

Rodriguez-Melendez R, Griffin JB, Zempleni J. Biotin supplementation increases expression of the cytochrome P450 1B1 gene in Jurkat cells, increasing the occurrence of single-stranded DNA breaks. J Nutr 2004; 134(9): 2222–2228

[104]

Dewhirst FE, Chen T, Izard J, Paster BJ, Tanner AC, Yu WH, Lakshmanan A, Wade WG. The human oral microbiome. J Bacteriol 2010; 192(19): 5002–5017

[105]

Michaud DS, Izard J, Wilhelm-Benartzi CS, You DH, Grote VA, Tjønneland A, Dahm CC, Overvad K, Jenab M, Fedirko V, Boutron-Ruault MC, Clavel-Chapelon F, Racine A, Kaaks R, Boeing H, Foerster J, Trichopoulou A, Lagiou P, Trichopoulos D, Sacerdote C, Sieri S, Palli D, Tumino R, Panico S, Siersema PD, Peeters PH, Lund E, Barricarte A, Huerta JM, Molina-Montes E, Dorronsoro M, Quirós JR, Duell EJ, Ye W, Sund M, Lindkvist B, Johansen D, Khaw KT, Wareham N, Travis RC, Vineis P, Bueno-de-Mesquita HB, Riboli E. Plasma antibodies to oral bacteria and risk of pancreatic cancer in a large European prospective cohort study. Gut 2013; 62(12): 1764–1770

[106]

Beck JD, Offenbacher S. Systemic effects of periodontitis: epidemiology of periodontal disease and cardiovascular disease. J Periodontol 2005; 76(11S): 2089–2100

[107]

Joshipura KJ, Rimm EB, Douglass CW, Trichopoulos D, Ascherio A, Willett WC. Poor oral health and coronary heart disease. J Dent Res 1996; 75(9): 1631–1636

[108]

Offenbacher S, Jared HL, O’Reilly PG, Wells SR, Salvi GE, Lawrence HP, Socransky SS, Beck JD. Potential pathogenic mechanisms of periodontitis associated pregnancy complications. Ann Periodontol 1998; 3(1): 233–250

[109]

Hujoel PP, Drangsholt M, Spiekerman C, Weiss NS. An exploration of the periodontitis-cancer association. Ann Epidemiol 2003; 13(5): 312–316

[110]

Famili P, Cauley JA, Greenspan SL. The effect of androgen deprivation therapy on periodontal disease in men with prostate cancer. J Urol 2007; 177(3): 921–924

[111]

Krieger JN, Nyberg L Jr, Nickel JC. NIH consensus definition and classification of prostatitis. JAMA 1999; 282(3): 236–237

[112]

Offenbacher S. Periodontal diseases: pathogenesis. Ann Periodontol 1996; 1(1): 821–878

[113]

Jang TL, Schaeffer AJ. The role of cytokines in prostatitis. World J Urol 2003; 21(2): 95–99

[114]

Van Dyke TE, van Winkelhoff AJ. Infection and inflammatory mechanisms. J Periodontol 2013; 84(4 Suppl): S1–S7

[115]

Joshi N, Bissada NF, Bodner D, Maclennan GT, Narendran S, Jurevic R, Skillicorn R. Association between periodontal disease and prostate-specific antigen levels in chronic prostatitis patients. J Periodontol 2010; 81(6): 864–869

[116]

Alwithanani N, Bissada NF, Joshi N. Periodontal treatment improves prostate symptoms and lowers serum PSA in men with high PSA and chronic periodontitis. Dentistry 2015; 5: 1–4

[117]

Hasui Y, Marutsuka K, Asada Y, Ide H, Nishi S, Osada Y. Relationship between serum prostate specific antigen and histological prostatitis in patients with benign prostatic hyperplasia. Prostate 1994; 25(2): 91–96

[118]

Kandirali E, Boran C, Serin E, Semercioz A, Metin A. Association of extent and aggressiveness of inflammation with serum PSA levels and PSA density in asymptomatic patients. Urology 2007; 70(4): 743–747

[119]

Noack B, Genco RJ, Trevisan M, Grossi S, Zambon JJ, De Nardin E. Periodontal infections contribute to elevated systemic C-reactive protein level. J Periodontol 2001; 72(9): 1221–1227

[120]

Estemalik J, Demko C, Bissada NF, Joshi N, Bodner D, Shankar E, Gupta S. Simultaneous detection of oral pathogens in subgingival plaque and prostatic fluid of men with periodontal and prostatic diseases. J Periodontol 2017; 88(9): 823–829

[121]

Kadowaki T, Nakayama K, Yoshimura F, Okamoto K, Abe N, Yamamoto K. Arg-gingipain acts as a major processing enzyme for various cell surface proteins in Porphyromonas gingivalis. J Biol Chem 1998; 273(44): 29072–29076

[122]

Saglie FR, Marfany A, Camargo P. Intragingival occurrence of Actinobacillus actinomycetemcomitans and Bacteroides gingivalis in active destructive periodontal lesions. J Periodontol 1988; 59(4): 259–265

[123]

Fan X, Alekseyenko AV, Wu J, Peters BA, Jacobs EJ, Gapstur SM, Purdue MP, Abnet CC, Stolzenberg-Solomon R, Miller G, Ravel J, Hayes RB, Ahn J. Human oral microbiome and prospective risk for pancreatic cancer: a population-based nested case-control study. Gut 2018; 67(1): 120–127

[124]

Stathopoulou PG, Benakanakere MR, Galicia JC, Kinane DF. The host cytokine response to Porphyromonas gingivalis is modified by gingipains. Oral Microbiol Immunol 2009; 24(1): 11–17

[125]

Duncan L, Yoshioka M, Chandad F, Grenier D. Loss of lipopolysaccharide receptor CD14 from the surface of human macrophage-like cells mediated by Porphyromonas gingivalis outer membrane vesicles. Microb Pathog 2004; 36(6): 319–325

[126]

Whiteside SA, Razvi H, Dave S, Reid G, Burton JP. The microbiome of the urinary tract—a role beyond infection. Nat Rev Urol 2015; 12(2): 81–90

[127]

Wolfe AJ, Toh E, Shibata N, Rong R, Kenton K, Fitzgerald M, . Evidence of uncultivated bacteria in the adult female bladder. J Clin Microbiol 2012; 50: 1376–1383

[128]

Hilt EE, McKinley K, Pearce MM, Rosenfeld AB, Zilliox MJ, Mueller ER, . Urine is not sterile: use of enhanced urine culture techniques to detect resident bacterial flora in the adult female bladder. J Clin Microbiol 2014; 52: 871–876

[129]

Lewis DA, Brown R, Williams J, White P, Jacobson SK, Marchesi JR, . The human urinary microbiome; bacterial DNA in voided urine of asymptomatic adults. Front Cell Infect Microbiol 2013; 3: 41

[130]

Porter CM, Shrestha E, Peiffer LB, Sfanos KS. The microbiome in prostate inflammation and prostate cancer. Prostate Cancer Prostatic Dis 2018; 21: 345–354

[131]

Kirby RS, Lowe D, Bultitude MI, Shuttleworth KE. Intra-prostatic urinary reflux: an aetiological factor in abacterial prostatitis. Br J Urol 1982; 54(6): 729–731

[132]

Fouts DE, Pieper R, Szpakowski S, Pohl H, Knoblach S, Suh MJ, . Integrated next-generation sequencing of 16S rDNA and metaproteomics differentiate the healthy urine microbiome from asymptomatic bacteriuria in neuropathic bladder associated with spinal cord injury. J Transl Med 2012; 28: 174

[133]

Nelson DE, Van Der Pol B, Dong Q, Revanna KV, Fan B, Easwaran S, . Characteristic male urine microbiomes associate with asymptomatic sexually transmitted infection. PLoS One 2010; 5: e14116

[134]

Dong Q, Nelson DE, Toh E, Diao L, Gao X, Fortenberry JD, . The microbial communities in male first catch urine are highly similar to those in paired urethral swab specimens. PLoS One 2011; 6: e19709

[135]

Cohen RJ, Shannon BA, McNeal JE, Shannon T, Garrett KL. Propionibacterium acnes associated with inflammation in radical prostatectomy specimens: a possible link to cancer evolution? J Urol 2005; 173(6): 1969–1974

[136]

Sfanos KS, Sauvageot J, Fedor HL, Dick JD, De Marzo AM, Isaacs WB. A molecular analysis of prokaryotic and viral DNA sequences in prostate tissue from patients with prostate cancer indicates the presence of multiple and diverse microorganisms. Prostate 2008; 68(3): 306–320

[137]

Mak TN, Yu SH, De Marzo AM, Brüggemann H, Sfanos KS. Multi-locus sequence typing (MLST) analysis of Propionibacterium acnes isolates from radical prostatectomy specimens. Prostate 2013; 73: 770–777

[138]

Sfanos KS, Isaacs WB. An evaluation of PCR primer sets used for detection of Propionibacterium acnes in prostate tissue samples. Prostate 2008; 68: 1492–1495

[139]

Davidsson S, Mölling P, Rider JR, Unemo M, Karlsson MG, Carlsson J, . Frequency and typing of Propionibacterium acnes in prostate tissue obtained from men with and without prostate cancer. Infect Agent Cancer 2016; 11: 26

[140]

Brede CM, Shoskes DA. The etiology and management of acute prostatitis. Nat Rev Urol 2011; 8: 207–212

[141]

Sasaki M, Yamaura C, Ohara-Nemoto Y, Tajika S, Kodama Y, Ohya T, Harada R, Kimura S. Streptococcus anginosus infection in oral cancer and its infection route. Oral Dis 2005; 11(3): 151–156

[142]

Shiga K, Tateda M, Saijo S, Hori T, Sato I, Tateno H, Matsuura K, Takasaka T, Miyagi T. Presence of Streptococcus infection in extra-oropharyngeal head and neck squamous cell carcinoma and its implication in carcinogenesis. Oncol Rep 2001; 8(2): 245–248

[143]

Fricke WF, Maddox C, Song Y, Bromberg JS. Human microbiota characterization in the course of renal transplantation. Am J Transplant 2014; 14: 416–427

[144]

Siddiqui H, Lagesen K, Nederbragt AJ, Jeansson SL, Jakobsen KS. Alterations of microbiota in urine from women with interstitial cystitis. BMC Microbiol 2012; 12: 205

[145]

Stapleton AE. Urinary tract infection pathogenesis: host factors. Infect Dis Clin North Am 2014; 28: 149–159

[146]

Ragnarsdóttir B, Lutay N, Grönberg-Hernandez J, Köves B, Svanborg C. Genetics of innate immunity and UTI susceptibility. Nat Rev Urol 2011; 8: 449–468

[147]

Gottschick C, Deng ZL, Vital M, Masur C, Abels C, Pieper DH, Wagner-Döbler I. The urinary microbiota of men and women and its changes in women during bacterial vaginosis and antibiotic treatment. Microbiome 2017; 5: 99

[148]

Pearce MM, Hilt EE, Rosenfeld AB, Zilliox MJ, Thomas-White K, Fok C, . The female urinary microbiome: a comparison of women with and without urgency urinary incontinence. MBio 2014; 5: e01283–14

[149]

Nienhouse V, Gao X, Dong Q, Nelson DE, Toh E, McKinley K, . Interplay between bladder microbiota and urinary antimicrobial peptides: mechanisms for human urinary tract infection risk and symptom severity. PLoS One 2014; 9: e114185

[150]

Siddiqui H, Nederbragt AJ, Lagesen K, Jeansson SL, Jakobsen KS. Assessing diversity of the female urine microbiota by high throughput sequencing of 16S rDNA amplicons. BMC Microbiol 2011; 11: 244

[151]

Micheli A, Ciampichini R, Oberaigner W, Ciccolallo L, de Vries E, Izarzugaza I, . The advantage of women in cancer survival: an analysis of EUROCARE-4 data. Eur J Cancer 2009; 45: 1017–1027

[152]

Sutcliffe S, Zenilman JM, Ghanem KG, Jadack RA, Sokoll LJ, Elliott DJ, . Sexually transmitted infections and prostatic inflammation/cell damage as measured by serum prostate specific antigen concentration. J Urol 2006; 175: 1937–1942

[153]

Huang WY, Hayes R, Pfeiffer R, Viscidi RP, Lee FK, Wang YF, Reding D, Whitby D, Papp JR, Rabkin CS. Sexually transmissible infections and prostate cancer risk. Cancer Epidemiol Biomarkers Prev 2008; 17(9): 2374–2381

[154]

Shoskes DA, Altemus J, Polackwich AS, Tucky B, Wang H, Eng C. The urinary microbiome differs significantly between patients with chronic prostatitis/chronic pelvic pain syndrome and controls as well as between patients with different clinical phenotypes. Urology 2016; 92: 26–32

[155]

Yu H, Meng H, Zhou F, Ni X, Shen S, Das UN. Urinary microbiota in patients with prostate cancer and benign prostatic hyperplasia. Arch Med Sci 2015; 11(2): 385–394

[156]

Holmes E, Li JV, Athanasiou T, Ashrafian H, Nicholson JK. Understanding the role of gut microbiome-host metabolic signal disruption in health and disease. Trends Microbiol 2011; 19: 349–359

[157]

Cox AJ, West NP, Cripps AW. Obesity, inflammation, and the gut microbiota. Lancet Diabetes Endocrinol 2015; 3: 207–215

[158]

Zitvogel L, Galluzzi L, Viaud S, Vétizou M, Daillère R, Merad M, Kroemer G. Cancer and the gut microbiota: an unexpected link. Sci Transl Med 2015; 7(271): 271ps1

[159]

Mandar R. Microbiota of male genital tract: impact on the health of man and his partner. Pharmacol Res 2013; 69: 32–41

[160]

Keay S, Zhang CO, Baldwin BR, Alexander RB. Polymerase chain reaction amplification of bacterial 16s rRNA genes in prostate biopsies from men without chronic prostatitis. Urology 1999; 53(3): 487–491

[161]

Krieger JN, Riley DE, Vesella RL, Miner DC, Ross SO, Lange PH. Bacterial DNA sequences in prostate tissue from patients with prostate cancer and chronic prostatitis. J Urol 2000; 164(4): 1221–1228

[162]

Yow MA, Tabrizi SN, Severi G, Bolton DM, Pedersen J; Australian Prostate Cancer BioResource, Giles GG, Southey MC. Characterisation of microbial communities within aggressive prostate cancer tissues. Infect Agent Cancer 2017; 12(1): 4

[163]

Salter SJ, Cox MJ, Turek EM, Calus ST, Cookson WO, Moffatt MF, . Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol 2014; 12: 87

[164]

Glassing A, Dowd SE, Galandiuk S, Davis B, Chiodini RJ. Inherent bacterial DNA contamination of extraction and sequencing reagents may affect interpretation of microbiota in low bacterial biomass samples. Gut Pathog 2016; 8(1): 24

[165]

Alfano M, Canducci F, Nebuloni M, Clementi M, Montorsi F, Salonia A. The interplay of extracellular matrix and microbiome in urothelial bladder cancer. Nat Rev Urol 2016; 13(2): 77–90

[166]

Caini S, Gandini S, Dudas M, Bremer V, Severi E, Gherasim A. Sexually transmitted infections and prostate cancer risk: a systematic review and meta-analysis. Cancer Epidemiol 2014; 38: 329–338

[167]

Yoon BI, Kim S, Han DS, Ha US, Lee SJ, Kim HW, Han CH, Cho YH. Acute bacterial prostatitis: how to prevent and manage chronic infection? J Infect Chemother 2012; 18(4): 444–450

[168]

Fair WR, Parrish RF. Antibacterial substances in prostatic fluid. Prog Clin Biol Res 1981; 75A: 247–264

[169]

Hall SH, Hamil KG, French FS. Host defense proteins of the male reproductive tract. J Androl 2002; 23(5): 585–597

[170]

Alexeyev OA, Marklund I, Shannon B, Golovleva I, Olsson J, Andersson C, Eriksson I, Cohen R, Elgh F. Direct visualization of Propionibacterium acnes in prostate tissue by multicolor fluorescent in situ hybridization assay. J Clin Microbiol 2007; 45(11): 3721–3728

[171]

Drott JB, Alexeyev O, Bergström P, Elgh F, Olsson J. Propionibacterium acnes infection induces upregulation of inflammatory genes and cytokine secretion in prostate epithelial cells. BMC Microbiol 2010; 10(1): 126–132

[172]

Palayoor ST, Youmell MY, Calderwood SK, Coleman CN, Price BD. Constitutive activation of IκB kinase α and NF-κB in prostate cancer cells is inhibited by ibuprofen. Oncogene 1999; 18(51): 7389–7394

[173]

Mora LB, Buettner R, Seigne J, Diaz J, Ahmad N, Garcia R, Bowman T, Falcone R, Fairclough R, Cantor A, Muro-Cacho C, Livingston S, Karras J, Pow-Sang J, Jove R. Constitutive activation of Stat3 in human prostate tumors and cell lines: direct inhibition of Stat3 signaling induces apoptosis of prostate cancer cells. Cancer Res 2002; 62(22): 6659–6666

[174]

Fassi Fehri L, Mak TN, Laube B, Brinkmann V, Ogilvie LA, Mollenkopf H, Lein M, Schmidt T, Meyer TF, Brüggemann H. Prevalence of Propionibacterium acnes in diseased prostates and its inflammatory and transforming activity on prostate epithelial cells. Int J Med Microbiol 2011; 301(1): 69–78

[175]

Ogino S, Nishihara R, VanderWeele TJ, Wang M, Nishi A, Lochhead P, . The role of molecular pathological epidemiology in the study of neoplastic and non-neoplastic diseases in the era of precision medicine. Epidemiology 2016; 27(4): 602–611

[176]

Ogino S, Lochhead P, Chan AT, Nishihara R, Cho E, Wolpin BM, . Molecular pathological epidemiology of epigenetics: emerging integrative science to analyze environment, host, and disease. Mod Pathol 2013; 26(4): 465–484

[177]

Kashyap PC, Chia N, Nelson H, Segal E, Elinav E. Microbiome at the frontier of personalized medicine. Mayo Clin Proc 2017; 92(12): 1855–1864

[178]

Hamada T, Nowak JA, Milner DA Jr. Song M, Ogino S. Integration of microbiology, molecular pathology, and epidemiology: a new paradigm to explore the pathogenesis of microbiome-driven neoplasms. J Pathol 2019; 247(5): 615–628

[179]

Ogino S, Nowak JA, Hamada T, Milner DA Jr, Nishihara R. Insights into pathogenic interactions among environment, host, and tumor at the crossroads of molecular pathology and epidemiology. Annu Rev Pathol 2019; 14: 83–103

[180]

Ogino S, Chan AT, Fuchs CS, Giovannucci E. Molecular pathological epidemiology of colorectal neoplasia: an emerging transdisciplinary and interdisciplinary field. Gut 2011; 60: 397–411

[181]

Hamada T, Keum N, Nishihara R, Ogino S. Molecular pathological epidemiology: new developing frontiers of big data science to study etiologies and pathogenesis. J Gastroenterol 2017; 52: 265–275

[182]

Whitaker NJ, Glenn WK, Sahrudin A, Orde MM, Delprado W, Lawson JS. Human papillomavirus and Epstein Barr virus in prostate cancer: koilocytes indicate potential oncogenic influences of human papillomavirus in prostate cancer. Prostate 2013; 73(3): 236–241

[183]

Nam YD, Kim HJ, Seo JG, Kang SW, Bae JW. Impact of pelvic radiotherapy on gut microbiota of gynecological cancer patients revealed by massive pyrosequencing. PLoS One 2013; 8(12): e82659

[184]

Banerjee S, Alwine JC, Wei Z, Tian T, Shih N, Sperling C, . Microbiome signatures in prostate cancer. Carcinogenesis 2019; 40(6): 749–764

[185]

Zambrano A, Kalantari M, Simoneau A, Jensen JL, Villarreal LP. Detection of human polyomaviruses and papillomaviruses in prostatic tissue reveals the prostate as a habitat for multiple viral infections. Prostate 2002; 53(4): 263–276

[186]

Blaheta RA, Weich E, Marian D, Bereiter-Hahn J, Jones J, Jonas D, Michaelis M, Doerr HW, Cinatl J Jr. Human cytomegalovirus infection alters PC3 prostate carcinoma cell adhesion to endothelial cells and extracellular matrix. Neoplasia 2006; 8(10): 807–816

[187]

Bhatt AP, Redinbo MR, Bultman SJ. The role of the microbiome in cancer development and therapy. CA Cancer J Clin 2017; 67(4): 326–344

[188]

Amirian ES, Petrosino JF, Ajami NJ, Liu Y, Mims MP, Scheurer ME. Potential role of gastrointestinal microbiota composition in prostate cancer risk. Infect Agent Cancer 2013; 8(1): 42

[189]

Haiser HJ, Turnbaugh PJ. Developing a metagenomic view of xenobiotic metabolism. Pharmacol Res 2013; 69(1): 21–31

[190]

Dutton RJ, Turnbaugh PJ. Taking a metagenomic view of human nutrition. Curr Opin Clin Nutr Metab Care 2012; 15(5): 448–454

[191]

La Thangue NB, Kerr DJ. Predictive biomarkers: a paradigm shift towards personalized cancer medicine. Nat Rev Clin Oncol 2011; 8(10): 587–596

[192]

Wong SH, Kwong TNY, Wu CY, Yu J. Clinical applications of gut microbiota in cancer biology. Semin Cancer Biol 2019; 55: 28– 36

[193]

Shetty Y, Prabhu P, Prabhakar B. Emerging vistas in theranostic medicine. Int J Pharm 2019; 558: 29–42

[194]

Behrouzi A, Nafari AH, Siadat SD. The significance of microbiome in personalized medicine. Clin Transl Med 2019; 8(1): 16

[195]

Alanee S, El-Zawahry A, Dynda D, Dabaja A, McVary K, Karr M, . A prospective study to examine the association of the urinary and fecal microbiota with prostate cancer diagnosis after transrectal biopsy of the prostate using 16sRNA gene analysis. Prostate 2019; 79(1): 81–87

[196]

Feng Y, Jaratlerdsiri W, Patrick SM, Lyons RJ, Haynes AM. Collins CC, . Metagenomic analysis reveals a rich bacterial content in high-risk prostate tumors from African men. Prostate 2019; 79(15): 1731–1738

[197]

Feng Y, Ramnarine VR, Bell R, Volik S, Davicioni E. Hayes VM, . Metagenomic and metatranscriptomic analysis of human prostate microbiota from patients with prostate cancer. BMC Genomics 2019; 20(1): 146

[198]

Ma X, Chi C, Fan L, Dong B, Shao X, Xie S, . The microbiome of prostate fluid is associated with prostate cancer. Front Microbiol 2019; 10: 1664

[199]

Rea D, Coppola G, Palma G, Barbieri A, Luciano A, Del Prete P. Microbiota effects on cancer: from risks to therapies. Oncotarget 2018; 9(25): 17915–17927

[200]

Wilson KM, Giovannucci EL, Mucci LA. Lifestyle and dietary factors in the prevention of lethal prostate cancer. Asian J Androl 2012; 14: 365–374

[201]

Punnen S, Hardin J, Cheng I, Klein EA, Witte JS. Impact of meat consumption, preparation, and mutagens on aggressive prostate cancer. PLoS One 2011; 6: e27711

[202]

Newmark HL, Heaney RP. Dairy products and prostate cancer risk. Nutr Cancer 2010; 62: 297–299

[203]

Richman EL, Kenfield SA, Stampfer MJ, Giovannucci EL, Chan JM. Egg, red meat, and poultry intake and risk of lethal prostate cancer in the prostate-specific antigen-era: incidence and survival. Cancer Prev Res (Phila) 2011; 4(12): 2110–2121

[204]

Astorg P. Dietary N-6 and N-3 polyunsaturated fatty acids and prostate cancer risk: a review of epidemiological and experimental evidence. Cancer Causes Control 2004; 15: 367–386

[205]

Joshi AD, Corral R, Catsburg C, Lewinger JP, Koo J, John EM, . Red meat and poultry, cooking practices, genetic susceptibility and risk of prostate cancer: results from a multiethnic case–control study. Carcinogenesis 2012; 33: 2108–2118

[206]

Travis RC, Appleby PN, Siddiq A, Allen NE, Kaaks R, Canzian F, . Genetic variation in the lactase gene, dairy product intake and risk for prostate cancer in the European prospective investigation into cancer and nutrition. Int J Cancer 2013; 132(8): 1901–1910

[207]

Amirian ES, Ittmann MM, Scheurer ME. Associations between arachidonic acid metabolism gene polymorphisms and prostate cancer risk. Prostate 2011; 71(13): 1382–1389

[208]

Massari F, Mollica V, Di Nunno V, Gatto L, Santoni M, Scarpelli M, . The human microbiota and prostate cancer: friend or foe? Cancers (Basel) 2019; 11(4):459

[209]

Kundu P, Blacher E, Elinav E, Pettersson S. Our gut microbiome: the evolving inner self. Cell 2017; 171(7): 1481–1493

[210]

Blaser M. Antibiotic overuse: stop the killing of beneficial bacteria. Nature 2011; 476(7361): 393–394

[211]

Boursi B, Mamtani R, Haynes K, Yang YX. Recurrent antibiotic exposure may promote cancer formation—another step in understanding the role of the human microbiota? Eur J Cancer 2015; 51(17): 2655–2664

[212]

Musso G, Gambino R, Cassader M. Obesity, diabetes, and gut microbiota: the hygiene hypothesis expanded? Diabetes Care 2010; 33(10): 2277–2284

[213]

Iebba V, Nicoletti M, Schippa S. Gut microbiota and the immune system: an intimate partnership in health and disease. Int J Immunopathol Pharmacol 2012; 25(4): 823–833

[214]

Ridlon JM, Ikegawa S, Alves JM, Zhou B, Kobayashi A, Iida T, . Clostridium scindens: a human gut microbe with a high potential to convert glucocorticoids into androgens. J Lipid Res 2013; 54(9): 2437–2449

[215]

Bisanz JE, Enos MK, Mwanga JR, Changalucha J, Burton JP, Gloor GB, . Randomized open-label pilot study of the influence of probiotics and the gut microbiome on toxic metal levels in Tanzanian pregnant women and school children. MBio 2014; 5(5): e01580–1514

[216]

Maleki Vareki S, Chanyi RM, Abdur-Rashid K, Brennan L, Burton JP. Moving on from Metchnikoff: thinking about microbiome therapeutics in cancer. Ecancermedicalscience 2018; 12: 867

[217]

Namasivayam N. Chemoprevention in experimental animals. Ann N Y Acad Sci 2011; 1215: 60–71

[218]

Flores R, Shi J, Fuhrman B, Xu X, Veenstra TD, Gail MH, . Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: a cross-sectional study. J Transl Med 2012; 10: 253

[219]

Cavalieri E, Rogan E. The molecular etiology and prevention of estrogeninitiated cancers. Mol Aspects Med 2014; 36: 1–55

[220]

Akaza H. Prostate cancer chemoprevention by soy isoflavones: role of intestinal bacteria as the “second human genome”. Cancer Sci 2012; 103(6): 969–975

[221]

Tsuji H, Moriyama K, Nomoto K, Miyanaga N, Akaza H. Isolation and characterization of the equol-producing bacterium Slackia sp. Strain NATTS. Arch Microbiol 2010; 192(4): 279–287

[222]

El-Demiry MI, Hargreave TB, Busuttil A, James K, Ritchie AW, Chisholm GD. Lymphocyte sub-populations in the male genital tract. Br J Urol 1985; 57(6): 769–774

[223]

McClinton S, Miller ID, Eremin O. An immunohistochemical characterisation of the inflammatory cell infiltrate in benign and malignant prostatic disease. Br J Cancer 1990; 61(3): 400–403

[224]

Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, Dai RM, Kiu H, Cardone M, Naik S, Patri AK, Wang E, Marincola FM, Frank KM, Belkaid Y, Trinchieri G, Goldszmid RS. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013; 342(6161): 967–970

[225]

Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, . The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 2013; 342: 971–976

[226]

Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Lei YM, Jabri B, Alegre ML, Chang EB, Gajewski TF. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015; 350(6264): 1084–1089

[227]

Johnstone TC, Park GY, Lippard SJ. Understanding and improving platinum anticancer drugs—phenanthriplatin. Anticancer Res 2014; 34(1): 471–476

[228]

Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, Fidelle M, Flament C, Poirier-Colame V, Opolon P, Klein C, Iribarren K, Mondragón L, Jacquelot N, Qu B, Ferrere G, Clémenson C, Mezquita L, Masip JR, Naltet C, Brosseau S, Kaderbhai C, Richard C, Rizvi H, Levenez F, Galleron N, Quinquis B, Pons N, Ryffel B, Minard-Colin V, Gonin P, Soria JC, Deutsch E, Loriot Y, Ghiringhelli F, Zalcman G, Goldwasser F, Escudier B, Hellmann MD, Eggermont A, Raoult D, Albiges L, Kroemer G, Zitvogel L. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018; 359(6371): 91–97

[229]

Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, Prieto PA, Vicente D, Hoffman K, Wei SC, Cogdill AP, Zhao L, Hudgens CW, Hutchinson DS, Manzo T, Petaccia de Macedo M, Cotechini T, Kumar T, Chen WS, Reddy SM, Szczepaniak Sloane R, Galloway-Pena J, Jiang H, Chen PL, Shpall EJ, Rezvani K, Alousi AM, Chemaly RF, Shelburne S, Vence LM, Okhuysen PC, Jensen VB, Swennes AG, McAllister F, Marcelo Riquelme Sanchez E, Zhang Y, Le Chatelier E, Zitvogel L, Pons N, Austin-Breneman JL, Haydu LE, Burton EM, Gardner JM, Sirmans E, Hu J, Lazar AJ, Tsujikawa T, Diab A, Tawbi H, Glitza IC, Hwu WJ, Patel SP, Woodman SE, Amaria RN, Davies MA, Gershenwald JE, Hwu P, Lee JE, Zhang J, Coussens LM, Cooper ZA, Futreal PA, Daniel CR, Ajami NJ, Petrosino JF, Tetzlaff MT, Sharma P, Allison JP, Jenq RR, Wargo JA. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018; 359(6371): 97–103

[230]

Parry RV, Chemnitz JM, Frauwirth KA, Lanfranco AR, Braunstein I, Kobayashi SV, Linsley PS, Thompson CB, Riley JL. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol 2005; 25(21): 9543–9553

[231]

Fife BT, Pauken KE, Eagar TN, Obu T, Wu J, Tang Q, Azuma M, . Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal. Nat Immunol 2009; 10: 1185–1192

[232]

Graff JN, Alumkal JJ, Drake CG, Thomas GV, Redmond WL, Farhad M, Cetnar JP, Ey FS, Bergan RC, Slottke R, Beer TM. Early evidence of anti-PD-1 activity in enzalutamide-resistant prostate cancer. Oncotarget 2016; 7(33): 52810–52817

[233]

Kuczma MP, Ding ZC, Li T, Habtetsion T, Chen T, Hao Z, . The impact of antibiotic usage on the efficacy of chemoimmunotherapy is contingent on the source of tumor-reactive T cells. Oncotarget 2017; 8(67): 111931–111942

[234]

Vande Voorde J, Sabuncuoğlu S, Noppen S, Hofer A, Ranjbarian F, Fieuws S, Balzarini J, Liekens S. Nucleoside-catabolizing enzymes in mycoplasma-infected tumor cell cultures compromise the cytostatic activity of the anticancer drug gemcitabine. J Biol Chem 2014; 289(19): 13054–13065

[235]

Ladoire S, Eymard JC, Zanetta S, Mignot G, Martin E, Kermarrec I, Mourey E, Michel F, Cormier L, Ghiringhelli F. Metronomic oral cyclophosphamide prednisolone chemotherapy is an effective treatment for metastatic hormone-refractory prostate cancer after docetaxel failure. Anticancer Res 2010; 30(10): 4317–4323

[236]

Kerbel RS, Kamen BA. The anti-angiogenic basis of metronomic chemotherapy. Nat Rev Cancer 2004; 4(6): 423–436

[237]

Ghiringhelli F, Menard C, Puig PE, Ladoire S, Roux S, Martin F, . Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T-cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol Immunother 2007; 56(5): 641–648

[238]

Nicolini A, Mancini P, Ferrari P, Anselmi L, Tartarelli G, Bonazzi V, . Oral low-dose cyclophosphamide in metastatic hormone refractory prostate cancer (MHRPC). Biomed Pharmacother 2004; 58(8): 447–450

[239]

Hellerstedt B, Pienta KJ, Redman BG, Esper P, Dunn R, Fardig J, . Phase II trial of oral cyclophosphamide, prednisone, and diethylstilbestrol for androgen-independent prostate carcinoma. Cancer 2003; 98(8): 1603–1610

[240]

Bracarda S, Tonato M, Rosi P, De Angelis V, Mearini E, Cesaroni S, Fornetti P, Porena M. Oral estramustine and cyclophosphamide in patients with metastatic hormone refractory prostate carcinoma: a phase II study. Cancer 2000; 88(6): 1438–1444

[241]

Johnstone TC, Suntharalingam K, Lippard SJ. The next generation of platinum drugs: targeted Pt(II) agents, nanoparticle delivery, and Pt(IV) prodrugs. Chem Rev 2016; 116(5): 3436–3486

[242]

Viaud S, Daillère R, Boneca IG, Lepage P, Langella P, Chamaillard M, Pittet MJ, Ghiringhelli F, Trinchieri G, Goldszmid R, Zitvogel L. Gut microbiome and anticancer immune response: really hot Sh*t! Cell Death Differ 2015; 22(2): 199–214

[243]

Westman EL, Canova MJ, Radhi IJ, Koteva K, Kireeva I, Waglechner N, . Bacterial inactivation of the anticancer drug doxorubicin. Chem Biol 2012; 19(10): 1255–1264

[244]

Harada N, Hanaoka R, Horiuchi H, Kitakaze T, Mitani T, Inui H, Yamaji R. Castration influences intestinal microflora and induces abdominal obesity in high-fat diet-fed mice. Sci Rep 2016; 6(1): 23001

[245]

Fijlstra M, Ferdous M, Koning AM, Rings EH, Harmsen HJ, Tissing WJ. Substantial decreases in the number and diversity of microbiota during chemotherapy-induced gastrointestinal mucositis in a rat model. Support Care Cancer 2015; 23(6): 1513– 1522

[246]

Sfanos KS, Markowski MC, Peiffer LB, Ernst SE, White JR, Pienta KJ, . Compositional differences in gastrointestinal microbiota in prostate cancer patients treated with androgen axis-targeted therapies. Prostate Cancer Prostatic Dis 2018; 21(4): 539–548

[247]

Bode HB, Zeggel B, Silakowski B, Wenzel SC, Reichenbach H, Müller R. Steroid biosynthesis in prokaryotes: identification of myxobacterial steroids and cloning of the first bacterial 2,3(S)-oxidosqualene cyclase from the myxobacterium Stigmatella aurantiaca. Mol Microbiol 2003; 47: 471–481

[248]

Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, Luke JJ, Gajewski TF. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018; 359(6371): 104–108

[249]

Chitapanarux I, Chitapanarux T, Traisathit P, Kudumpee S, Tharavichitkul E, Lorvidhaya V. Randomized controlled trial of live Lactobacillus acidophilus plus Bifidobacterium bifidum in prophylaxis of diarrhea during radiotherapy in cervical cancer patients. Radiat Oncol 2010; 5(1): 31

[250]

Seidel DV, Azcárate-Peril MA, Chapkin RS, Turner ND. Shaping functional gut microbiota using dietary bioactives to reduce colon cancer risk. Semin Cancer Biol 2017; 46: 191–204

[251]

Conlon MA, Bird AR. The impact of diet and lifestyle on gut microbiota and human health. Nutrients 2014; 7(1): 17–44

[252]

Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CP, Poirier-Colame V, Roux A, Becharef S, Formenti S, Golden E, Cording S, Eberl G, Schlitzer A, Ginhoux F, Mani S, Yamazaki T, Jacquelot N, Enot DP, Bérard M, Nigou J, Opolon P, Eggermont A, Woerther PL, Chachaty E, Chaput N, Robert C, Mateus C, Kroemer G, Raoult D, Boneca IG, Carbonnel F, Chamaillard M, Zitvogel L. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015; 350(6264): 1079–1084

[253]

Guarner F, Malagelada JR. Gut flora in health and disease. Lancet 2003; 361: 512–519

[254]

Horinaka M. Yoshida T, Kishi A, Akatani K, Yasuda T, Kouhara J, . Lactobacillus strains induce TRAIL production and facilitate natural killer activity against cancer cells. FEBS Lett 2010; 584(3): 577–582

[255]

Sanders ME, Klaenhammer TR. The scientific basis of Lactobacillus acidophilus NCFM functionality as a probiotic. J Dairy Sci 2001; 84: 319–331

[256]

Montes RG, Bayless TM, Saavedra JM, Perman JA. Effect of milks inoculated with Lactobacillus acidophilus or a yogurt starter culture in lactosemaldigesting children. J Dairy Sci 1995; 78: 1657–1664

[257]

Davani-Davari D, Negahdaripour M, Karimzadeh I, Seifan M, Mohkam M, Masoumi SJ, . Prebiotics: definition, types, sources, mechanisms, and clinical applications. Foods 2019; 8(3): 92

[258]

Wang H, Geier MS, Howarth GS. Prebiotics: a potential treatment strategy for the chemotherapy-damaged gut? Crit Rev Food Sci Nutr 2016; 56(6): 946–956

[259]

Sambi M, Bagheri L, Szewczuk MR. Current challenges in cancer immunotherapy: multimodal approaches to improve efficacy and patient response rates. J Oncol 2019; 2019: 4508794

[260]

Arora M, Weuve J, Fall K, Pedersen NL, Mucci LA. An exploration of shared genetic risk factors between periodontal disease and cancers: a prospective co-twin study. Am J Epidemiol 2010; 171(2): 253–259

[261]

Miyake M, Ohnishi K, Hori S, Nakano A, Nakano R, Yano H, . Mycoplasma genitalium infection and chronic inflammation in human prostate cancer: detection using prostatectomy and needle biopsy specimens. Cells 2019; 8(3): 212

[262]

Al-Marhoon MS, Ouhtit A, Al-Abri AO, Venkiteswaran KP, Al-Busaidi Q, Mathew J, . Molecular evidence of Helicobacter pylori infection in prostate tumors. Curr Urol 2015; 8(3): 138–143

[263]

Alanee S, El-Zawahry A, Dynda D, McVary K, Karr M, Braundmeier-Fleming A. Prospective examination of the changes in the urinary microbiome induced by transrectal biopsy of the prostate using 16S rRNA gene analy sis. Prostate Cancer Prostatic Dis 2019; 22(3): 446–452

RIGHTS & PERMISSIONS

Higher Education Press

AI Summary AI Mindmap
PDF (790KB)

5481

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/