Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2437 2022-08-30 16:55:34 |
2 layout + 6 word(s) 2443 2022-09-01 03:07:26 | |
3 layout Meta information modification 2443 2022-09-01 03:08:57 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Kumar, N.B.;  Hogue, S.;  Pow-Sang, J.;  Poch, M.;  Manley, B.J.;  Li, R.;  Dhillon, J.;  Yu, A.;  Byrd, D.A. Green Tea Catechins in Chemoprevention for Prostate Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/26684 (accessed on 28 March 2024).
Kumar NB,  Hogue S,  Pow-Sang J,  Poch M,  Manley BJ,  Li R, et al. Green Tea Catechins in Chemoprevention for Prostate Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/26684. Accessed March 28, 2024.
Kumar, Nagi B., Stephanie Hogue, Julio Pow-Sang, Michael Poch, Brandon J. Manley, Roger Li, Jasreman Dhillon, Alice Yu, Doratha A. Byrd. "Green Tea Catechins in Chemoprevention for Prostate Cancer" Encyclopedia, https://encyclopedia.pub/entry/26684 (accessed March 28, 2024).
Kumar, N.B.,  Hogue, S.,  Pow-Sang, J.,  Poch, M.,  Manley, B.J.,  Li, R.,  Dhillon, J.,  Yu, A., & Byrd, D.A. (2022, August 30). Green Tea Catechins in Chemoprevention for Prostate Cancer. In Encyclopedia. https://encyclopedia.pub/entry/26684
Kumar, Nagi B., et al. "Green Tea Catechins in Chemoprevention for Prostate Cancer." Encyclopedia. Web. 30 August, 2022.
Green Tea Catechins in Chemoprevention for Prostate Cancer
Edit

Human PCa is a complex heterogeneous disease. The central driving forces of prostate carcinogenesis include acquisitions of diverse sets of hallmark capabilities, aberrant functioning of androgen receptor signaling, deregulation of vital cell physiological processes, inactivation of tumor-suppressive activity, and disruption of prostate gland-specific cellular homeostasis. Green tea is known for its health benefits deriving from molecules called green tea catechins (GTCs). GTCs have been demonstrated to influence molecular pathways to halt the progression of prostate cancer (PCa) and may be of particular benefit to men with low-risk PCa who are placed on active surveillance. Administering GTCs may provide patients an opportunity to be actively engaged in their treatment and help prevent cancer progression. Importantly, the trillions of microbes in the gut (the gut microbiome) metabolize GTCs, making them more accessible to the body to exert their health effects. Additionally, the gut microbiome influences multiple other processes likely involved in PCa progression, including regulating inflammation, hormones, and other known/unknown pathways. 

prostate cancer green tea catechins microbiome

1. Introduction

The American Cancer Society estimates that there will be 268,490 and 34,500 new cases of and deaths due to prostate cancer (PCa) in the United States (US) in 2022, respectively [1]. Over the past two decades, PCa screening via serum prostate-specific antigen (PSA) led to substantial increases in detection of low-risk PCas (Gleason score ≤ 6), which pose little risk of either metastatic spread or death [2][3][4][5]. Conversely, over-treatment is a well-documented consequence of over-detection of PCa, predominantly occurring among men with low-risk PCa who may be subject to multiple treatment-related morbidities with negligible or no benefit towards cancer-specific survival [4][6]. Thus, the recommended guideline for the management of low-risk disease is active surveillance (AS). However, there are several identified challenges with AS, ranging from concerns with under-grading [7][8][9][10][11][12][13], patient-related factors (e.g., anxiety, depression, doubts about the possible progression of disease), and higher decisional conflict regarding the selection of AS [14][15][16], leading many to ultimately opt for a treatment that does not beneficially change tumor characteristics. On the other hand, men on AS are a highly motivated subgroup eager to make positive lifestyle changes to reduce their risk of PCa progression [16][17][18][19][20][21], providing an optimal opportunity to intervene during this window with promising chemopreventive agents for PCa.
Previous strategies for PCa chemoprevention included 5-alpha-reductase inhibitors, finasteride, dutasteride [22][23][24], trace element selenomethionine, and/or vitamin E. Collectively, these agents demonstrated greater risk for high grade disease [25] or no reduction in risk of PCa progression in large phase III trials, severely limiting their clinical adoption [23]. To date, there is minimal evidence available for the efficacy of any one agent or strategy for chemoprevention of PCa among men on AS. Therefore, the goal of our team for PCa chemoprevention is to utilize a systematic, broad-spectrum approach [26] that involves an agent shown to (a) be bioavailable; (b) have an excellent safety profile; (c) produce robust targeting of multiple relevant molecular pathways; and (d) modulate measurable intermediate endpoint biomarkers correlated with early clinical progression of PCa—an approach that collectively may be more effective than agents evaluated to date. Our team and others have evaluated several approaches (i.e., diet interventions) and agents (selenium, vitamin E, isoflavones, lycopene n-3 fatty acids, and green tea catechins, or GTCs) targeting prostate carcinogenesis.
Human PCa is a complex heterogeneous disease. The central driving forces of prostate carcinogenesis include acquisitions of diverse sets of hallmark capabilities, aberrant functioning of androgen receptor signaling, deregulation of vital cell physiological processes, inactivation of tumor-suppressive activity, and disruption of prostate gland-specific cellular homeostasis. Thus, the molecular complexity and redundancy of oncoprotein signaling in PCa demands for concurrent inhibition of multiple hallmark-associated pathways [27]. The ultimate goal for clinical cancer chemoprevention is to utilize a systematic, broad-spectrum approach that involves identifying and evaluating agents that can: (a) produce robust and concurrent inhibition of multiple hallmark-associated pathways in the target tissue/microenvironment; (b) address the underlying biology of carcinogenesis; and (c) enhance bioavailability and half-life with minimal toxicity in exceptionally high-risk populations [26][28]. GTCs comprise (−)-epigallocatechin-3-gallate (EGCG), (−)-epicatechin, (−)-epigallocatechin (EGC), and (−)-epicatechin-3-gallate. Among the agents evaluated to date, EGCG in particular has been demonstrated to affect molecular pathways implicated in prostate carcinogenesis.

2. Green Tea Catechins: Promising Agent for Prostate Cancer Chemoprevention

The most abundant constituents of green tea are the polyphenols, which are catechins that represent 30–40% of the dry weight of the tea leaves. The catechins in green tea belong to the flavon-3-ols of the polyphenol family [29]. Laboratory studies have identified EGCG as the most potent modulator of molecular pathways thought to be relevant to prostate carcinogenesis [30][31][32][33]. In the past two decades, research studies have shown that GTCs influence multiple biochemical and molecular cascades that inhibit several hallmarks of carcinogenesis relevant to prostate carcinogenesis. With an acceptable safety profile, GTCs are ideal candidates for PCa chemoprevention. Laboratory studies demonstrate that EGCG can affect several cancer-related proteins, including p27, Bcl-2 or Bcr-Abl oncoproteins, Bax, matrix metalloproteinases (MMP-2 and MMP-9), the androgen receptor (particularly important in PCa development and progression), epidermal growth factor receptor, activator protein 1, and some cell cycle regulators [30][34]. Using cell culture systems, Adhami et al. [35] were able to show that EGCG induces apoptosis, cyclin kinase inhibitor WAF-1/p21-mediated cell cycle-dysregulation, and cell growth inhibition. In cDNA microarrays, EGCG treatment of LNCaP cells induced genes that exhibit growth-inhibitory effects and repressed genes belonging to the G-protein signaling network [36]. The ubiquitin/proteasome pathway plays a critical role in activation of the cellular apoptotic program and the regulation of apoptosis [37]. Our work demonstrated that GTC specifically inhibits the chymotrypsin-like activity of the proteasome in several tumor and transformed cell lines, including prostate cell lines, resulting in the accumulation of two natural proteasome substrates–p27 (Kip1) and nuclear factor kappa B (NF-ĸB) inhibitor alpha, which inhibit transcription factor NF-ĸB, leading to growth arrest in the G(1) phase of the cell cycle. Synthetic analogs of EGCG were observed to be more potent as proteasome inhibitors compared to EGCG. Polyphenon E® (Poly E) and Sunphenon® 90D are standardized formulations of green tea containing 50% of the catechins from EGCG. Searchers observed that Poly E® (>50% EGCG, 80% total catechins) preferentially inhibits the proteasomal chymotrypsin-like activities over other activities, with an IC50 value of 0.88 µM [37][38][39][40]. Standardized GTC formulations of Poly E® and Sunphenon® 90D in equal concentrations were evaluated in vitro. Pre-treatment with Sunphenon® 90D downregulated NF-ĸB in H2O2-treated C2C12 cells, while activating caspase-3 (Figure 1) [41]. Incubation of human primary osteoblasts with Sunphenon® 90D significantly reduced oxidative stress and improved cell viability [42]. EGCG has been shown to have both anti-inflammatory properties, such as through the influence of T-cell proliferation and inhibition of NF-ĸB, and neuroprotective properties by acting as a free radical scavenger [43][44]. More specifically, EGCG’s antioxidant properties deplete reactive oxygen species, thus preventing DNA damage and inhibiting NF-ĸB-induced inflammation, angiogenesis, and cell survival that could otherwise propel cancer development and progression [45]. In summary, researchers and others have reported convincing evidence suggesting that GTCs inhibit proliferation and cell cycle events and induce apoptosis through multiple mechanisms.
Figure 1. Mechanistic pathway by which GTCs prevent PCa progression. In vitro studies [37][38][39][40] demonstrate that GTCs block proteasomal activity in PCa cells, leading to build-up of proteasomal substrates Kip1 and Ik-b α that subsequently downregulate the activity of NF-κB. This inhibits the cell cycle and elicits apoptosis in these PCa cells. GTCs, green tea catechins; Ik-b α, NF-κB inhibitor alpha; NF-κB, nuclear factor kappa B; PCa, prostate cancer. Created with Biorender.com (accessed on 1 July 2022).
The association of green tea intake with PCa risk has been investigated in several epidemiological studies. In a meta-analysis of 9 case-control studies, there was a statistically significant 57% lower risk of PCa, comparing subjects with the highest relative to lowest green tea consumption, whereas there was a null association in a meta-analysis of 4 cohort studies [46]. Similar results were observed in a more recent meta-analysis of 3 case-control and 4 cohort studies: no statistically significant associations were observed across cohort studies, while a statistically significant 55% lower odds of PCa was observed for highest versus lowest green tea intake in the case-control studies [47]. This inconsistency could be due, in part, to differences in study design, residual confounding factors such as by diet/lifestyle and biological factors, and varying formulations and subtypes of green tea studied. These studies were mostly limited to men in Asian countries, where approximately 20% of green tea is consumed globally and where mortality from PCa is the lowest compared to Western populations [31], where green tea consumption is a more recent phenomenon. Asian men who migrate to the US have a relatively increased risk of PCa compared to their counterparts in their countries of origin, potentially as a result of acculturation and adoption of Western diets [33]. Although the above study findings have been mixed—potentially due to confounding by variation in geographical location, tobacco and alcohol use, and other lifestyle factors (mainly diets) [32][48]—taken together, studies among Asian populations demonstrate a protective effect of GTCs as related to PCa [32][33][49]. Another highly plausible confounder of GTC-PCa associations is the gut microbiome, which has increasingly been implicated in the modulation of carcinogenesis. The gut microbiome comprises densely populated commensal and symbiotic microbes [50] whose composition is highly influenced by the host’s dietary intake. The gut microbiome also produces metabolically active metabolites that interact with host-signaling pathways and gene expression, impacting cancer initiation and progression [51][52]. Multiple studies have observed differences in the gut microbiome between various racial and ethnic groups, even amongst those living in the same community. These differences are potentially attributed to lifestyle, dietary, social, and other uncharacterized exposures that result in variations across racial and ethnic groups [53][54]. Using fecal shotgun metagenomic data analyzed amongst 106 Japanese individuals compared with those of 11 other nations, the composition of the Japanese gut microbiome was more abundant in the phylum Actinobacteria, in particular, genus Bifidobacterium, compared to others [55]. In line with increased PCa rates in Asian populations living in the US, studies have shown that the gut microbiome of Southeast Asian immigrants changes after migration to the US [56], potentially indicative of an incompatibility between the incorporation of Western lifestyles with the traditionally harbored microbiome of this population [57]. These studies have provided the basis for understanding that the gut microbiome can act as an important mediating factor in investigations of diet and lifestyle differences that potentially promote cancer risk.

3. The Gut Microbiome, Prostate Cancer, and Green Tea Catechins

Predictive biomarkers of responses to secondary chemoprevention are presently lacking. Identification of biomarkers, such as the gut microbiome, predictive of favorable clinical responses to secondary chemoprevention has the potential to substantially facilitate clinical decision-making. Numerous studies found that the gut microbiome directly effects drug metabolism, efficacy, and toxicity, potentially affecting disease development and progression [58]. For example, in oncology, there exists convincing evidence to support that the antitumor effects of immunotherapies can be enhanced or inhibited by the gut microbiome [59][60].
The gut microbiome likely has critical roles in regulating the bioavailability of GTCs and absorption of bioactive phenolic GTC metabolites, as demonstrated in laboratory and pre-clinical models (Figure 2). Although dietary polyphenols are absorbed by the small intestine, accumulating evidence suggests that they are metabolized to a greater extent in the colon by bacterial enzymes [61][62]. EGCG is hydrolyzed by bacteria to gallic acid or EGC and further converted to multiple metabolites, such as 5-(3,5-dihydroxyphenyl)-4-hydroxyvaleric acid and 5-(3′,5′-dihydroxyphenyl)-g-valerolactone [62]. These metabolites are then either taken up via the portal vein and transported to the liver or excreted in the feces.
Figure 2. Examples of chemopreventive effects of GTCs in the context of PCa via gut microbiome modulation. (A) GTCs like EGCG have been evidenced to alter microbial composition, such as increasing abundance of Bifidobacterium [62]. This genus, for example, is known to increase production of SCFAs [62][63] which inhibit inflammatory pathways initiated by NF-κB that would otherwise propel carcinogenesis [64]. (B) The gut microbiome can enzymatically alter GTCs like EGCG to produce metabolites including gallic acid, EGC, valeric acid, and valerolactone, that subsequently travel to the bloodstream to exert potential chemopreventive benefits (e.g., regulating HDAC 1 and 2 and suppressing cell-cycle-related genes) [62][65][66][67]. EGC, epigallocatechin; EGCG, epigallocatechin gallate; GTCs, green tea catechins; HDAC, histone deacetylase; IL-6, interleukin-6; NF-κB, nuclear factor kappa B; SCFA, short chain fatty acid; TNF-α, tumor necrosis factor alpha. Created with Biorender.com (accessed on 1 July 2022).
On the other hand, several pharmacologic agents, including GTCs, were shown to influence gut microbiome composition and function. For example, in a study of 10 volunteers who drank 1000 mL of green tea daily for 10 days, Bifidobacteria abundance was increased [68]. In multiple animal studies, green tea polyphenols had similar effects on Bifidobacteria and other effects, including decreasing the Firmicutes/Bacteroidetes ratio [69][70]. In turn, gut microbiome composition and function may directly and indirectly influence PCa progression, such as through production of metabolically active metabolites or regulation of hormones and inflammation, as described below [71][72][73]. Given the substantial preliminary evidence for interrelationships among GTCs, the gut microbiome, and prostate carcinogenesis, it is highly likely that the gut microbiome may mediate etiological effects of GTCs, including effects on PCa progression and development of adverse events; however, little is known regarding these interrelationships among humans.
The gut microbiome has biologically plausible roles in PCa such as via its influence on hormone and inflammation regulation and production of metabolically active metabolites [71][72][73]. For example, gut microbes produce sex hormones, such as androgen, and, in a study by Pernigoni et al., multiple species among mice and humans produced androgens from androgen precursors, in turn promoting progression of castrate resistant PCa [74]. In a study of both mice and PCa patients, Proteobacteria was increased after antibiotic exposure, and was in turn associated with development of PCa in mice and with metastasis of PCa among humans [75]. In a study of mice on a high-fat diet, the resultant alterations to the mice fecal microbiome promoted histamine biosynthesis and increased inflammatory cancer cell growth [76]. Previous human studies of the gut microbiome and PCa included a case-control study comparing 16S rRNA sequenced fecal bacteria among 64 men with PCa and 41 without PCa, finding differences in beta diversity, higher abundances of Bacteroides and Streptococcus species, and differences in folate and arginine pathways [77]. Another case-control study compared the gut metagenome among 8 men with benign prostatic conditions and 12 men with intermediate or high risk clinically localized PCa, finding higher relative abundance of Bacteriodes massiliensis and lower relative abundances of Faecalibacterium prausnitzii and Eubacterium rectalie amongst men with intermediate/high-risk PCa [78]. In a comparison of men with and without prostate enlargement, the ratio of Firmicutes to Bacteroidetes was higher among men with enlarged prostates, potentially related to prostate inflammation [79]. Finally, evidence supports the study of the gut microbiome across the disease continuum of PCa, with evidence demonstrating that the gut microbiome may be modified by PCa treatment, including androgen deprivation therapy, among more advanced PCa patients [80].

References

  1. American Cancer Society. Available online: http://www.cancer.org/Cancer/ProstateCancer/DetailedGuide/prostate-cancer-key-statistics (accessed on 1 July 2022).
  2. Bruinsma, S.M.; Bangma, C.H.; Carroll, P.R.; Leapman, M.S.; Rannikko, A.; Petrides, N.; Weerakoon, M.; Bokhorst, L.P.; Roobol, M.J.; Movember, G.A.P.C. Active surveillance for prostate cancer: A narrative review of clinical guidelines. Nat. Rev. Urol. 2016, 13, 151–167.
  3. Ip, S.; Dahabreh, I.J.; Chung, M.; Yu, W.W.; Balk, E.M.; Iovin, R.C.; Mathew, P.; Luongo, T.; Dvorak, T.; Lau, J. An evidence review of active surveillance in men with localized prostate cancer. Evid. Rep. Technol. Assess. 2011, 204, 1–341.
  4. Klotz, L. Active Surveillance for Prostate Cancer: For Whom? J. Clin. Oncol. 2005, 23, 8165–8169.
  5. Thompson, I.; Thrasher, J.B.; Aus, G.; Burnett, A.L.; Canby-Hagino, E.D.; Cookson, M.S.; D’Amico, A.V.; Dmochowski, R.R.; Eton, D.T.; Forman, J.D.; et al. Guideline for the management of clinically localized prostate cancer: 2007 update. J. Urol. 2007, 177, 2106–2131.
  6. Hamdy, F.C.; Donovan, J.L.; Neal, D.E. 10-Year Outcomes in Localized Prostate Cancer. N. Engl. J. Med. 2017, 376, 180.
  7. Cooperberg, M.R.; Carroll, P.R. Trends in Management for Patients With Localized Prostate Cancer, 1990–2013. JAMA 2015, 314, 80–82.
  8. D’Amico, A.V. Personalizing the Use of Active Surveillance As an Initial Approach for Men With Newly Diagnosed Prostate Cancer. J. Clin. Oncol. 2015, 33, 3365–3366.
  9. Klotz, L. Active surveillance for low-risk prostate cancer. Curr. Urol. Rep. 2015, 16, 24.
  10. Klotz, L. Active surveillance and focal therapy for low-intermediate risk prostate cancer. Transl. Androl. Urol. 2015, 4, 342–354.
  11. Klotz, L.; Zhang, L.; Lam, A.; Nam, R.; Mamedov, A.; Loblaw, A. Clinical results of long-term follow-up of a large, active surveillance cohort with localized prostate cancer. J. Clin. Oncol. 2010, 28, 126–131.
  12. Maurice, M.J.; Abouassaly, R.; Kim, S.P.; Zhu, H. Contemporary Nationwide Patterns of Active Surveillance Use for Prostate Cancer. JAMA Intern. Med. 2015, 175, 1569–1571.
  13. Oon, S.F.; Watson, R.W.; O’Leary, J.J.; Fitzpatrick, J.M. Epstein criteria for insignificant prostate cancer. BJU Int. 2011, 108, 518–525.
  14. NCCN. NCCN Guidelines for Prostate Cancer; NCCN: Bethesda, MD, USA, 2017.
  15. Orom, H.; Underwood, W., 3rd; Biddle, C. Emotional Distress Increases the Likelihood of Undergoing Surgery among Men with Localized Prostate Cancer. J. Urol. 2017, 197, 350–355.
  16. Watts, S.; Leydon, G.; Eyles, C.; Moore, C.M.; Richardson, A.; Birch, B.; Prescott, P.; Powell, C.; Lewith, G. A quantitative analysis of the prevalence of clinical depression and anxiety in patients with prostate cancer undergoing active surveillance. BMJ Open 2015, 5, e006674.
  17. Avery, K.N.; Donovan, J.L.; Horwood, J.; Neal, D.E.; Hamdy, F.C.; Parker, C.; Wade, J.; Lane, A. The importance of dietary change for men diagnosed with and at risk of prostate cancer: A multi-centre interview study with men, their partners and health professionals. BMC Fam. Pr. 2014, 15, 81.
  18. Horwood, J.P.; Avery, K.N.; Metcalfe, C.; Donovan, J.L.; Hamdy, F.C.; Neal, D.E.; Lane, J.A. Men’s knowledge and attitudes towards dietary prevention of a prostate cancer diagnosis: A qualitative study. BMC Cancer 2014, 14, 812.
  19. Kelloff, G.J.; Lieberman, R.; Steele, V.E.; Boone, C.W.; Lubet, R.A.; Kopelovitch, L.; Malone, W.A.; Crowell, J.A.; Sigman, C.C. Chemoprevention of prostate cancer: Concepts and strategies. Eur. Urol. 1999, 35, 342–350.
  20. Kumar, N.; Chornokur, G. Molecular Targeted Therapies Using Botanicals for Prostate Cancer Chemoprevention. Transl. Med. 2012, S2, 005.
  21. Lieberman, R. Prostate cancer chemoprevention: Strategies for designing efficient clinical trials. Urology 2001, 57, 224–229.
  22. Andriole, G.L.; Bostwick, D.G.; Brawley, O.W.; Gomella, L.G.; Marberger, M.; Montorsi, F.; Pettaway, C.A.; Tammela, T.L.; Teloken, C.; Tindall, D.J.; et al. Effect of dutasteride on the risk of prostate cancer. N. Engl. J. Med. 2010, 362, 1192–1202.
  23. Hamilton, R.J.; Kahwati, L.C.; Kinsinger, L.S. Knowledge and use of finasteride for the prevention of prostate cancer. Cancer Epidemiol. Biomark. Prev. 2010, 19, 2164–2171.
  24. Thompson, I.M.; Goodman, P.J.; Tangen, C.M.; Lucia, M.S.; Miller, G.J.; Ford, L.G.; Lieber, M.M.; Cespedes, R.D.; Atkins, J.N.; Lippman, S.M.; et al. The influence of finasteride on the development of prostate cancer. N. Engl. J. Med. 2003, 349, 215–224.
  25. Lippman, S.M.; Klein, E.A.; Goodman, P.J.; Lucia, M.S.; Thompson, I.M.; Ford, L.G.; Parnes, H.L.; Minasian, L.M.; Gaziano, J.M.; Hartline, J.A.; et al. Effect of selenium and vitamin E on risk of prostate cancer and other cancers: The Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA 2009, 301, 39–51.
  26. Block, K.I.; Gyllenhaal, C.; Lowe, L.; Amedei, A.; Amin, A.; Amin, A.; Aquilano, K.; Arbiser, J.; Arreola, A.; Arzumanyan, A.; et al. Designing a broad-spectrum integrative approach for cancer prevention and treatment. Semin. Cancer Biol. 2015, 35, S276–S304.
  27. Datta, D.; Aftabuddin, M.; Gupta, D.K.; Raha, S.; Sen, P. Human Prostate Cancer Hallmarks Map. Sci. Rep. 2016, 6, 30691.
  28. Kumar, N.B.; Pow-Sang, J.; Spiess, P.; Dickinson, S.; Schell, M.J. A phase II randomized clinical trial using aglycone isoflavones to treat patients with localized prostate cancer in the pre-surgical period prior to radical prostatectomy. Oncotarget 2020, 11, 1218–1234.
  29. Pérez-Burillo, S.; Navajas-Porras, B.; López-Maldonado, A.; Hinojosa-Nogueira, D.; Pastoriza, S.; Rufián-Henares, J. Green Tea and Its Relation to Human Gut Microbiome. Molecules 2021, 26, 3907.
  30. Connors, S.K.; Chornokur, G.; Kumar, N.B. New insights into the mechanisms of green tea catechins in the chemoprevention of prostate cancer. Nutr. Cancer 2012, 64, 4–22.
  31. Ito, K. Prostate cancer in Asian men. Nat. Rev. Urol. 2014, 11, 197–212.
  32. Jian, L.; Xie, L.P.; Lee, A.H.; Binns, C.W. Protective effect of green tea against prostate cancer: A case-control study in southeast China. Int. J. Cancer 2004, 108, 130–135.
  33. Yuan, J.M. Cancer prevention by green tea: Evidence from epidemiologic studies. Am. J. Clin. Nutr. 2013, 98, 1676S–1681S.
  34. Khan, N.; Mukhtar, H. Modulation of signaling pathways in prostate cancer by green tea polyphenols. Biochem. Pharm. 2013, 85, 667–672.
  35. Adhami, V.M.; Siddiqui, I.A.; Sarfaraz, S.; Khwaja, S.I.; Hafeez, B.B.; Ahmad, N.; Mukhtar, H. Effective Prostate Cancer Chemopreventive Intervention with Green Tea Polyphenols in the TRAMP Model Depends on the Stage of the Disease. Clin. Cancer Res. 2009, 15, 1947–1953.
  36. Adhami, V.M.; Ahmad, N.; Mukhtar, H. Molecular Targets for Green Tea in Prostate Cancer Prevention. J. Nutr. 2003, 133, 2417S–2424S.
  37. Kazi, A.; Daniel, K.G.; Smith, D.M.; Kumar, N.B.; Dou, Q.P. Inhibition of the proteasome activity, a novel mechanism associated with the tumor cell apoptosis-inducing ability of genistein. Biochem. Pharm. 2003, 66, 965–976.
  38. Kazi, A.; Wang, Z.; Kumar, N.; Falsetti, S.C.; Chan, T.H.; Dou, Q.P. Structure-activity relationships of synthetic analogs of (-)-epigallocatechin-3-gallate as proteasome inhibitors. Anticancer Res. 2004, 24, 943–954.
  39. Nam, S.; Smith, D.M.; Dou, Q.P. Ester bond-containing tea polyphenols potently inhibit proteasome activity in vitro and in vivo. J. Biol. Chem. 2001, 276, 13322–13330.
  40. Smith, D.M.; Wang, Z.; Kazi, A.; Li, L.H.; Chan, T.H.; Dou, Q.P. Synthetic analogs of green tea polyphenols as proteasome inhibitors. Mol. Med. 2002, 8, 382–392.
  41. Sivakumar, A.S.; Hwang, I. Effects of Sunphenon and Polyphenon 60 on proteolytic pathways, inflammatory cytokines and myogenic markers in H2O2-treated C2C12 cells. J. Biosci. 2015, 40, 53–59.
  42. Vester, H.; Holzer, N.; Neumaier, M.; Lilianna, S.; Nussler, A.K.; Seeliger, C. Green Tea Extract (GTE) improves differentiation in human osteoblasts during oxidative stress. J. Inflamm. 2014, 11, 15.
  43. Aktas, O.; Prozorovski, T.; Smorodchenko, A.; Savaskan, N.E.; Lauster, R.; Kloetzel, P.M.; Infante-Duarte, C.; Brocke, S.; Zipp, F. Green tea epigallocatechin-3-gallate mediates T cellular NF-kappa B inhibition and exerts neuroprotection in autoimmune encephalomyelitis. J. Immunol. 2004, 173, 5794–5800.
  44. Sun, Q.; Zheng, Y.; Zhang, X.; Hu, X.; Wang, Y.; Zhang, S.; Zhang, D.; Nie, H. Novel immunoregulatory properties of EGCG on reducing inflammation in EAE. Front. Biosci. 2013, 18, 332–342.
  45. Hayakawa, S.; Ohishi, T.; Miyoshi, N.; Oishi, Y.; Nakamura, Y.; Isemura, M. Anti-Cancer Effects of Green Tea Epigallocatchin-3-Gallate and Coffee Chlorogenic Acid. Molecules 2020, 25, 4553.
  46. Zheng, J.; Yang, B.; Huang, T.; Yu, Y.; Yang, J.; Li, D. Green tea and black tea consumption and prostate cancer risk: An exploratory meta-analysis of observational studies. Nutr. Cancer 2011, 63, 663–672.
  47. Guo, Y.; Zhi, F.; Chen, P.; Zhao, K.; Xiang, H.; Mao, Q.; Wang, X.; Zhang, X. Green tea and the risk of prostate cancer: A systematic review and meta-analysis. Medicine 2017, 96, e6426.
  48. Liu, J.; Li, X.; Hou, J.; Sun, J.; Guo, N.; Wang, Z. Dietary Intake of N-3 and N-6 Polyunsaturated Fatty Acids and Risk of Cancer: Meta-Analysis of Data from 32 Studies. Nutr. Cancer 2021, 73, 901–913.
  49. Lee, J.; Demissie, K.; Lu, S.E.; Rhoads, G.G. Cancer incidence among Korean-American immigrants in the United States and native Koreans in South Korea. Cancer Control 2007, 14, 78–85.
  50. Shen, J.; Obin, M.S.; Zhao, L. The gut microbiota, obesity and insulin resistance. Mol. Asp. Med. 2013, 34, 39–58.
  51. Trefflich, I.; Jabakhanji, A.; Menzel, J.; Blaut, M.; Michalsen, A.; Lampen, A.; Abraham, K.; Weikert, C. Is a vegan or a vegetarian diet associated with the microbiota composition in the gut? Results of a new cross-sectional study and systematic review. Crit. Rev. Food Sci. Nutr. 2020, 60, 2990–3004.
  52. Yang, Q.; Liang, Q.; Balakrishnan, B.; Belobrajdic, D.P.; Feng, Q.J.; Zhang, W. Role of Dietary Nutrients in the Modulation of Gut Microbiota: A Narrative Review. Nutrients 2020, 12, 381.
  53. Byrd, D.A.; Carson, T.L.; Williams, F.; Vogtmann, E. Elucidating the role of the gastrointestinal microbiota in racial and ethnic health disparities. Genome Biol. 2020, 21, 192.
  54. Dwiyanto, J.; Hussain, M.H.; Reidpath, D.; Ong, K.S.; Qasim, A.; Lee, S.W.H.; Lee, S.M.; Foo, S.C.; Chong, C.W.; Rahman, S. Ethnicity influences the gut microbiota of individuals sharing a geographical location: A cross-sectional study from a middle-income country. Sci. Rep. 2021, 11, 2618.
  55. Nishijima, S.; Suda, W.; Oshima, K.; Kim, S.W.; Hirose, Y.; Morita, H.; Hattori, M. The gut microbiome of healthy Japanese and its microbial and functional uniqueness. DNA Res. 2016, 23, 125–133.
  56. Vangay, P.; Johnson, A.J.; Ward, T.L.; Al-Ghalith, G.A.; Shields-Cutler, R.R.; Hillmann, B.M.; Lucas, S.K.; Beura, L.K.; Thompson, E.A.; Till, L.M.; et al. US Immigration Westernizes the Human Gut Microbiome. Cell 2018, 175, 962–972.
  57. Sankaranarayanan, R.; Ramadas, K.; Qiao, Y.L. Managing the changing burden of cancer in Asia. BMC Med. 2014, 12, 3.
  58. Wilson, I.D.; Nicholson, J.K. Gut microbiome interactions with drug metabolism, efficacy, and toxicity. Transl. Res. 2017, 179, 204–222.
  59. Cheng, W.Y.; Wu, C.Y.; Yu, J. The role of gut microbiota in cancer treatment: Friend or foe? Gut 2020, 69, 1867–1876.
  60. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103.
  61. Gan, R.Y.; Li, H.B.; Sui, Z.Q.; Corke, H. Absorption, metabolism, anti-cancer effect and molecular targets of epigallocatechin gallate (EGCG): An updated review. Crit. Rev. Food Sci. Nutr. 2018, 58, 924–941.
  62. Guo, T.; Song, D.; Cheng, L.; Zhang, X. Interactions of tea catechins with intestinal microbiota and their implication for human health. Food Sci. Biotechnol. 2019, 28, 1617–1625.
  63. Azad, M.A.K.; Sarker, M.; Li, T.; Yin, J. Probiotic Species in the Modulation of Gut Microbiota: An Overview. BioMed Res. Int. 2018, 2018, 9478630.
  64. Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277.
  65. Chiou, Y.-S.; Wu, J.-C.; Huang, Q.; Shahidi, F.; Wang, Y.-J.; Ho, C.-T.; Pan, M.-H. Metabolic and colonic microbiota transformation may enhance the bioactivities of dietary polyphenols. J. Funct. Foods 2014, 7, 3–25.
  66. Jang, Y.G.; Ko, E.B.; Choi, K.C. Gallic acid, a phenolic acid, hinders the progression of prostate cancer by inhibition of histone deacetylase 1 and 2 expression. J. Nutr. Biochem. 2020, 84, 108444.
  67. Liu, K.C.; Huang, A.C.; Wu, P.P.; Lin, H.Y.; Chueh, F.S.; Yang, J.S.; Lu, C.C.; Chiang, J.H.; Meng, M.; Chung, J.G. Gallic acid suppresses the migration and invasion of PC-3 human prostate cancer cells via inhibition of matrix metalloproteinase-2 and -9 signaling pathways. Oncol. Rep. 2011, 26, 177–184.
  68. Jin, J.S.; Touyama, M.; Hisada, T.; Benno, Y. Effects of green tea consumption on human fecal microbiota with special reference to Bifidobacterium species. Microbiol. Immunol. 2012, 56, 729–739.
  69. Liao, Z.L.; Zeng, B.H.; Wang, W.; Li, G.H.; Wu, F.; Wang, L.; Zhong, Q.P.; Wei, H.; Fang, X. Impact of the Consumption of Tea Polyphenols on Early Atherosclerotic Lesion Formation and Intestinal Bifidobacteria in High-Fat-Fed ApoE−/− Mice. Front. Nutr. 2016, 3, 42.
  70. Wang, J.; Tang, L.; Zhou, H.; Zhou, J.; Glenn, T.C.; Shen, C.L.; Wang, J.S. Long-term treatment with green tea polyphenols modifies the gut microbiome of female sprague-dawley rats. J. Nutr. Biochem. 2018, 56, 55–64.
  71. Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546.
  72. Schluter, J.; Peled, J.U.; Taylor, B.P.; Markey, K.A.; Smith, M.; Taur, Y.; Niehus, R.; Staffas, A.; Dai, A.; Fontana, E.; et al. The gut microbiota is associated with immune cell dynamics in humans. Nature 2020, 588, 303–307.
  73. Shin, J.-H.; Park, Y.-H.; Sim, M.; Kim, S.-A.; Joung, H.; Shin, D.-M. Serum level of sex steroid hormone is associated with diversity and profiles of human gut microbiome. Res. Microbiol. 2019, 170, 192–201.
  74. Pernigoni, N.; Zagato, E.; Calcinotto, A.; Troiani, M.; Mestre, R.P.; Calì, B.; Attanasio, G.; Troisi, J.; Minini, M.; Mosole, S.; et al. Commensal bacteria promote endocrine resistance in prostate cancer through androgen biosynthesis. Science 2021, 374, 216–224.
  75. Zhong, W.; Wu, K.; Long, Z.; Zhou, X.; Zhong, C.; Wang, S.; Lai, H.; Guo, Y.; Lv, D.; Lu, J.; et al. Gut dysbiosis promotes prostate cancer progression and docetaxel resistance via activating NF-κB-IL6-STAT3 axis. Microbiome 2022, 10, 94.
  76. Matsushita, M.; Fujita, K.; Hatano, K.; Hayashi, T.; Kayama, H.; Motooka, D.; Hase, H.; Yamamoto, A.; Uemura, T.; Yamamichi, G.; et al. High-fat diet promotes prostate cancer growth through histamine signaling. Int. J. Cancer 2022, 151, 623–636.
  77. Liss, M.A.; White, J.R.; Goros, M.; Gelfond, J.; Leach, R.; Johnson-Pais, T.; Lai, Z.; Rourke, E.; Basler, J.; Ankerst, D.; et al. Metabolic Biosynthesis Pathways Identified from Fecal Microbiome Associated with Prostate Cancer. Eur. Urol. 2018, 74, 575–582.
  78. Golombos, D.M.; Ayangbesan, A.; O’Malley, P.; Lewicki, P.; Barlow, L.M.; Barbieri, C.E.; Chan, C.; DuLong, C.; Abu-Ali, G.; Huttenhower, C.; et al. The Role of Gut Microbiome in the Pathogenesis of Prostate Cancer: A Prospective, Pilot Study. Urology 2018, 111, 122–128.
  79. Takezawa, K.; Fujita, K.; Matsushita, M.; Motooka, D.; Hatano, K.; Banno, E.; Shimizu, N.; Takao, T.; Takada, S.; Okada, K.; et al. The Firmicutes/Bacteroidetes ratio of the human gut microbiota is associated with prostate enlargement. Prostate 2021, 81, 1287–1293.
  80. Kure, A.; Tsukimi, T.; Ishii, C.; Aw, W.; Obana, N.; Nakato, G.; Hirayama, A.; Kawano, H.; China, T.; Shimizu, F.; et al. Gut environment changes due to androgen deprivation therapy in patients with prostate cancer. Prostate Cancer Prostatic Dis. 2022.
More
Information
Subjects: Others
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , ,
View Times: 407
Entry Collection: Biopharmaceuticals Technology
Revisions: 3 times (View History)
Update Date: 01 Sep 2022
1000/1000