Resveratrol and Tumor Microenvironment: Mechanistic Basis and Therapeutic Targets
<p>Resveratrol chemical structure in cis (<b>A</b>) and <span class="html-italic">trans</span> (<b>B</b>) isoforms. <span class="html-italic">Trans</span> is the biologically active form. Created using Biorender software.</p> "> Figure 2
<p>Tumor microenvironment. Various host cells and proteins work together to support cancer cell survival and progression. Created using Biorender software.</p> ">
Abstract
:1. Introduction
2. Resveratrol: Sources, Bioavailability, Absorption and Metabolism
3. Tumor Microenvironment
4. Targets of Resveratrol in Tumor Microenvironment
4.1. Reactive Oxygen Species (ROS)
4.2. Tumor Associated Macrophages and Indoleamine 2,3-Dioxygenase in Dendritic Cells
4.3. Vascular Endothelial Growth Factor (VEGF)
4.4. Fibrosis
4.5. Interleukin-6 (IL-6)
5. Resveratrol in Anti-Cancer Clinical Studies
5.1. Beneficial Effects of Resveratrol and Its Pharmaceutics
5.2. Completed Clinical Studies
6. Resveratrol in the Pharmaceutical Industry and Supplement Market
7. Resveratrol and Cancer Prevention
8. Safety Aspects of Resveratrol Treatment
9. Conclusions and Future Perspectives
Funding
Acknowledgments
Conflicts of Interest
References
- Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
- World Health Organization. Global Health Observatory; World Health Organization: Geneva, Switzerland, 2018; p. 10. [Google Scholar]
- Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.M.C.S.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef]
- Donaldson, M.S. Nutrition and cancer: A review of the evidence for an anti-cancer diet. Nutrition 2004, 21, 1–21. [Google Scholar] [CrossRef] [Green Version]
- Talib, W.H. Consumption of garlic and lemon aqueous extracts combination reduces tumor burden by angiogenesis inhibition, apoptosis induction, and immune system modulation. Nutrition 2017, 43, 89–97. [Google Scholar] [CrossRef]
- Vecchia, C.L.; Bosetti, C. Diet and cancer risk in Mediterranean countries: Open issues. Public Health Nutr. 2006, 9, 1077–1082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, J.; Yang, B.; Huang, T.; Yu, Y.; Zheng, J.; Yang, B.; Huang, T. Green Tea and Black Tea Consumption and Prostate Cancer Risk: An Exploratory Meta-Analysis of Observational Studies Green Tea and Black Tea Consumption and Prostate Cancer Risk: An Exploratory Meta-Analysis of Observational Studies. Nutr. Cancer 2011, 37–41. [Google Scholar]
- Block, K.I.; Gyllenhaal, C.; Lowe, L.; Amedei, A.; Amin, A.R.M.R.; Amin, A.; Aquilano, K.; Arbiser, J.; Arreola, A.; Arzumanyan, A.; et al. Seminars in Cancer Biology Designing a broad-spectrum integrative approach for cancer prevention and treatment. Semin. Cancer Biol. 2015, 35, S276–S304. [Google Scholar] [CrossRef] [PubMed]
- Al-hadid, S.A.; Ali, M.B.W.; Al-yasari, I.H.; Ali, M.R.A. Role of curcumin in regulating p53 in breast cancer: An overview of the mechanism of action. Breast Cancer-Targets Ther. 2018, 10, 207–217. [Google Scholar]
- Falah, R.R.; Talib, W.H.; Shbailat, S.J. Combination of metformin and curcumin targets breast cancer in mice by angiogenesis inhibition, immune system modulation and induction of p53 independent apoptosis. Ther. Adv. Med Oncol. 2017, 235–252. [Google Scholar] [CrossRef]
- Talib, W.H.; Alkury, L.T. Biomedicine & Pharmacotherapy Parthenolide inhibits tumor-promoting effects of nicotine in lung cancer by inducing P53-dependent apoptosis and inhibiting VEGF expression. Biomed. Pharmacother. 2018, 107, 1488–1495. [Google Scholar]
- Banerjee, S.; Li, Y.; Wang, Z.; Sarkar, F.H. Multi-targeted therapy of cancer by genistein. Cancer Lett. 2008, 269, 226–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Talib, W.H. Regressions of Breast Carcinoma Syngraft Following Treatment with Piperine in Combination with Thymoquinone. Sci. Pharm. 2017, 85, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hinshaw, D.C.; Shevde, L.A. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res. 2019, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kollár, P.; Hotolová, H. Biological Effects of Resveratrol and Other Constituents of Wine. Ceska Slov. Farm. 2003, 52, 272–281. [Google Scholar]
- Catalgol, B.; Batirel, S.; Taga, Y.; Ozer, N.K. Resveratrol: French paradox revisited. Front. Pharmacol. 2012, 3, 1–18. [Google Scholar] [CrossRef] [Green Version]
- Almeida, L.; Vaz-da-Silva, M.; Falcão, A.; Soares, E.; Costa, R.; Loureiro, A.I.; Fernandes-Lopes, C.; Rocha, J.F.; Nunes, T.; Wright, L.; et al. Pharmacokinetic and safety profile of trans-resveratrol in a rising multiple-dose study in healthy volunteers. Mol. Nutr. Food Res. 2009, 53 (Suppl. 1), 7–15. [Google Scholar] [CrossRef]
- Kroon, P.A.; Iyer, A.; Chunduri, P.; Chan, V.; Brown, L. The Cardiovascular Nutrapharmacology of Resveratrol: Pharmacokinetics, Molecular Mechanisms and Therapeutic Potential. Curr. Med. Chem. 2010, 17, 2442–2455. [Google Scholar] [CrossRef]
- Gambini, J.; Inglés, M.; Olaso, G.; Lopez-Grueso, R.; Bonet-Costa, V.; Gimeno-Mallench, L.; Mas-Bargues, C.; Abdelaziz, K.M.; Gomez-Cabrera, M.C.; Vina, J.; et al. Properties of Resveratrol: In Vitro and In Vivo Studies about Metabolism, Bioavailability, and Biological Effects in Animal Models and Humans. Oxidative Med. Cell. Longev. 2015, 2015. [Google Scholar] [CrossRef] [Green Version]
- Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef] [Green Version]
- Wenzel, E.; Somoza, V. Metabolism and bioavailability of trans -resveratrol. Mol. Cell. Biol. 2005, 49, 472–481. [Google Scholar]
- Sergides, C.; Chirilă, M.; Silvestro, L.; Pitta, D.; Pittas, A. Bioavailability and safety study of resveratrol 500 mg tablets in healthy male and female volunteers. Exp. Ther. Med. 2016, 11, 164–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vitaglione, P.; Sforza, S.; Galaverna, G.; Ghidini, C.; Caporaso, N.; Vescovi, P.P.; Fogliano, V.; Marchelli, R. Bioavailability of trans-resveratrol from red wine in humans. Mol. Nutr. Food Res. 2005, 49, 495–504. [Google Scholar] [CrossRef] [PubMed]
- Delmas, D.; Aires, V.; Limagne, E.; Dutartre, P.; Mazué, F.; Ghiringhelli, F.; Latruffe, N. Transport, stability, and biological activity of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 48–59. [Google Scholar] [CrossRef] [PubMed]
- Boocock, D.J.; Faust, G.E.S.; Patel, K.R.; Schinas, A.M.; Brown, V.A.; Ducharme, M.P.; Booth, T.D.; Crowell, J.A.; Perloff, M.; Gescher, A.J. Phase I dose escalation pharmacokinetic study in healthy volunteers of resveratrol, a potential cancer chemopreventive agent. Cancer Epidemiol. Prev. Biomark. 2007, 16, 1246–1252. [Google Scholar] [CrossRef] [Green Version]
- Marier, J.F.; Vachon, P.; Gritsas, A.; Zhang, J.; Moreau, J.P.; Ducharme, M.P. Metabolism and disposition of resveratrol in rats: Extent of absorption, glucuronidation, and enterohepatic recirculation evidenced by a linked-rat model. J. Pharmacol. Exp. Ther. 2002, 302, 369–373. [Google Scholar] [CrossRef] [Green Version]
- Bode, L.M.; Bunzel, D.; Huch, M.; Cho, G.-S.; Ruhland, D.; Bunzel, M.; Bub, A.; Franz, C.M.A.P.; Kulling, S.E. In vivo and in vitro metabolism of trans -resveratrol by human gut. Am. J. Clin. Nutr. 2013, 295–309. [Google Scholar] [CrossRef]
- Goldberg, D.M.; Yan, J.; Soleas, G.J. Absorption of three wine-related polyphenols in three different matrices by healthy subjects. Clin. Biochem. 2003, 36, 79–87. [Google Scholar] [CrossRef]
- Ndiaye, M.; Kumar, R.; Ahmad, N. Resveratrol in cancer management: Where are we and where we go from here? Resveratrol in cancer management: Where are we and where we go from here? Ann. N. Y. Acad. Sci. 2011, 1215, 144–149. [Google Scholar] [CrossRef]
- Vitrac, X.; Desmoulière, A.; Brouillaud, B.; Krisa, S.; Deffieux, G.; Barthe, N.; Rosenbaum, J.; Mérillon, J.M. Distribution of [14C]-trans-resveratrol, a cancer chemopreventive polyphenol, in mouse tissues after oral administration. Life Sci. 2003, 72, 2219–2233. [Google Scholar] [CrossRef]
- Manach, C.; Milenkovic, D.; Van de Wiele, T.; Rodriguez-Mateos, A.; de Roos, B.; Garcia-Conesa, M.T.; Landberg, R.; Gibney, E.R.; Heinonen, M.; Tomás-Barberán, F.; et al. Addressing the inter-individual variation in response to consumption of plant food bioactives: Towards a better understanding of their role in healthy aging and cardiometabolic risk reduction. Mol. Nutr. Food Res. 2017, 61, 1–16. [Google Scholar] [CrossRef] [Green Version]
- Ritter, J.K. Intestinal UGTs as potential modifiers of pharmacokinetics and biological responses to drugs and xenobiotics. Expert Opin. Drug Metab. Toxicol. 2007, 3, 93–107. [Google Scholar] [CrossRef]
- Brown, V.A.; Patel, K.R.; Viskaduraki, M.; Crowell, J.A.; Perloff, M.; Booth, T.D.; Vasilinin, G.; Sen, A.; Schinas, A.M.; Piccirilli, G.; et al. Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: Safety, pharmacokinetics, and effect on the insulin-like growth factor axis. Cancer Res. 2010, 70, 9003–9011. [Google Scholar] [CrossRef] [Green Version]
- Wightman, E.L.; Reay, J.L.; Haskell, C.F.; Williamson, G.; Dew, T.P.; Kennedy, D.O. Effects of resveratrol alone or in combination with piperine on cerebral blood flow parameters and cognitive performance in human subjects: A randomised, double-blind, placebo-controlled, cross-over investigation. Br. J. Nutr. 2014, 203–213. [Google Scholar] [CrossRef]
- De Santi, C.; Pietrabissa, A.; Spisni, R.; Mosca, F.; Pacifici, G.M. Sulphation of resveratrol, a natural compound present in wine, and its inhibition by natural avonoids. Xenobiotica 2000, 30, 857–866. [Google Scholar] [CrossRef]
- Soleas, B.G.J.; Angelini, M.; Grass, L.; Diamandis, E.P.; Goldberg, D.M. Absorption of trans-Resveratrol in Rats. Method Enzymol. 2001, 335, 145–154. [Google Scholar]
- Basu, N.K.; Kole, L.; Basu, M.; McDonagh, A.F.; Owens, I.S. Targeted inhibition of glucuronidation markedly improves drug efficacy in mice—A model. Biochem. Biophys. Res. Commun. 2007, 360, 7–13. [Google Scholar] [CrossRef] [Green Version]
- Hoshino, J.; Park, E.-j.; Kondratyuk, T.P.; Marler, L.; Pezzuto, J.M.; Breemen, R.B.V.; Mo, S.; Li, Y.; Cushman, M. Selective Synthesis and Biological Evaluation of Sulfate-Conjugated Resveratrol Metabolites. Am. Chem. Soc. 2010, 5033–5043. [Google Scholar] [CrossRef] [Green Version]
- Patel, K.R.; Scott, E.; Brown, V.A.; Gescher, A.J.; Steward, W.P.; Brown, K. Clinical trials of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 161–169. [Google Scholar] [CrossRef]
- Menet, M.C.; Baron, S.; Taghi, M.; Diestra, R.; Dargère, D.; Laprévote, O.; Nivet-Antoine, V.; Beaudeux, J.L.; Bédarida, T.; Cottart, C.H. Distribution of trans-resveratrol and its metabolites after acute or sustained administration in mouse heart, brain, and liver. Mol. Nutr. Food Res. 2017, 61, 1–33. [Google Scholar] [CrossRef]
- Storniolo, C.E.; Moreno, J.J. Resveratrol metabolites have an antiproliferative effect on intestinal epithelial cancer cells. Food Chem. 2012, 134, 1385–1391. [Google Scholar] [CrossRef]
- Lasa, A.; Churruca, I.; Eseberri, I.; Andrés-Lacueva, C.; Portillo, M.P. Delipidating effect of resveratrol metabolites in 3T3-L1 adipocytes. Mol. Nutr. Food Res. 2012, 56, 1559–1568. [Google Scholar] [CrossRef] [PubMed]
- Zamora-Ros, R.; Urpi-Sarda, M.; Lamuela-Raventós, R.M.; Martínez-González, M.Á.; Salas-Salvadó, J.; Arós, F.; Fitó, M.; Lapetra, J.; Estruch, R.; Andres-Lacueva, C. High urinary levels of resveratrol metabolites are associated with a reduction in the prevalence of cardiovascular risk factors in high-risk patients. Pharmacol. Res. 2012, 65, 615–620. [Google Scholar] [CrossRef] [PubMed]
- Burkon, A.; Somoza, V. Quantification of free and protein-bound trans-resveratrol metabolites and identification of Two novel resveratrol metabolites in human plasma. Mol. Nutr. Food Res. 2008, 52, 549–557. [Google Scholar] [CrossRef]
- Andres-Lacueva, C.; MacArulla, M.T.; Rotches-Ribalta, M.; Boto-Ordóñez, M.; Urpi-Sarda, M.; Rodríguez, V.M.; Portillo, M.P. Distribution of resveratrol metabolites in liver, adipose tissue, and skeletal muscle in rats fed different doses of this polyphenol. J. Agric. Food Chem. 2012, 60, 4833–4840. [Google Scholar] [CrossRef]
- Miksits, M.; Wlcek, K.; Svoboda, M.; Kunert, O.; Haslinger, E.; Thalhammer, T.; Szekeres, T.; Jäger, W. Antitumor activity of resveratrol and its sulfated metabolites against human breast cancer cells. Planta Med. 2009, 75, 1227–1230. [Google Scholar] [CrossRef]
- Kiev, A.; Anumonye, A. A Comparative Study. Int. J. Ment. Health 1976, 5, 50–59. [Google Scholar] [CrossRef]
- Bird, J.K.; Raederstorff, D.; Weber, P.; Steinert, R.E. Cardiovascular and antiobesity effects of resveratrol mediated through the gut microbiota. Adv. Nutr. 2017, 8, 839–849. [Google Scholar] [CrossRef]
- Albini, A.; Sporn, M.B. The tumour microenvironment as a target for chemoprevention. Nat. Rev. Cancer 2007, 7, 139–147. [Google Scholar] [CrossRef]
- Sounni, N.E.; Noel, A. Targeting the tumor microenvironment for cancer therapy. Clin. Chem. 2013, 59, 85–93. [Google Scholar] [CrossRef] [Green Version]
- Lu, P.; Weaver, V.M.; Werb, Z. The extracellular matrix: A dynamic niche in cancer progression. J. Cell Biol. 2012, 196, 395–406. [Google Scholar] [CrossRef]
- Hanahan, D.; Coussens, L.M. Accessories to the Crime: Functions of Cells Recruited to the Tumor Microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef] [Green Version]
- Bissell, M.J.; Hall, H.G.; Parry, G. How does the extracellular matrix direct gene expression? J. Theor. Biol. 1982, 99, 31–68. [Google Scholar] [CrossRef]
- Dvorak, H.F. Tumors: Wounds that do not heal. Similarities between tumor stroma generation and wound healing. N. Engl. J. Med. 1986, 315, 1650–1659. [Google Scholar]
- Kannan, K.; Jain, S.K. Oxidative stress and apoptosis. Pathophysiology 2000, 7, 153–163. [Google Scholar] [CrossRef]
- Macip, S.; Igarashi, M.; Berggren, P.; Yu, J.; Lee, S.W.; Aaronson, S.A. Influence of Induced Reactive Oxygen Species in p53-Mediated Cell Fate Decisions. Mol. Cell. Biol. 2003, 23, 8576–8585. [Google Scholar] [CrossRef] [Green Version]
- Azmi, A.S.; Bhat, S.H.; Hanif, S.; Hadi, S.M. Plant polyphenols mobilize endogenous copper in human peripheral lymphocytes leading to oxidative DNA breakage: A putative mechanism for anticancer properties. FEBS Lett. 2006, 580, 533–538. [Google Scholar] [CrossRef] [Green Version]
- Chandel, N.S.; Vander Heiden, M.G.; Thompson, C.B.; Schumacker, P.T. Redox regulation of p53 during hypoxia. Oncogene 2000, 19, 3840–3848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bode, A.M.; Dong, Z. Post-translational modification of p53 in tumorigenesis. Nat. Rev. Cancer 2004, 4, 793–805. [Google Scholar] [CrossRef]
- Luo, H.; Yang, A.; Schulte, B.A.; Wargovich, M.J.; Wang, G.Y. Resveratrol Induces Premature Senescence in Lung Cancer Cells via ROS-Mediated DNA Damage. PLoS ONE 2013, 8, e60065. [Google Scholar] [CrossRef] [Green Version]
- Zhou, X.; Chen, M.; Zeng, X.; Yang, J.; Deng, H.; Yi, L.; Mi, M.T. Resveratrol regulates mitochondrial reactive oxygen species homeostasis through Sirt3 signaling pathway in human vascular endothelial cells. Cell Death Dis. 2014, 5, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heo, J.R.; Kim, S.M.; Hwang, K.A.; Kang, J.H.; Choi, K.C. Resveratrol induced reactive oxygen species and endoplasmic reticulum stress-mediated apoptosis, and cell cycle arrest in the A375SM malignant melanoma cell line. Int. J. Mol. Med. 2018, 42, 1427–1435. [Google Scholar] [CrossRef] [PubMed]
- Ji, S.; Zheng, Z.; Liu, S.; Ren, G.; Gao, J.; Zhang, Y.; Li, G. Resveratrol Promotes Oxidative Stress to Drive DLC1 Mediated Cellular Senescence in Cancer Cells; Elsevier Inc.: Houston, TX, USA, 2018; Volume 370, pp. 292–302. [Google Scholar]
- Lee, Y.-J.; Lee, G.J.; Shin, S.; Heo, S.-H.; Park, C.-R.; Nam, H.-S.; Cho, M.-K.; Lee, S.-H. Cisplatin and resveratrol induce apoptosis and autophagy following oxidative stress in malignant mesothelioma cells. Food Chem. Toxicol. 2016, 97, 96–107. [Google Scholar] [CrossRef]
- Physiology, C. FOXO1, a Potential Therapeutic Target, Regulates Autophagic Flux, Oxidative Stress, Mitochondrial Dysfunction, and Apoptosis in Human Cholangiocarcinoma QBC939 Cells. Cell. Physiol. Biochem. 2018, 45, 1506–1514. [Google Scholar]
- Hadi, S.M.; Asad, S.F.; Singh, S.; Ahmad, A. Putative mechanism for anticancer and apoptosis-inducing properties of plant-derived polyphenolic compounds. IUBMB Life 2000, 50, 167–171. [Google Scholar]
- Kerkar, S.P.; Restifo, N.P. Cellular constituents of immune escape within the tumor microenvironment. Cancer Res. 2012, 72, 3125–3130. [Google Scholar] [CrossRef] [Green Version]
- Qian, B.-Z.; Pollard, J.W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weiss, J.M.; Subleski, J.J.; Wigginton, J.M.; Wiltrout, R.H. Immunotherapy of cancer by IL-12-based cytokine combinations. Export. Opin. 2007, 1705–1722. [Google Scholar] [CrossRef]
- Fiorentino, D.F.; Zlotnik, A.; Vieira, P.; Mosmann, T.R.; Howard, M.; Moore, K.W.; Garra, A. IL-10 acts on the antigen-presenting cell to inhibit cytokine production by Th1 cells. J. Immunol. 1991, 146, 3444–3451. [Google Scholar]
- Fan, Q.-M.; Jing, Y.-Y.; Yu, G.-F.; Kou, X.-R.; Ye, F.; Gao, L.; Li, R.; Zhao, Q.-D.; Yang, Y.; Lu, Z.-H.; et al. Tumor-associated macrophages promote cancer stem cell-like properties via transforming growth factor-beta1-induced epithelial–mesenchymal transition in hepatocellular carcinoma. Cancer Lett. 2014, 352, 160–168. [Google Scholar] [CrossRef]
- Casey, S.C.; Amedei, A.; Aquilano, K.; Azmi, A.S.; Benencia, F.; Bhakta, D.; Bilsland, A.E.; Boosani, C.S.; Chen, S.; Ciriolo, M.R.; et al. Cancer prevention and therapy through the modulation of the tumor microenvironment. Semin. Cancer Biol. 2015, 35, S199–S223. [Google Scholar] [CrossRef]
- Lyford-Pike, S.; Peng, S.; Young, G.D.; Taube, J.M.; Westra, W.H.; Akpeng, B.; Bruno, T.C.; Richmon, J.D.; Wang, H.; Bishop, J.A. Evidence for a role of the PD-1: PD-L1 pathway in immune resistance of HPV-associated head and neck squamous cell carcinoma. Cancer Res. 2013, 73, 1733–1741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, L.; Chen, B.; Jiang, R.; Li, J.; Wang, B. Resveratrol inhibits lung cancer growth by suppressing M2-like polarization of tumor associated macrophages. Cell. Immunol. 2017, 311, 86–93. [Google Scholar] [CrossRef]
- Kimura, Y.; Sumiyoshi, M. Resveratrol prevents tumor growth and metastasis by inhibiting lymphangiogenesis and M2 macrophage activation and differentiation in tumor-associated macrophages. Nutr. Cancer 2016, 68, 667–678. [Google Scholar] [CrossRef] [PubMed]
- Mukherjee, S.; Hussaini, R.; White, R.; Atwi, D.; Fried, A.; Sampat, S.; Piao, L.; Pan, Q.; Banerjee, P. TriCurin, a synergistic formulation of curcumin, resveratrol, and epicatechin gallate, repolarizes tumor-associated macrophages and triggers an immune response to cause suppression of HPV+ tumors. Cancer Immunol. Immunother. 2018, 67, 761–774. [Google Scholar] [CrossRef] [PubMed]
- Jeong, S.K.; Yang, K.; Park, Y.S.; Choi, Y.J.; Oh, S.J.; Lee, C.W.; Lee, K.Y.; Jeong, M.H.; Jo, W.S. Interferon gamma induced by resveratrol analog, HS-1793, reverses the properties of tumor associated macrophages. Int. Immunopharmacol. 2014, 22, 303–310. [Google Scholar] [CrossRef] [PubMed]
- Riboldi, E.; Musso, T.; Moroni, E.; Bernasconi, S.; Rusnati, M.; Presta, M.; Sozzani, S. Cutting Edge: Proangiogenic Properties of Alternatively Activated Dendritic Cells. J. Immunol. 2005, 175, 2788–2792. [Google Scholar] [CrossRef]
- Lutz, M.B.; Schuler, G. Immature, semi-mature and fully mature dendritic cells: which signals induce tolerance or immunity? Trends Immunol. 2002, 23, 445–449. [Google Scholar] [CrossRef]
- Munn, D.H.; Mellor, A.L. Indoleamine 2,3 dioxygenase and metabolic control of immune responses. Trends Immunol. 2013, 34, 137–143. [Google Scholar] [CrossRef] [Green Version]
- Sharma, M.D.; Baban, B.; Chandler, P.; Hou, D.-Y.; Singh, N.; Yagita, H.; Azuma, M.; Blazar, B.R.; Mellor, A.L.; Munn, D.H. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J. Clin. Investig. 2007, 117, 2570–2582. [Google Scholar] [CrossRef] [Green Version]
- Frumento, G.; Rotondo, R.; Tonetti, M.; Damonte, G.; Benatti, U.; Ferrara, G.B. Tryptophan-derived Catabolites Are Responsible for Inhibition of T and Natural Killer Cell Proliferation Induced by Indoleamine 2,3-Dioxygenase. J. Exp. Med. 2002, 196, 459–468. [Google Scholar] [CrossRef] [Green Version]
- Zamanakou, M.; Germenis, A.E.; Karanikas, V. Tumor immune escape mediated by indoleamine 2,3-dioxygenase. Immunol. Lett. 2007, 111, 69–75. [Google Scholar] [CrossRef] [PubMed]
- Noh, K.T.; Chae, S.H.; Chun, S.H.; Jung, I.D.; Kang, H.K.; Park, Y.-M. Resveratrol suppresses tumor progression via the regulation of indoleamine 2,3-dioxygenase. Biochem. Biophys. Res. Commun. 2013, 431, 348–353. [Google Scholar] [CrossRef]
- Gualdoni, G.A.; Fuchs, D.; Zlabinger, G.J.; Gostner, J.M. Resveratrol intake enhances indoleamine-2,3-dioxygenase activity in humans. Pharmacol. Rep. 2016, 68, 1065–1068. [Google Scholar] [CrossRef] [PubMed]
- Zeng, Z.; Li, Y.; Pan, Y.; Lan, X.; Song, F.; Sun, J.; Zhou, K.; Liu, X.; Ren, X.; Wang, F. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat. Commun. 2018, 9, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bergers, G.; Benjamin, L.E. Tumorigenesis and the angiogenic switch. Nat. Rev. Cancer 2003, 3, 401–410. [Google Scholar] [CrossRef]
- Ferrara, N.; Gerber, H.-P.; LeCouter, J. The biology of VEGF and its receptors. Nat. Med. 2003, 9, 669–676. [Google Scholar] [CrossRef]
- Takahashi, H.; Shibuya, M. The vascular endothelial growth factor (VEGF)/VEGF receptor system and its role under physiological and pathological conditions. Clin. Sci. 2005, 109, 227–241. [Google Scholar] [CrossRef] [Green Version]
- Lapeyre-Prost, A.; Terme, M.; Pernot, S.; Pointet, A.-L.; Voron, T.; Tartour, E.; Taieb, J. Immunomodulatory activity of VEGF in cancer. In International Review of Cell and Molecular Biology; Elsevier: Amsterdam, The Netherlands, 2017; Volume 330, pp. 295–342. [Google Scholar]
- Marigo, I.; Dolcetti, L.; Serafini, P.; Zanovello, P.; Bronte, V. Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells. Immunol. Rev. 2008, 222, 162–179. [Google Scholar] [CrossRef]
- Sebolt-Leopold, J.S.; English, J.M. Mechanisms of drug inhibition of signalling molecules. Nature 2006, 441, 457–462. [Google Scholar] [CrossRef]
- Aggarwal, B.B.; Bhardwaj, A.; Aggarwal, R.S.; Seeram, N.P.; Shishodia, S.; Takada, Y. Role of resveratrol in prevention and therapy of cancer: Preclinical and clinical studies. Anticancer Res. 2004, 24, 2783–2840. [Google Scholar] [PubMed]
- Hu, W.-H.; Chan, G.K.-L.; Duan, R.; Wang, H.-Y.; Kong, X.-P.; Dong, T.T.-X.; Tsim, K.W.-K. Synergy of Ginkgetin and Resveratrol in Suppressing VEGF-Induced Angiogenesis: A Therapy in Treating Colorectal Cancer. Cancers 2019, 11, 1828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, M.-T.; Yen, M.-L.; Lin, C.-Y.; Kuo, M.-L. Inhibition of vascular endothelial growth factor-induced angiogenesis by resveratrol through interruption of Src-dependent vascular endothelial cadherin tyrosine phosphorylation. Mol. Pharmacol. 2003, 64, 1029–1036. [Google Scholar] [CrossRef] [PubMed]
- Trapp, V.; Parmakhtiar, B.; Papazian, V.; Willmott, L.; Fruehauf, J.P. Anti-angiogenic effects of resveratrol mediated by decreased VEGF and increased TSP1 expression in melanoma-endothelial cell co-culture. Angiogenesis 2010, 13, 305–315. [Google Scholar] [CrossRef] [Green Version]
- Zhang, M.; Li, W.; Yu, L.; Wu, S. The suppressive effect of resveratrol on HIF-1α and VEGF expression after warm ischemia and reperfusion in rat liver. PLoS ONE 2014, 9, e109589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seong, H.; Ryu, J.; Jeong, J.Y.; Chung, I.Y.; Han, Y.S.; Hwang, S.H.; Park, J.M.; Kang, S.S.; Seo, S.W. Resveratrol suppresses vascular endothelial growth factor secretion via inhibition of CXC-chemokine receptor 4 expression in ARPE-19 cells. Mol. Med. Rep. 2015, 12, 1479–1484. [Google Scholar] [CrossRef] [Green Version]
- Bishayee, A.; Petit, D.; Samtani, K. Angioprevention is implicated in resveratrol chemoprevention of experimental hepatocarcinogenesis. J. Carcinog. Mutagen 2010, 1, 102. [Google Scholar] [CrossRef]
- Cao, Z.; Fang, J.; Xia, C.; Shi, X.; Jiang, B.-H. trans-3,4,5′-Trihydroxystibene inhibits hypoxia-inducible factor 1α and vascular endothelial growth factor expression in human ovarian cancer cells. Clin. Cancer Res. 2004, 10, 5253–5263. [Google Scholar] [CrossRef] [Green Version]
- Piersma, B.; Hayward, M.; Weaver, V.M. Fibrosis and cancer: A strained relationship. Biochim. Biophys. Acta Rev. Cancer 2020, 1873, 188356. [Google Scholar] [CrossRef]
- Kalluri, R.; Zeisberg, M. Fibroblasts in cancer. Nat. Rev. Cancer 2006, 6, 392–401. [Google Scholar] [CrossRef]
- Orimo, A.; Gupta, P.B.; Sgroi, D.C.; Arenzana-Seisdedos, F.; Delaunay, T.; Naeem, R.; Carey, V.J.; Richardson, A.L.; Weinberg, R.A. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 2005, 121, 335–348. [Google Scholar] [CrossRef]
- Giannoni, E.; Bianchini, F.; Masieri, L.; Serni, S.; Torre, E.; Calorini, L.; Chiarugi, P. Reciprocal activation of prostate cancer cells and cancer-associated fibroblasts stimulates epithelial-mesenchymal transition and cancer stemness. Cancer Res. 2010, 70, 6945–6956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ha, S.Y.; Yeo, S.-Y.; Xuan, Y.-h.; Kim, S.-H. The prognostic significance of cancer-associated fibroblasts in esophageal squamous cell carcinoma. PLoS ONE 2014, 9, e99955. [Google Scholar] [CrossRef] [PubMed]
- Wikberg, M.L.; Edin, S.; Lundberg, I.V.; Van Guelpen, B.; Dahlin, A.M.; Rutegård, J.; Stenling, R.; Öberg, Å.; Palmqvist, R. High intratumoral expression of fibroblast activation protein (FAP) in colon cancer is associated with poorer patient prognosis. Tumor Biol. 2013, 34, 1013–1020. [Google Scholar] [CrossRef] [Green Version]
- Tran, E.; Chinnasamy, D.; Yu, Z.; Morgan, R.A.; Lee, C.-C.R.; Restifo, N.P.; Rosenberg, S.A. Immune targeting of fibroblast activation protein triggers recognition of multipotent bone marrow stromal cells and cachexia. J. Exp. Med. 2013, 210, 1125–1135. [Google Scholar] [CrossRef]
- Özdemir, B.C.; Pentcheva-Hoang, T.; Carstens, J.L.; Zheng, X.; Wu, C.-C.; Simpson, T.R.; Laklai, H.; Sugimoto, H.; Kahlert, C.; Novitskiy, S.V. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 2014, 25, 719–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramakrishnan, P.; Loh, W.M.; Gopinath, S.C.; Bonam, S.R.; Fareez, I.M.; Mac Guad, R.; Sim, M.S.; Wu, Y.S. Selective phytochemicals targeting pancreatic stellate cells as new anti-fibrotic agents for chronic pancreatitis and pancreatic cancer. Acta Pharm. Sin. B 2020, 10, 399–413. [Google Scholar] [CrossRef]
- Yan, B.; Cheng, L.; Jiang, Z.; Chen, K.; Zhou, C.; Sun, L.; Cao, J.; Qian, W.; Li, J.; Shan, T. Resveratrol inhibits ROS-promoted activation and glycolysis of pancreatic stellate cells via suppression of miR-21. Oxidative Med. Cell. Longev. 2018, 2018. [Google Scholar] [CrossRef]
- Lin, Z.; Zheng, L.-C.; Zhang, H.-J.; Tsang, S.W.; Bian, Z.-X. Anti-fibrotic effects of phenolic compounds on pancreatic stellate cells. BMC Complement. Altern. Med. 2015, 15, 1–7. [Google Scholar] [CrossRef] [Green Version]
- Yao, X.; Huang, J.; Zhong, H.; Shen, N.; Faggioni, R.; Fung, M.; Yao, Y. Targeting interleukin-6 in inflammatory autoimmune diseases and cancers. Pharmacol. Ther. 2014, 141, 125–139. [Google Scholar] [CrossRef]
- Johnson, D.E.; O’Keefe, R.A.; Grandis, J.R. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat. Rev. Clin. Oncol. 2018, 15, 234. [Google Scholar] [CrossRef]
- Rossi, J.-F.; Lu, Z.-Y.; Jourdan, M.; Klein, B. Interleukin-6 as a therapeutic target. Clin. Cancer Res. 2015, 21, 1248–1257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, Y.; Xu, F.; Lu, T.; Duan, Z.; Zhang, Z. Interleukin-6 signaling pathway in targeted therapy for cancer. Cancer Treat. Rev. 2012, 38, 904–910. [Google Scholar] [CrossRef] [PubMed]
- Lippitz, B.E.; Harris, R.A. Cytokine patterns in cancer patients: A review of the correlation between interleukin 6 and prognosis. Oncoimmunology 2016, 5, e1093722. [Google Scholar] [CrossRef] [Green Version]
- Huynh, P.T.; Beswick, E.J.; Coronado, Y.A.; Johnson, P.; O’Connell, M.R.; Watts, T.; Singh, P.; Qiu, S.; Morris, K.; Powell, D.W. CD 90+ stromal cells are the major source of IL-6, which supports cancer stem-like cells and inflammation in colorectal cancer. Int. J. Cancer 2016, 138, 1971–1981. [Google Scholar] [CrossRef] [Green Version]
- Lesina, M.; Wörmann, S.M.; Neuhöfer, P.; Song, L.; Algül, H. Interleukin-6 in Inflammatory and Malignant Diseases of the Pancreas; Seminars in Immunology; Elsevier: Amsterdam, The Netherlands, 2014; pp. 80–87. [Google Scholar]
- Pop, V.-V.; Seicean, A.; Lupan, I.; Samasca, G.; Burz, C.-C. IL-6 roles–Molecular pathway and clinical implication in pancreatic cancer–A systemic review. Immunol. Lett. 2017, 181, 45–50. [Google Scholar] [CrossRef] [PubMed]
- Swartz, M.A.; Iida, N.; Roberts, E.W.; Sangaletti, S.; Wong, M.H.; Yull, F.E.; Coussens, L.M.; DeClerck, Y.A. Tumor microenvironment complexity: Emerging roles in cancer therapy. AACR 2012, 2473–2480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chang, Q.; Daly, L.; Bromberg, J. The IL-6 Feed-forward Loop: A Driver of Tumorigenesis; Seminars in Immunology; Elsevier: Amsterdam, The Netherlands, 2014; pp. 48–53. [Google Scholar]
- Dijkgraaf, E.; Santegoets, S.; Reyners, A.; Goedemans, R.; Wouters, M.; Kenter, G.; Van Erkel, A.; Van Poelgeest, M.; Nijman, H.; Van Der Hoeven, J. A phase I trial combining carboplatin/doxorubicin with tocilizumab, an anti-IL-6R monoclonal antibody, and interferon-α2b in patients with recurrent epithelial ovarian cancer. Ann. Oncol. 2015, 26, 2141–2149. [Google Scholar] [CrossRef]
- Vainer, N.; Dehlendorff, C.; Johansen, J.S. Systematic literature review of IL-6 as a biomarker or treatment target in patients with gastric, bile duct, pancreatic and colorectal cancer. Oncotarget 2018, 9, 29820. [Google Scholar] [CrossRef] [Green Version]
- Lee, M.-H.; Kundu, J.K.; Keum, Y.-S.; Cho, Y.-Y.; Surh, Y.-J.; Choi, B.Y. Resveratrol inhibits IL-6-induced transcriptional activity of AR and STAT3 in human prostate cancer LNCaP-FGC cells. Biomol. Ther. 2014, 22, 426. [Google Scholar] [CrossRef] [Green Version]
- Chun, J.Y.; Nadiminty, N.; Dutt, S.; Lou, W.; Yang, J.C.; Kung, H.-J.; Evans, C.P.; Gao, A.C. Interleukin-6 regulates androgen synthesis in prostate cancer cells. Clin. Cancer Res. 2009, 15, 4815–4822. [Google Scholar] [CrossRef] [Green Version]
- Van der Poel, H.; Zevenhoven, J.; Bergman, A. Pim1 regulates androgen-dependent survival signaling in prostate cancer cells. Urol. Int. 2010, 84, 212–220. [Google Scholar] [CrossRef] [PubMed]
- Ha, S.; Iqbal, N.J.; Mita, P.; Ruoff, R.; Gerald, W.L.; Lepor, H.; Taneja, S.S.; Lee, P.; Melamed, J.; Garabedian, M.J. Phosphorylation of the androgen receptor by PIM1 in hormone refractory prostate cancer. Oncogene 2013, 32, 3992–4000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zemskova, M.; Sahakian, E.; Bashkirova, S.; Lilly, M. The PIM1 kinase is a critical component of a survival pathway activated by docetaxel and promotes survival of docetaxel-treated prostate cancer cells. J. Biol. Chem. 2008, 283, 20635–20644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baur, J.A.; Sinclair, D.A. Therapeutic potential of resveratrol: The in vivo evidence. Nat. Rev. Drug Discov. 2006, 5, 493–506. [Google Scholar] [CrossRef] [PubMed]
- Soleas, G.J.; Grass, L.; Josephy, P.D.; Goldberg, D.M.; Diamandis, E.P. A comparison of the anticarcinogenic properties of four red wine polyphenols. Clin. Biochem. 2006, 39, 492–497. [Google Scholar] [CrossRef]
- Nguyen, A.V.; Martinez, M.; Stamos, M.J.; Moyer, M.P.; Planutis, K.; Hope, C.; Holcombe, R.F. Results of a phase I pilot clinical trial examining the effect of plant-derived resveratrol and grape powder on Wnt pathway target gene expression in colonic mucosa and colon cancer. Cancer Manag. Res. 2009, 1, 25. [Google Scholar]
- Patel, K.R.; Brown, V.A.; Jones, D.J.; Britton, R.G.; Hemingway, D.; Miller, A.S.; West, K.P.; Booth, T.D.; Perloff, M.; Crowell, J.A. Clinical pharmacology of resveratrol and its metabolites in colorectal cancer patients. Cancer Res. 2010, 70, 7392–7399. [Google Scholar] [CrossRef] [Green Version]
- Howells, L.M.; Berry, D.P.; Elliott, P.J.; Jacobson, E.W.; Hoffmann, E.; Hegarty, B.; Brown, K.; Steward, W.; Gescher, A.J. Phase I randomized, double-blind pilot study of micronized resveratrol (SRT501) in patients with hepatic metastases—Safety, pharmacokinetics, and pharmacodynamics. Cancer Prev. Res. 2011, 4, 1419–1425. [Google Scholar] [CrossRef] [Green Version]
- Popat, R.; Plesner, T.; Davies, F.; Cook, G.; Cook, M.; Elliott, P.; Jacobson, E.; Gumbleton, T.; Oakervee, H.; Cavenagh, J. A phase 2 study of SRT 501 (resveratrol) with bortezomib for patients with relapsed and or refractory multiple myeloma. Br. J. Haematol. 2013, 160, 714–717. [Google Scholar] [CrossRef]
- Paller, C.J.; Rudek, M.A.; Zhou, X.C.; Wagner, W.D.; Hudson, T.S.; Anders, N.; Hammers, H.J.; Dowling, D.; King, S.; Antonarakis, E.S.; et al. A phase I study of muscadine grape skin extract in men with biochemically recurrent prostate cancer: Safety, tolerability, and dose determination. Prostate 2015, 75, 1518–1525. [Google Scholar] [CrossRef] [Green Version]
- Zhu, W.; Qin, W.; Zhang, K.; Rottinghaus, G.E.; Chen, Y.-C.; Kliethermes, B.; Sauter, E.R. Trans-resveratrol alters mammary promoter hypermethylation in women at increased risk for breast cancer. Nutr. Cancer 2012, 64, 393–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, W.; Qin, W.; Hewett, J.E.; Sauter, E.R. Quantitative evaluation of DNA hypermethylation in malignant and benign breast tissue and fluids. Int. J. Cancer 2010, 126, 474–482. [Google Scholar] [CrossRef] [Green Version]
- Masferrer, J.L.; Leahy, K.M.; Koki, A.T.; Zweifel, B.S.; Settle, S.L.; Woerner, B.M.; Edwards, D.A.; Flickinger, A.G.; Moore, R.J.; Seibert, K. Antiangiogenic and antitumor activities of cyclooxygenase-2 inhibitors. Cancer Res. 2000, 60, 1306–1311. [Google Scholar]
- Jogie-Brahim, S.; Feldman, D.; Oh, Y. Unraveling insulin-like growth factor binding protein-3 actions in human disease. Endocr. Rev. 2009, 30, 417–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kjaer, T.N.; Ornstrup, M.J.; Poulsen, M.M.; Jørgensen, J.O.; Hougaard, D.M.; Cohen, A.S.; Neghabat, S.; Richelsen, B.; Pedersen, S.B. Resveratrol reduces the levels of circulating androgen precursors but has no effect on, testosterone, dihydrotestosterone, PSA levels or prostate volume. A 4-month randomised trial in middle-aged men. Prostate 2015, 75, 1255–1263. [Google Scholar] [CrossRef]
- Cai, H.; Scott, E.; Kholghi, A.; Andreadi, C.; Rufini, A.; Karmokar, A.; Britton, R.G.; Horner-Glister, E.; Greaves, P.; Jawad, D. Cancer chemoprevention: Evidence of a nonlinear dose response for the protective effects of resveratrol in humans and mice. Sci. Transl. Med. 2015, 7, 298ra117. [Google Scholar] [CrossRef] [Green Version]
- Singh, C.K.; Ndiaye, M.A.; Ahmad, N. Resveratrol and cancer: Challenges for clinical translation. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2015, 1852, 1178–1185. [Google Scholar] [CrossRef] [Green Version]
- Hormones, E.; Group, B.C.C. Endogenous sex hormones and breast cancer in postmenopausal women: Reanalysis of nine prospective studies. J. Natl. Cancer Inst. 2002, 94, 606–616. [Google Scholar]
- Chow, H.S.; Garland, L.L.; Heckman-Stoddard, B.M.; Hsu, C.-H.; Butler, V.D.; Cordova, C.A.; Chew, W.M.; Cornelison, T.L. A pilot clinical study of resveratrol in postmenopausal women with high body mass index: Effects on systemic sex steroid hormones. J. Transl. Med. 2014, 12, 223. [Google Scholar] [CrossRef] [Green Version]
- Renehan, A.G.; Zwahlen, M.; Minder, C.; O’Dwyer, S.T.; Shalet, S.M.; Egger, M. Insulin-like growth factor (IGF)-I, IGF binding protein-3, and cancer risk: Systematic review and meta-regression analysis. Lancet 2004, 363, 1346–1353. [Google Scholar] [CrossRef]
- Chow, H.S.; Garland, L.L.; Hsu, C.-H.; Vining, D.R.; Chew, W.M.; Miller, J.A.; Perloff, M.; Crowell, J.A.; Alberts, D.S. Resveratrol modulates drug-and carcinogen-metabolizing enzymes in a healthy volunteer study. Cancer Prev. Res. 2010, 3, 1168–1175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Juan, M.E.; Vinardell, M.P.; Planas, J.M. The daily oral administration of high doses of trans-resveratrol to rats for 28 days is not harmful. J. Nutr. 2002, 132, 257–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Crowell, J.A.; Korytko, P.J.; Morrissey, R.L.; Booth, T.D.; Levine, B.S. Resveratrol-associated renal toxicity. Toxicol. Sci. Off. J. Soc. Toxicol. 2004, 82, 614–619. [Google Scholar] [CrossRef] [Green Version]
- Johnson, W.; Morrissey, R.; Usborne, A.; Kapetanovic, I.; Crowell, J.; Muzzio, M.; McCormick, D. Subchronic oral toxicity and cardiovascular safety pharmacology studies of resveratrol, a naturally occurring polyphenol with cancer preventive activity. Food Chem. Toxicol. 2011, 49, 3319–3327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Metabolites | Model of Experiment | Effect, Disease |
---|---|---|
Trans-resveratrol | Human [23,39] | antioxidant, cancer [23] |
Trans-resveratrol-4′-O-glucuronide | Human [23,39] Mouse [40] | antioxidant, colon cancer [41] Delipidating age obesity [42] |
Trans-resveratrol-3-O-glucuronide | Human [23,39] | Change lipid profile & control heart rate, Cardiovascular disease [43] |
Trans-resveratrol-diglucuronide | Human [44] Mouse [40] | N |
Trans-resveratrol-3-O-sulfate | Human [20,39] Mouse [40] | antioxidant, colon cancer [41] |
Trans-resveratrol-4′-O-sulfate | Human [20,39] Rat [45] | antitumor, breast cancer [46] |
Cis-resveratrol-3-O-sulfate | Rat [45] | change lipid profile, cardiovascular [43] |
Trans-resveratrol-3,4′-disulfate | Human [39] | antitumor, breast cancer [46] |
Trans-resveratrol-glucuronide-sulfate | Mouse [40] | N |
Dihydroresveratrol | Human [27] | antiproliferative, prostate cancer [47] |
Dihydroresveratrol-glucuronide | Human [19] | N |
Dihydroresveratrol-sulfate | Human [19] Mouse [40] | N |
Dihydroresveratrol-glucuronide-sulfate | Mouse [40] | N |
3,4′-dihydroxy-trans-stilbene | Human [27] | lower cholesterol level, cardiovascular [48] |
Lunularin | Human [27] | lowering food intake, obesity [48] |
Tumor Microenvironment Targets | Resveratrol Effect | Mechanism of Action |
---|---|---|
Reactive oxygen species (ROS) | Inhibition and activation | Increases ROS generation [52] Scavenger of reactive oxygen species [58] |
Tumor associated macrophages | Inhibition | Inhibition of M2 polarization of macrophage [66], inhibition of M2 macrophage activation [67], activates repolarization of macrophage from M2 to M1 phenotype [68] |
Indoleamine 2,3-dioxygenase | Inhibition and activation | Inhibition of IDO expression and activity (50 mg/kg every 2 days for 3 weeks, animal study) [76] Increase in the activity of IDO (oral administration of 5 g resveratrol by 8 volunteers [77] |
Vascular endothelial growth factor (VEGF) | Inhibition | Suppressing VEGF production [97] Inhibiting the production of HIF-1 [103] |
Fibrosis | Inhibition | Inhibit activation, invasion, migration and glycolysis of cells involved in fibrogenesis process [113] Reducing several major fibrogenic mediators, such as a-SMA, type I collagen and fibronectin [114] |
IL-6 | Inhibition | Reduced IL-6-induced AR activity [127] |
Resveratrol Formulation | Dosage Administration | Cancer Type | Sample Size and Phase | Outcome of the Study | Ref |
---|---|---|---|---|---|
Resveratrol | 20 or 80 mg/day for 14 days | Colon | N = 8 Phase 1 | Reduction in the expression of a panel of Wnt target genes indicated inhibition of Wnt signaling in normal colonic mucosa | [131] |
Grape Powder | 80 or 120 g/day for 14 days | ||||
Resveratrol | 0.5 or 1.0 g for 8 days | Colon | N = 20 | Quantification of Resveratrol and its metabolites found in colon tissue with a higher value obtained at right side of the colon. Tumor cell proliferation was reduced by 5% (Ki-67 immunostaining, p = 0.05). | [132] |
Micronized resveratrol (SRT501) | 5.0 g for 20 consecutive days in a 21-day cycle for a max of 12 cycles | Multiple myeloma | N = 24 Phase 2 | Clinical trial conducted for assessment. SRT501 with or without bortezomib was given to multiple myeloma patients. Study was immediately terminated because of the appearance of serious adverse events and observed and minimal effectiveness in relapsed/refractory multiple myeloma patients. However, the previous outcome emphasized the risks of development and administration of the drug such populations. | [33] |
Micronized resveratrol (SRT5001) | 5 g for 14 days | Colorectal cancer patients with hepatic metastasis | N = 6 | Resveratrol was detected on hepatic tissue and a 39% increase in the content of cleaved caspase-3 in malignant hepatic tissue. | [133] |
Micronized resveratrol, (SRT501) | 5.0 g for 14 days | Colorectal with hepatic metastases | N = 9 Phase 1 | The detectable resveratrol in hepatic tissue that increased the cleavage of caspase-3 by 39% was well-tolerated with higher plasma resveratrol level (3.6-fold) in malignant hepatic tissue. | [134] |
Pulverized muscadine grape skin Extract (MPX) | 4000 mg/patient | Biochemically recurrent Prostate cancer patients | N = 14 | MPX was safe but needs further investigation in d phase II trial with dose evaluation | [135] |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Talib, W.H.; Alsayed, A.R.; Farhan, F.; Al Kury, L.T. Resveratrol and Tumor Microenvironment: Mechanistic Basis and Therapeutic Targets. Molecules 2020, 25, 4282. https://doi.org/10.3390/molecules25184282
Talib WH, Alsayed AR, Farhan F, Al Kury LT. Resveratrol and Tumor Microenvironment: Mechanistic Basis and Therapeutic Targets. Molecules. 2020; 25(18):4282. https://doi.org/10.3390/molecules25184282
Chicago/Turabian StyleTalib, Wamidh H., Ahmad Riyad Alsayed, Faten Farhan, and Lina T. Al Kury. 2020. "Resveratrol and Tumor Microenvironment: Mechanistic Basis and Therapeutic Targets" Molecules 25, no. 18: 4282. https://doi.org/10.3390/molecules25184282
APA StyleTalib, W. H., Alsayed, A. R., Farhan, F., & Al Kury, L. T. (2020). Resveratrol and Tumor Microenvironment: Mechanistic Basis and Therapeutic Targets. Molecules, 25(18), 4282. https://doi.org/10.3390/molecules25184282