Molecular Targets of Natural Compounds with Anti-Cancer Properties
Abstract
:1. Introduction
2. Curcumin
3. Acetogenins from Annona muricata
4. Resveratrol
5. Epigallocatechin-3-gallate
6. Quercetin
7. Lycopene
8. Clinical Trials Involving Natural Compounds
9. Conclusions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Hanahan, D.; Wienberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prasad, V. Do cancer drugs improve survival or quality of life? BMJ 2017, 359, 4528. [Google Scholar] [CrossRef] [Green Version]
- Hong, I.S.; Lee, H.Y.; Nam, J.S. Cancer stem cells: The ‘Achilles heel’ of chemoresistant tumors. Recent Pat. Antic. Drug Discov. 2015, 10, 2–22. [Google Scholar] [CrossRef]
- Jiang, W.; Peng, J.; Zhang, Y.; Cho, W.C.; Jin, K. The implications of cancer stem cells for cancer therapy. Int. J. Mol. Sci. 2012, 13, 16636–16657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, P.; Li, B.; Liu, F.; Zhang, M.; Wang, Q.; Liu, Y.; Yao, Y.; Li, D. The epithelial to mesenchymal transition (EMT) and cancer stem cells: Implication for treatment resistance in pancreatic cancer. Mol. Cancer 2017, 16, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haslam, E.; Lilley, T.H.; Warminski, E.; Liao, H.; Cai, Y.; Martin, R.; Gaffney, S.H.; Goulding, P.N.; Luck, G. Polyphenol complexation. A study in molecular recognition. ACS Symp. Ser. 1992, 506, 8–50. [Google Scholar]
- Magrone, T.; Russo, M.A.; Jirillo, E. Cocoa and dark chocolate polyphenols: From biology to clinical applications. Front. Immunol. 2017, 8, 677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Doss, M.H.; Dey, C.; Rasool, M.K. Targeting inflammatory mediators with ferulic acid, a dietary polyphenol, for the suppression of monosodium urate crystal-induced inflammation in rats. Life Sci. 2016, 148, 201–210. [Google Scholar] [CrossRef]
- Li, Y.H.; Yu, C.Y.; Li, X.X.; Zhang, P.; Tang, J.; Yang, Q.; Fu, T.; Zhang, X.; Cui, X.; Tu, G.; et al. Therapeutic target database update 2018 enriched resource for facilitating bench-to-clinic research of targeted therapeutics. Nucleic Acids Res. 2018, 46, D1121–D1127. [Google Scholar] [CrossRef] [PubMed]
- The Role of Immune Checkpoints in Immunity and Cancer. Available online: https://www.bio-rad-antibodies.com/immune-checkpoint-minireview.html (accessed on 15 December 2016).
- Primeau, A.S.B.; Controlled Amino Acid Therapy and Cancer. Cancer Therapy Advisor. Available online: https://www.cancertherapyadvisor.com/home/tools/fact-sheets/controlled-amino-acid-therapy-and-cancer (accessed on 15 December 2016).
- Li, W.W.; Li, V.W.; Hutnik, M.; Chiou, A.S. Tumor angiogenesis as target for dietary cancer prevention. J. Oncol. 2012, 2012, 879623. [Google Scholar] [CrossRef] [Green Version]
- Huang, Z. Bcl-2 family proteins as targets for anticancer drug design. Oncogene 2000, 19, 6627–6631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhagwat, A.S.; Vakoc, C.R. Targeting transcription factors in cancer. Trends Cancer 2015, 1, 53–65. [Google Scholar] [CrossRef] [Green Version]
- Eckschlager, T.; Plc, J.; Stiborova, M.; Hrabeta, J. Histone deacetylase inhibitors as anticancer drugs. Int. J. Mol. Sci. 2017, 18, 1414. [Google Scholar] [CrossRef]
- Ireson, C.; Orr, S.; Jones, D.J.; Verschoyle, R.; Lim, C.K.; Luo, J.L.; Howells, L.; Plummer, S.; Jukes, R.; Williams, M.; et al. Characterization of metabolites of the chemopreventive agent curcumin in human and rat hepatocytes and in the rat in vivo, and evaluation of their ability to inhibit phorbol ester-induced prostaglandin E2 production. Cancer Res. 2001, 61, 1058–1064. [Google Scholar] [PubMed]
- Nguyen, T.A.; Tang, Q.D.; Doan, D.C.T.; Dang, M.C. Micro and nano liposome vesicles containing curcumin for a drug delivery system. Adv. Nat. Sci. Nanosci. Nanotechnol. 2016, 7, 035003. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Xu, X.; Zhang, Y.; Zhang, Y.; Zhu, Y.; Shi, J.; Sun, Y.; Huang, Q. Encapsulation of curcumin within poly(amidoamine) dendrimers for delivery to cancer cells. J. Mater. Sci. Mater. Med. 2013, 24, 2137–2144. [Google Scholar] [CrossRef] [PubMed]
- Dei Cas, M.; Ghidoni, R. Dietary curcumin: Correlation between bioavailability and health potential. Nutrients 2019, 11, 2147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Teiten, M.H.; Dicato, M.; Diederich, M. Curcumin as a regulator of epigenetic events. Mol. Nutr. Food Res. 2013, 57, 1619–1629. [Google Scholar] [CrossRef] [PubMed]
- Bilia, A.R.; Piazzini, V.; Guccione, C.; Risaliti, L.; Asprea, M.; Capecchi, G.; Bergonzi, M.C. Improving on nature: The role of nanomedicine in the development of clinical natural drugs. Planta Med. 2017, 83, 366–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lestari, M.L.A.D.; Indrayanto, G. Curcumin. In Profiles of Drug Substances, Excipients and Related Methodology; Academic Press: Cambridge, MA, USA, 2014; Volume 39, ISBN 9780128001738. [Google Scholar]
- Yang, M.; Akbar, U.; Mohan, C. Curcumin in autoimmune and rheumatic diseases. Nutrients 2019, 11, 1004. [Google Scholar] [CrossRef] [Green Version]
- Gupta, S.C.; Patchva, S.; Aggarwal, B.B. Therapeutic roles of curcumin: Lessons learned from clinical trials. AAPS J. 2013, 15, 195–218. [Google Scholar] [CrossRef] [Green Version]
- Pivari, F.; Mingione, A.; Brasacchio, C.; Soldati, L. Curcumin and type 2 diabetes mellitus: Prevention and treatment. Nutrients 2019, 11, 1837. [Google Scholar] [CrossRef] [Green Version]
- Chin, K.Y. The spice for joint inflammation: Anti-inflammatory role of curcumin in treating osteoarthritis. Drug Des. Dev. Ther. 2016, 10, 3029–3042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leblhuber, F.; Ehrlich, D.; Steiner, K.; Geisler, S.; Fuchs, D.; Lanser, L.; Kurz, K. The Immunopathogenesis of Alzheimer’s Disease Is Related to the Composition of Gut Microbiota. Nutrients 2021, 13, 361. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.; Clifton, P. Curcumin, cardiometabolic health and dementia. Int. J. Environ. Res. Public Health 2018, 15, 2093. [Google Scholar] [CrossRef] [Green Version]
- Kumar, P.; Barua, C.; Sulakhiya, K.; Kumar Sharma, R. Curcumin Ameliorates Cisplatin-Induced Nephrotoxicity and Potentiates Its Anticancer Activity in SD Rats: Potential Role of Curcumin in Breast Cancer Chemotherapy. Front. Pharm. 2017, 8, 213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nirumand, M.C.; Hajialyani, M.; Rahimi, R.; Farzaei, M.H.; Zingue, S.; Nabavi, S.M.; Bishayee, A. Dietary plants for the prevention and management of kidney stones: Preclinical and clinical evidence and molecular mechanisms. Int. J. Mol. Sci. 2018, 19, 765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jain, K.; Sood, S.; Gowthamarajan, K. Modulation of cerebral malaria by curcumin as an adjunctive therapy. Braz. J. Infect. Dis. 2013, 17, 579–591. [Google Scholar] [CrossRef] [Green Version]
- Bisht, S.; Feldmann, G.; Soni, S.; Ravi, R.; Karikar, C.; Maitra, A. Polymeric nanoparticle-encapsulated curcumin (“nanocurcumin”): A novel strategy for human cancer therapy. J. Nanobiotechnol. 2007, 5, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Narayanan, N.K.; Nargi, D.; Randolph, C.; Narayanan, B.A. Liposome encapsulation of curcumin and resveratrol in combination reduces prostate cancer incidence in PTEN knockout mice. Int. J. Cancer 2009, 125, 1–8. [Google Scholar] [CrossRef]
- Gupta, S.C.; Sung, B.; Kim, J.H.; Prasad, S.; Li, S.; Aggarwal, B.B. Multitargeting by turmeric, the golden spice: From kitchen to clinic. Mol. Nutr. Food Res. 2013, 57, 1510–1528. [Google Scholar] [CrossRef]
- Lapenna, S.; Bilia, A.R.; Morris, G.A.; Nilsson, M. Novel artemisinin and curcumin micellar formulations: Drug solubility studies by NMR spectroscopy. J. Pharm. Sci. 2009, 98, 3666–3675. [Google Scholar] [CrossRef] [PubMed]
- Zielińska, A.; Alves, H.; Marques, V.; Durazzo, A.; Lucarini, M.; Alves, T.F.; Morsink, M.; Willemen, N.; Eder, P.; Chaud, M.V.; et al. Properties, extraction methods, and delivery systems for curcumin as a natural source of beneficial health effects. Medicina 2020, 56, 336. [Google Scholar] [CrossRef] [PubMed]
- Alven, S.; Aderibigbe, B.A. Efficacy of polymer-based nanocarriers for co-delivery of curcumin and selected anticancer drugs. Nanomaterials 2020, 10, 1556. [Google Scholar] [CrossRef]
- Sun, C.-Y.; Zhang, Q.-Y.; Zheng, G.-J.; Feng, B. Phytochemicals: Current strategy to sensitize cancer cells to cisplatin. Biomed. Pharmacother. 2019, 110, 518–527. [Google Scholar] [CrossRef] [PubMed]
- Ponte, L.G.S.; Pavan, I.C.B.; Mancini, M.C.S.; da Silva, L.G.S.; Morelli, A.P.; Severino, M.B.; Bezerra, R.M.N.; Simabuco, F.M. The hallmarks of flavonoids in cancer. Molecules 2021, 26, 2029. [Google Scholar] [CrossRef] [PubMed]
- Mizushina, Y. Specific inhibitors of mammalian DNA polymerase species. Biosci. Biotechnol. Biochem. 2009, 73, 1239–1251. [Google Scholar] [CrossRef]
- Wallace, J.M. Nutritional and botanical modulation of the inflammatory cascade–eicosanoids, cyclooxygenases, and lipoxygenases as an adjunct in cancer therapy. Integr Cancer Ther. 2002, 1, 7–37. [Google Scholar]
- Chakraborti, S.; Das, L.; Kapoor, N.; Das, A.; Dwived, V.; Poddar, A.; Chakraborti, G.; Janik, M.; Basu, G.; Panda, D.; et al. Curcumin recognizes a unique binding site of tubulin. J. Med. Chem. 2011, 54, 6183–6196. [Google Scholar] [CrossRef] [PubMed]
- Li, D.J.; Zhu, M.; Xu, C.; Chen, J.J.; Ji, B.M. The effect of Cu2+ or Fe3+ on the noncovalent binding of rutin with bovine serum albumin by spectroscopic analysis. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2011, 78, 74–79. [Google Scholar] [CrossRef]
- Cao, S.H.; Jiang, X.Y.; Chen, J.W. Effect of Zinc (II) on the interactions of bovine serum albumin with flavonols bearing a different number of hydroxyl substituent on B-ring. J. Inorg. Biochem. 2010, 104, 146–152. [Google Scholar] [CrossRef] [PubMed]
- Mbese, Z.; Khwaza, V.; Aderibigbe, B.A. Curcumin and its derivatives as potential therapeutic agents in prostate, colon and breast cancers. Molecules 2019, 24, 4386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, Z.; Xi, Y. MicroRNAs mediate therapeutic and preventive effects of natural agents in breast cancer. Chin. J. Nat. Med. 2016, 14, 881–887. [Google Scholar] [CrossRef] [Green Version]
- Tilghman, S.L.; Melyssa, L.V.; Bratton, R.; Carriere, P.; Preyan, L.C.; Boue, S.M.; Vasaitis, T.S.; McLachlan, J.A.; Burow, M.E. Phytoalexins, miRNAs and breast cancer: A review of phytochemical-mediated miRNA regulation in breast cancer. J. Health Care Poor Underserved 2013, 24, 36–46. [Google Scholar] [CrossRef]
- Wong, K.E.; Ngai, S.C.; Chan, K.G.; Lee, L.H.; Goh, B.H.; Chuah, L.H. Curcumin Nanoformulations for Colorectal Cancer: A Review. Front. Pharmacol. 2019, 10, 152. [Google Scholar] [CrossRef] [PubMed]
- Zhai, T.; Li, S.; Hu, W.; Li, D.; Leng, S. Potential Micronutrients and Phytochemicals against the Pathogenesis of Chronic Obstructive Pulmonary Disease and Lung Cancer. Nutrients 2018, 10, 813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hong, H.; Ahn, K.S.; Bae, E.; Jeon, S.S.; Choi, H.Y. The effects of curcumin on the invasiveness of prostate cancer in vitro and in vivo. Prostate Cancer Prostatic Dis. 2006, 9, 147–152. [Google Scholar] [CrossRef] [PubMed]
- Rivera, M.; Ramos, Y.; Rodríguez-Valentín, M.; López-Acevedo, S.; Cubano, L.A.; Zou, J.; Zhang, Q.; Wang, G.; Boukli, N.M. Targeting multiple pro-apoptotic signaling pathways with curcumin in prostate cancer cells. PLoS ONE 2017, 12, e0179587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, A.J.; Jiang, G.; Li, L.T.; Zheng, J.N. Curcumin induces apoptosis through mitochondrial pathway and caspases activation in human melanoma cells. Mol. Biol. Rep. 2015, 42, 267–275. [Google Scholar] [CrossRef] [PubMed]
- Siwak, D.R.; Shishodia, S.; Aggarwal, B.B.; Kurzrock, R. Curcumin-induced antiproliferative and proapoptotic effects in melanoma cells are associated with suppression of I kappa B kinase and nuclear factor kappa B activity and are independent of the B-Raf/mitogen-activated/extracellular signal-regulated protein kinase pathway and the Akt pathway. Cancer 2005, 104, 879–890. [Google Scholar]
- Glienke, W.; Maute, L.; Wicht, J.; Bergmann, L. Curcumin inhibits constitutive STAT3 phosphorylation in human pancreatic cancer cell lines and downregulation of survivin/BIRC5 gene expression. Cancer Investig. 2010, 28, 166–171. [Google Scholar] [CrossRef] [PubMed]
- Alexandrow, M.G.; Song, L.J.; Altiok, S.; Gray, J.; Haura, E.B.; Kumar, N.B. Curcumin: A novel Stat3 pathway inhibitor for chemoprevention of lung cancer. Eur. J. Cancer Prev. 2012, 21, 407–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peisch, S.F.; Van Blarigan, E.L.; Chan, J.M.; Stampfer, M.J.; Kenfield, S.A. Prostate cancer progression and mortality: A review of diet and lifestyle factors. World J. Urol. 2017, 35, 867–874. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Shan, M.; Liu, T.; Shi, Q.; Zhong, Z.; Wei, W.; Pang, D. Analysis of TRRAP as a Potential Molecular Marker and Therapeutic Target for Breast Cancer. J. Breast Cancer 2016, 19, 61–67. [Google Scholar] [CrossRef] [PubMed]
- Aruna, S.; Jaiswal, A.S.; Marlow, B.P.; Gupta, N.; Narayan, S. β-Catenin-mediated transactivation and cell–cell adhesion pathways are important in curcumin (diferuylmethane)-induced growth arrest and apoptosis in colon cancer cells. Oncogene 2002, 21, 8414–8427. [Google Scholar]
- Han, S.S.; Chung, S.T.; Robertson, D.A.; Ranjan, D.; Bondada, S. Curcumin causes the growth arrest and apoptosis of B cell lymphoma by downregulation of egr-1, C-myc, Bcl-XL, NF-κB, and p53. Clin. Immunol. 1999, 93, 152–161. [Google Scholar] [CrossRef]
- Shanmugam, M.K.; Arfuso, F.; Sng, J.C.; Bishayee, A.; Kumar, A.P.; Seth, G. Epigenetic effects of curcumin in cancer prevention. Epigenet. Cancer Prev. 2019, 8, 107–128. [Google Scholar]
- Yu, J.; Peng, Y.; Wu, L.-C.; Xie, Z.; Deng, Y.; Hughes, T.; He, S.; Mo, X.K.; Chiu, M.; Wang, Q.E.; et al. Curcumin down-regulates DNA methyltransferase 1 and plays an anti-leukemic role in acute myeloid leukemia. PLoS ONE 2013, 8, e55934. [Google Scholar] [CrossRef] [PubMed]
- Yen, H.; Tsao, C.W.; Lin, J.-W.; Kuo, C.-C.; Tsao, C.H.; Liu, C.-Y. Regulation of carcinogenesis and modulation through Wnt/β-catenin signaling by curcumin in an ovarian cancer cell line. Sci. Rep. 2019, 9, 17267. [Google Scholar] [CrossRef] [Green Version]
- Qiu, Y.; Yu, T.; Wang, W.; Pan, K.; Shi, D.; Sun, H. Curcumin-induced melanoma cell death is associated with mitochondrial permeability transition pore (mPTP) opening. Biochem. Biophys. Res. Commun. 2014, 448, 15–21. [Google Scholar] [CrossRef]
- Koohpar, Z.K.; Entezari, M.; Movafagh, A.; Hashemi, M. Anticancer activity of curcumin on human breast adenocarcinoma: Role of Mcl-1 gene. Iran. J. Cancer Prev. 2015, 8, e2331. [Google Scholar] [CrossRef] [Green Version]
- Medina-Franco, J.L.; López-Vallejo, F.; Kuck, D.; Lyko, F. Natural compounds as DNA methyltransferase inhibitors: A computer-aided discovery approach. Mol. Divers. 2011, 15, 293–304. [Google Scholar] [CrossRef] [PubMed]
- Link, A.; Balaguer, F.; Shen, Y.; Lozano, J.-J.; Leung, H.-C.E.; Boland, C.R.; Goel, A. Curcumin modulates DNA methylation in colorectal cancer cells. PLoS ONE 2013, 8, e57709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shu, L.; Khor, T.O.; Lee, J.H.; Boyanapalli, S.S.; Huang, Y.; Wu, T.Y.; Saw, C.L.; Cheung, K.L.; Kong, A.N. Epigenetic CpG demethylation of the promoter and reactivation of the expression of Neurog1 by curcumin in prostate LNCaP cells. AAPS J. 2011, 13, 606–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, S.; Khor, T.O.; Cheung, K.L.; Li, W.; Wu, T.Y.; Huang, Y.; Foster, B.A.; Kan, Y.W.; Kong, A.N. Nrf2 expression is regulated by epigenetic mechanisms in prostate cancer of TRAMP mice. PLoS ONE 2010, 5, e8579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rajendran, P.; Ho, E.; Williams, D.E.; Dashwood, R.H. Dietary phytochemicals, HDAC inhibition, and DNA damage/repair defects in cancer cells. Clin. Epigenetics 2011, 3, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ito, K.; Ito, M.; Elliott, W.M.; Cosio, B.G.; Caramori, G.; Kon, O.M.; Barczyk, A.; Hayashi, S.; Adcock, I.; Hogg, J.C.; et al. Decreased histone deacetylase activity in chronic obstructive pulmonary disease. N. Engl. J. Med. 2005, 352, 1967–1976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marcu, M.G.; Jung, Y.J.; Lee, S.; Chung, E.J.; Lee, M.J.; Trepel, J.; Neckers, L. Curcumin is an inhibitor of p300 histone acetylatrans-ferase. Med. Chem. 2006, 2, 169–174. [Google Scholar]
- Balasubramanyam, K.; Varier, R.A.; Altaf, M.; Swaminathan, V.; Siddappa, N.B.; Ranga, U.; Kundu, T.K. Curcumin, a novel p300/CREB-binding protein-specific inhibitor of acetyltransferase, represses the acetylation of histone/nonhistone proteins and histone acetyltransferase-dependent chromatin transcription. J. Biol. Chem. 2004, 279, 51163–51171. [Google Scholar] [CrossRef] [Green Version]
- Liu, H.L.; Chen, Y.; Cui, G.H.; Zhou, J.F. Curcumin, a potent anti- tumor reagent, is a novel histone deacetylase inhibitor regulating B- NHL cell line Raji proliferation. Acta Pharmacol. Sin. 2005, 26, 603–609. [Google Scholar] [CrossRef] [Green Version]
- Gao, S.M.; Yang, J.J.; Chen, C.Q.; Chen, J.J.; Ye, L.P.; Wang, L.Y.; Wu, J.B.; Xing, C.Y.; Yu, K. Pure curcumin decreases the expression of WT1 by upregulation of miR-15a and miR-16–1 in leukemic cells. J. Exp. Clin. Cancer Res. 2012, 31, 27. [Google Scholar] [CrossRef] [Green Version]
- Li, W.; Zhou, Y.; Yang, J.; Li, H.; Zhang, H.; Zheng, P. Curcumin induces cell death and protective autophagy in human gastric cancer cells. Oncol. Rep. 2017, 37, 3459–3466. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Y.; Bu, S. Curcumin induces autophagy, apoptosis, and cell cycle arrest in human pancreatic cancer cells. Evid.-Based Complement. Altern. Med. 2017, 2017, 5787218. [Google Scholar] [CrossRef] [PubMed]
- Bhattacharyya, S.; Mandal, D.; Saha, B.; Sen, G.S.; Das, T.; Sa, G. Curcumin prevents tumor-induced T cell apoptosis through Stat-5a-mediated Bcl-2 induction. J. Biol. Chem. 2007, 282, 15954–15964. [Google Scholar] [CrossRef] [Green Version]
- Musial, C.; Siedlecka-Kroplewska, K.; Kmiec, Z.; Gorska-Ponikowska, M. Modulation of autophagy in cancer cells by dietary polyphenols. Antioxidants 2021, 10, 123. [Google Scholar] [CrossRef] [PubMed]
- Rashmi, R.; Kumar, S.; Karunagaran, D. Human colon cancer cells lacking Bax resist curcumin-induced apoptosis and Bax requirement is dispensable with ectopic expression of Smac or downregulation of Bcl-XL. Carcinogenesis 2005, 26, 713–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choi, B.H.; Kim, C.G.; Lim, Y.; Shin, S.Y.; Lee, Y.H. Curcumin down-regulates the multidrug-resistance mdr1b gene by inhibiting the PI3K/Akt/NF kappa B pathway. Cancer Lett. 2008, 259, 111–118. [Google Scholar] [CrossRef] [PubMed]
- Lao, C.D.; Ruffin, M.T.; Normolle, D.; Heath, D.D.; Murray, S.I.; Bailey, J.M.; Boggs, M.E.; Crowell, J.; Rock, C.L.; Brenner, D.E. Dose escalation of a curcuminoid formulation. BMC Complement. Altern. Med. 2006, 6, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, R.A.; Euden, S.A.; Platton, S.L.; Cooke, D.N.; Shafayat, A.; Hewitt, H.R.; Marczylo, T.H.; Morgan, B.; Hemingway, D.; Plummer, S.M. Phase I clinical trial of oral curcumin: Biomarkers of systemic activity and compliance. Clin. Cancer Res. 2004, 10, 6847–6854. [Google Scholar] [CrossRef] [Green Version]
- Jang, M.; Choi, M.S.; Jung, U.J.; Kim, M.J.; Kim, H.J.; Jeon, S.M.; Shin, S.K.; Seong, C.N.; Lee, M.K. Beneficial effects of curcumin on hyperlipidemia and insulin resistance in high-fat-fed hamsters. Metabolism 2008, 57, 1576–1583. [Google Scholar] [CrossRef]
- Reddy, P.H.; Manczak, M.; Yin, X.; Grady, M.C.; Mitchell, A.; Tonk, S.; Kuruva, C.S.; Bhatti, J.S.; Kandimalla, R.; Vijayan, M.; et al. Protective effects of indian spice curcumin against amyloid-beta in Alzheimer’s disease. J. Alzheimers Dis. 2018, 61, 843–866. [Google Scholar] [CrossRef] [PubMed]
- Chan, W.J.; McLachlan, A.J.; Hanrahan, J.R.; Harnett, J.E. The safety and tolerability of Annona muricata leaf extract: A systematic review. J. Pharm. Pharmacol. 2020, 72, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- George, V.C.; Kumar, D.; Rajkumar, V.; Suresh, P.; Kumar, R.A. Quantitative assessment of the relative antineoplastic potential of the n-butanolic leaf extract of Annona Muricata Linn. in normal and immortalized human cell lines. Asia Pac. J. Cancer Prev. 2012, 12, 699–704. [Google Scholar] [CrossRef] [PubMed]
- Moghadamtousi, S.Z.; Fadaeinasab, M.; Nikzad, S.; Mohan, G.; Ali, M.H.; Kadir, H.A. Annona muricata (Annonaceae): A review of its traditional uses, isolated acetogenins and biological activities. Int. J. Mol. Sci. 2015, 16, 15625–15658. [Google Scholar] [CrossRef]
- Mishra, S.; Ahmad, S.; Kumar, N.; Sharma, B.K. Annona muricata (the cancer killer): A review. Glob. J. Pharm. Res. 2013, 2, 1613–1618. [Google Scholar]
- Rady, I.; Bloch, M.B.; Chamcheu, R.-C.N.; Mbeumi, S.B.; Anwar, M.R.; Mohamed, H.; Babatunde, A.S.; Kuiate, J.-R.; Noubissi, F.-K.; El Sayed, K.A.; et al. Anticancer Properties of Graviola (Annona muricata): A Comprehensive Mechanistic Review. Oxid. Med. Cell Longev. 2018, 2018, 1826170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eposti, M.; Gheli, A.; Ratta, M.; Cortes, D.; Estomell, E. Natural substances (acetogenins) from the family Annonaceae are powerful inhibitors of mitochondrial NADH dehydrogenase (Complex I). J. Biochem. 1994, 301, 161–167. [Google Scholar]
- Orak, H.H.; Bahrisefit, I.S.; Sabudak, T. Antioxidant activity of extracts of soursop (Annona muricata L.) leaves, fruit pulps, peels, and seeds. Pol. J. Food Nutr. Sci. 2019, 69, 359–366. [Google Scholar] [CrossRef]
- Moghadamtousi, S.Z.; Karimian, H.; Rouhollahi, E.; Paydar, M.; Fadaeinasab, M.; Kadir, H.A. Annona muricata leaves induce G1 cell cycle arrest and apoptosis through mitochondria-mediated pathway in human HCT-116 and HT-29 colon cancer cells. J. Ethnopharmacol. 2014, 156, 277–289. [Google Scholar] [CrossRef]
- Deep, G.; Kumar, R.; Jain, A.K.; Dhar, D.; Panigrahi, G.K.; Hussain, A.; Agarwal, C.; El-Elimat, T.; Sica, V.P.; Oberlies, N.H.; et al. Graviola inhibits hypoxia-induced NADPH oxidase activity in prostate cancer cells reducing their proliferation and clonogenicity. Sci. Rep. 2016, 10, 23135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lannuzel, A.; Michel, P.P.; Höglinger, G.U.; Champy, P.; Jousset, A.; Medja, F.; Lombès, A.; Darios, F.; Gleye, C.; Laurens, A.; et al. The mitochondrial complex I inhibitor annonacin is toxic to mesencephalic dopaminergic neurons by impairment of energy metabolism. Neuroscience 2003, 121, 287–296. [Google Scholar] [CrossRef]
- Torres, M.P.; Rachagani, S.; Purohit, V.; Pandey, P.; Joshi, S.; Moore, E.D.; Johansson, S.L.; Singh, P.K.; Ganti, A.K.; Batra, S.K. Graviola: A novel promising natural-derived drug that inhibit tumorigeneicity and metastasis of pancreatic cancer cells in vitro and in vivo through altering cell metabolism. Cancer Lett. 2012, 323, 29–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Höllerhage, M.; Rösler, T.W.; Berjas, M.; Luo, R.; Tran, K.; Richards, K.M.; Sabaa-Srur, A.U.; Maia, J.G.S.; De Moraes, M.R.; Godoy, H.T.; et al. Neurotoxicity of dietary supplements from Annonaceae species. Int. J. Toxicol. 2015, 34, 543–550. [Google Scholar] [CrossRef] [Green Version]
- Luo, R.R.; Maia, J.G.; Moraes, M.R.; Godoy, H.T.; Sabaa-Srur, A.; Tran, K.; Richards, K.M.; Monroe, D.M.; Vocque, R.H.; Smith, R.E. NMR Analysis of Potentially Neurotoxic Annonaceous Fruits. Nat. Prod. J. 2013, 3, 230–241. [Google Scholar]
- Champy, P.; Höglinger, G.U.; Féger, J.; Gleye, C.; Hocquemiller, R.; Laurens, A.; Guérineau, V.; Laprévote, O.; Medja, F.; Lombès, A.; et al. Annonacin, a lipophilic inhibitor of mitochondrial complex I, induces nigral and striatal neurodegeneration in rats: Possible relevance for atypical parkinsonism in Guadeloupe. J. Neurochem. 2004, 88, 63–69. [Google Scholar] [CrossRef] [PubMed]
- Bonnefont-Rousselot, D. Resveratrol and Cardiovascular Diseases. Nutrients 2016, 8, 250. [Google Scholar] [CrossRef]
- Farghali, H.; Kutinová Canová, N.; Lekić, N. Resveratrol and related compounds as antioxidants with an allosteric mechanism of action in epigenetic drug targets. Physiol. Res. 2013, 62, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Kundu, J.K.; Shin, Y.K.; Kim, S.H.; Surh, Y.J. Resveratrol inhibits phorbol ester-induced expression of COX-2 and activation of NF-kappaB in mouse skin by blocking IkappaB kinase activity. Carcinogenesis 2006, 27, 1465–1474. [Google Scholar] [CrossRef] [PubMed]
- Ren, Z.; Wang, L.; Cui, J.; Huoc, Z.; Xue, J.; Cui, H.; Mao, Q.; Yang, R. Resveratrol inhibits NF-κB signaling through suppression of p65 and IkappaB kinase activities. Pharmazie 2013, 68, 689–694. [Google Scholar] [PubMed]
- Mitani, T.; Harada, N.; Tanimori, S.; Nakano, Y.; Inui, H.; Yamaji, R. Resveratrol inhibits hypoxia-inducible factor-1α-mediated androgen receptor signaling and represses tumor progression in cas-tration-resistant prostate cancer. J. Nutr. Sci. Vitaminol. 2014, 60, 276–282. [Google Scholar] [CrossRef] [Green Version]
- Mitani, T.; Ito, Y.; Harada, N.; Nakano, Y.; Inui, H.; Ashida, H.; Yamaji, R. Resveratrol reduces the hypoxia-induced resistance to doxorubicin in breast cancer cells. J. Nutr. Sci. Vitaminol. 2014, 60, 122–128. [Google Scholar] [CrossRef] [Green Version]
- Bishayee, A.; Petit, D.M.; Samtani, K. Angioprevention is implicated in resveratrol chemoprevention of experimental hepatocarcinogenesis. J. Carcinog. Mutagen. 2010, 1, 102. [Google Scholar] [CrossRef]
- Muqbil, I.; Beck, F.W.; Bao, B.; Sarkar, F.H.; Mohammad, R.M.; Hadi, S.M.; Azmi, A.S. Old wine in a new bottle: The Warburg effect and anticancer mechanisms of resveratrol. Curr. Pharm. Des. 2012, 18, 1645–1654. [Google Scholar] [CrossRef] [PubMed]
- Shankar, S.; Nall, D.; Tang, S.N.; Meeker, D.; Passarini, J.; Sharma, J.; Srivastava, R.K. Resveratrol inhibits pancreatic cancer stem cell characteristics in human and KrasG12D transgenic mice by inhibiting pluripotency maintaining factors and epithelial-mesenchymal transition. PLoS ONE 2010, 6, e16530. [Google Scholar] [CrossRef] [Green Version]
- Follo-Martinez, A.; Banerjee, N.; Li, X.; Safe, S.; Mertens-Talcott, S. Resveratrol and quercetin in combination have anticancer activity in colon cancer cells and repress oncogenic microRNA-27a. Nutr. Cancer 2013, 65, 494–504. [Google Scholar] [CrossRef] [PubMed]
- Kumar, A.; Rimando, A.M.; Levenson, A.S. Resveratrol and pterostilbene as a microRNA-mediated chemopreventive and therapeutic strategy in prostate cancer. Ann. N. Y. Acad. Sci. 2017, 1043, 15–26. [Google Scholar] [CrossRef] [PubMed]
- Wu, F.; Cui, L. Resveratrol suppresses melanoma by inhibiting NF-κB/miR-221 and inducing TFG expression. Arch. Dermatol. Res. 2017, 309, 823–831. [Google Scholar] [CrossRef] [PubMed]
- Venkatadri, R.; Muni, T.; Iyer, A.K.V.; Yakisich, J.S.; Azad, N. Role of apoptosis-related miRNAs in resveratrol-induced breast cancer cell death. Cell Death Dis. 2018, 7, e2104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dhar, S.; Kumar, A.; Li, K.; Tzivion, G.; Levenson, A.S. Resveratrol regulates PTEN/Akt pathway through inhibition ofMTA1/HDAC unit of the NuRD complex in prostate cancer. Biochim. Biophys. Acta 2015, 1853, 265–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Daitoku, H.; Sakamaki, J.; Fukamizu, A. Regulation of FoxO transcription factors by acetylation andprotein–protein interactions. Biochim. Biophys. Acta 2011, 1813, 1954–1960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sunita, B.; Jones, S.B.; De Primo, S.E.; Whitfield, M.; Brooks, J. Resveratrol-induced gene expression profiles in human prostate cancer cells. Cancer Epidemiol. Biomark. Prev. 2005, 14, 596–604. [Google Scholar]
- Farhan, M.; Silva, M.; Xingan, X.; Huang, Y.; Zheng, W. Role of FoxO transcription factors in cancer metabolism and angiogenesis. Cells 2020, 9, 1586. [Google Scholar] [CrossRef] [PubMed]
- Tili, E.; Michaille, J.J.; Alder, H.; Volinia, S.; Delmas, D.; Latruffe, N.; Croce, C.M. Resveratrol modulates the levels of microRNAs targeting genes encoding tumor-suppressors and effectors of TGFβ signaling pathway in SW480 cells. Biochem. Pharmacol. 2010, 80, 2057–2065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, G.; Dai, F.; Yu, K.; Jia, Z.; Zhang, A.; Huang, Q.; Kang, C.; Jiang, H.; Pu, P. Resveratrol inhibits glioma cell growth via targeting oncogenic microRNAs and multiple signaling pathways. Int. J. Oncol. 2015, 46, 1739–1747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, H.; Jia, Z.; Li, A.; Jenkins, G.; Yang, X.; Hu, J.; Guo, W. Resveratrol repressed viability of U251 cells by miR-21 inhibiting of NF-κB pathway. Mol. Cell. Biochem. 2013, 382, 137–143. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Tomé-Carneiro, J.; Gonzálvez, M.; Larrosa, M.; Yáñez-Gascón, M.J.; García-Almagro, F.J.; Ruiz-Ros, J.A.; Tomás-Barberán, F.A.; García-Conesa, M.T.; Espín, J.C. Grape resveratrol increases serum adiponectin and downregulates inflammatory genes in peripheral blood mononuclear cells: A triple-blind, placebo-controlled, one-year clinical trial in patients with stable coronary artery disease. Cardiovasc. Drugs Ther. 2013, 27, 37–48. [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]
- Bode, L.M.; Bunzel, D.; Huch, M.; Cho, G.S.; Ruhland, D.; Bunzel, M.; Bub, A.; Franz, C.M.; Kulling, S.E. In vivo and in vitro metabolism of trans-resveratrol by human gut microbiota. Am. J. Clin. Nutr. 2013, 97, 295–309. [Google Scholar] [CrossRef]
- Bastianetto, S.; Ménard, C.; Quirion, R. Neuroprotective action of resveratrol. Biochim. Biophys. Acta 2015, 1852, 1195–1201. [Google Scholar] [CrossRef] [Green Version]
- Baur, J.A.; Pearson, K.J.; Price, N.L.; Jamieson, H.A.; Lerin, C.; Kalra, A.; Prabhu, V.V.; Allard, J.S.; Lopez Lluch, G.; Lewis, K.; et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature 2006, 444, 337–342. [Google Scholar] [CrossRef]
- Wilson, T.; Knight, T.J.; Beitz, D.C.; Lewis, D.S.; Engen, R.L. Resveratrol promotes atherosclerosis in hypercholesterolemic rabbits. Life Sci. 1996, 59, PL15–PL21. [Google Scholar] [CrossRef]
- Ferry-Dumazet, H.; Garnier, O.; Mamani-Matsuda, M.; Vercauteren, J.; Belloc, F.; Billiard, C.; Dupouy, M.; Thiolat, D.; Kolb, J.P.; Marit, G.; et al. Resveratrol inhibits the growth and induces the apoptosis of both normal and leukemic hematopoietic cells. Carcinogenesis 2002, 23, 1327–1333. [Google Scholar] [CrossRef] [PubMed]
- Min, K.J.; Kwon, T.K. Anticancer effects and molecular mechanisms of epigallocatechin-3-gallate. Integr. Med. Res. 2014, 3, 16–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, G.Y.; Liao, J.; Li, C.J.; Yurkow, E.J.; Ho, C.T.; Yang, C.S. Effect of black and green tea polyphenols on c-jun phosphorylation and H2O2 production in transformed and non-transformed human bronchial cell lines: Possible mechanisms of cell growth inhibition and apoptosis induction. Carcinogenesis 2000, 21, 2035–2039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, B.N.; Shankar, S.; Srivastava, R.K. Green tea catechin, epigallocatechin-3-gallate (EGCG): Mechanisms, perspectives and clinical applications. Biochem Pharmacol. 2011, 82, 1807–1821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, L.; Xie, J.; Gan, R.; Wu, Z.; Luo, H.; Chen, X.; Lu, Y.; Wu, L.; Zheng, D. Synergistic inhibition of lung cancer cells by EGCG and NF-κB inhibitor BAY11-7082. J. Cancer 2019, 10, 6543–6556. [Google Scholar] [CrossRef]
- Rao, S.D.; Pagidas, K. Epigallocatechin-3-gallate, a natural polyphenol, inhibits cell proliferation and induces apoptosis in human ovarian cancer cells. Anticancer Res. 2010, 30, 2519–2523. [Google Scholar] [PubMed]
- Zhang, G.; Wang, Y.; Zhang, Y.; Wan, X.; Li, J.; Liu, K.; Wang, F.; Liu, K.; Liu, Q.; Yang, C.; et al. Anti-cancer activities of tea epigallocatechin-3-gallate in breast cancer patients under radiotherapy. Curr. Mol. Med. 2012, 12, 163–176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Ren, X.; Deng, C.; Yang, L.; Yan, E.; Guo, T.; Li, Y.; Xu, M.X. Mechanism of the inhibition of the STAT3 signaling pathway by EGCG. Oncol. Rep. 2013, 30, 2691–2696. [Google Scholar] [CrossRef] [PubMed]
- Tang, S.N.; Fu, J.; Shankar, S.; Srivastava, R.K. EGCG enhances the therapeutic potential of gemcitabine and CP690550 by inhibiting STAT3 signaling pathway in human pancreatic cancer. PLoS ONE 2012, 7, e31067. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Bian, S.; Yang, C.S. Green tea polyphenol EGCG suppresses lung cancer cell growth through upregulating miR-210 expression caused by stabilizing HIF-1α. Carcinogenesis 2011, 32, 1881–1889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Larsen, C.A.; Dashwood, R.H. (−)-Epigallocatechin-3-gallate inhibits Met signaling, proliferation, and invasiveness in human colon cancer cells. Arch. Biochem. Biophys. 2010, 501, 52–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chu, C.; Deng, J.; Man, Y.; Qu, Y. Green tea extracts epigallocatechin-3-gallate for different treatments. BioMed Res. Int. 2017, 2017, 5615647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yiannakopoulou, E.C. Targeting DNA methylation with green tea catechins. Pharmacology 2015, 95, 111–116. [Google Scholar] [CrossRef]
- Fang, M.Z.; Wang, Y.; Ai, N.; Hou, Z.; Sun, Y.; Lu, H.; Welsh, W.; Yang, C.S. Tea polyphenol (-)-epigallocatechin-3-gallate inhibits DNA methyltransferase and reactivates methylation-silenced genes in cancer cell lines. Cancer Res. 2003, 63, 7563–7570. [Google Scholar] [PubMed]
- Gilbert, E.R.; Liu, D. Flavonoids influence epigenetic-modifying enzyme activity: Structure-function relationships and the therapeutic potential for cancer. Curr. Med. Chem. 2010, 17, 1756–1768. [Google Scholar] [CrossRef] [PubMed]
- Lee, W.J.; Shim, J.Y.; Zhu, B.T. Mechanisms for the inhibition of DNA methyltransferases by tea catechins and bioflavonoids. Mol. Pharmacol. 2005, 68, 1018–1030. [Google Scholar] [CrossRef] [Green Version]
- Katiyar, S.K.; Singh, T.; Prasad, R.; Sun, Q.; Vaid, M. Epigenetic alterations in ultraviolet radiation-induced skin carcinogenesis: Interaction of bioactive dietary components on epigenetic targets. Photochem. Photobiol. 2012, 88, 1066–1074. [Google Scholar] [CrossRef] [PubMed]
- Lu, Q.Y.; Jin, Y.S.; Zhang, Z.F.; Le, A.D.; Heber, D.; Li, F.P.; Dubinett, S.M.; Rao, J.Y. Green tea induces annexin-I expression in human lung adenocarcinoma A549 cells: Involvement of an- nexin-I in actin remodeling. Lab. Investig. 2007, 87, 456–465. [Google Scholar] [CrossRef] [Green Version]
- Xiao, G.S.; Jin, Y.S.; Lu, Q.Y.; Zhang, Z.F.; Belldegrun, A.; Figlin, R.; Pantuck, A.; Yen, Y.; Li, F.; Rao, J. Annexin-I as a potential target for green tea extract induced actin remodeling. Int. J. Cancer 2007, 120, 111–120. [Google Scholar] [CrossRef] [PubMed]
- Gao, Z.; Xu, Z.; Hung, M.S.; Lin, Y.C.; Wang, T.; Gong, M.; Zhi, X.; Jablon, D.M.; You, L. Promoter demethylation of WIF-1 by epigallocatechin-3-gallate in lung cancer cells. Anticancer Res. 2009, 29, 2025–2030. [Google Scholar] [PubMed]
- Choi, K.C.; Jung, M.G.; Lee, Y.H.; Yoon, J.C.; Kwon, S.H.; Kang, H.B.; Kim, M.J.; Jun, W.J.; Lee, M.J.; Yoon, H.G. Epigallocatechin-3-gallate, a histone acetyltransferase inhibitor, inhibits EBV-induced B lymphocyte transformation via suppression of RelA acetylation. Cancer Res. 2009, 69, 583–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Soliman, H.; Mediavilla-Varela, M.; Antonia, S. Indoleamine 2,3-Dioxygenase. Is It an Immune Suppressor? Cancer J. 2010, 16, 10. [Google Scholar] [CrossRef] [Green Version]
- 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] [PubMed] [Green Version]
- Mándi, Y.; Vécsei, L. The kynurenine system and immunoregulation. J. Neural. Transm. 2012, 119, 197–209. [Google Scholar] [CrossRef] [PubMed]
- Ogawa, K.; Hara, T.; Shimizu, M.; Nagano, J.; Ohno, T.; Hoshi, M.; Ito, H.; Tsurumi, H.; Saito, K.; Seishima, M.; et al. (-)-Epigallocatechin gallate inhibits the expression of indoleamine 2,3-dioxygenase in human colorectal cancer cells. Oncol. Lett. 2012, 4, 546–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mor, A.; Tankiewicz-Kwedlo, A.; Pawlak, D. Kynurenines as a Novel Target for the treatment. Pharmaceuticals 2021, 14, 606. [Google Scholar] [CrossRef] [PubMed]
- Mellor, A.L.; Munn, D.H. IDO expression by dendritic cells: Tolerance and tryptophan catabolism. Nat. Rev. Immunol. 2004, 4, 762–774. [Google Scholar] [CrossRef] [PubMed]
- Rawangkan, A.; Wongsirisin, P.; Namiki, K.; Iida, K.; Kobayashi, Y.; Shimizu, Y.; Hirota Fujiki, H.; Suganuma, M. Green Tea Catechin Is an Alternative Immune Checkpoint Inhibitor that Inhibits PD-L1 Expression and Lung Tumor Growth. Molecules 2018, 23, 2071. [Google Scholar] [CrossRef] [Green Version]
- Kim, S.N.; Kwon, H.-J.; Akindehin, S.; Jeong, H.W. Effects of epigallocatechin-3-gallate on autophagic lipolysis in adipocytes. Nutrients 2017, 9, 680. [Google Scholar] [CrossRef] [Green Version]
- Shanafelt, T.D.; Call, T.G.; Zent, C.S.; La Plant, B.; Bowen, D.A.; Roos, M.; Secreto, C.R.; Ghosh, A.K.; Kabat, B.F.; Lee, M.-J.; et al. Phase I trial of daily oral Polyphenon E in patients with asymptomatic Rai stage 0 to II chronic lymphocytic leukemia. J. Clin. Oncol. 2009, 27, 3808–3814. [Google Scholar] [CrossRef]
- Saakre, M.; Mathew, D.; Ravisankar, V. Perspectives on plant flavonoid quercetin-based drugs for novel SARS-CoV-2. Beni-Suef Univ. J. Basic Appl. Sci. 2021, 10, 21. [Google Scholar] [CrossRef] [PubMed]
- Maalik, A.; Khan, F.A.; Mumtaz, A.; Mehmood, A.; Azhar, S.; Atif, M.; Karim, S.; Altaf, Y.; Tariq, I.; Trop, J. Pharmacological applications of quercetin and its derivatives: A short review. Trop. J. Pharm. Res. 2014, 13, 1561–1566. [Google Scholar] [CrossRef]
- Jafarinia, M.; Sadat Hosseini, M.; Kasiri, N.; Fazel, N.; Fathi, F.; Ganjalikhani Hakemi, M.; Eskandari, N. Quercetin with the potential effect on allergic diseases. Allergy Asthma Clin. Immunol. 2020, 16, 36. [Google Scholar] [CrossRef] [PubMed]
- Granato, M.; Rizello, C.; Montani, M.S.G.; Cuomo, L.; Vitillo, M.; Santarelli, R.; Gonella, R.; D’Orazi, G.; Faggioni, A.; Cirone, M. Quercetin induces apoptosis and autophagy in primary effusion lymphoma cells by inhibiting PI3K/AKT/mTOR and STAT3 signaling pathways. J. Nutr. Biochem. 2017, 41, 124–136. [Google Scholar] [CrossRef]
- Costa, L.G.; Garrick, J.M.; Roquè, P.J.; Pellacani, C. Mechanisms of neuroprotection by quercetin: Counteracting oxidative stress and more. Oxidative Med. Cell. Longev. 2016, 2016, 2986796. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Yao, J.; Han, C.; Yang, J.; Chaudhry, M.T.; Wang, S.; Liu, H.; Yin, Y. Quercetin, inflammation and immunity. Nutrients 2016, 8, 167. [Google Scholar] [CrossRef] [PubMed]
- Khan, H.; Ullah, H.; Aschner, M.; Cheang, W.S.; Akkol, E.K. Neuroprotective effects of quercetin in Alzheimer’s disease. Biomolecules 2019, 10, 59. [Google Scholar] [CrossRef] [Green Version]
- Solimani, R. The flavonols quercetin, rutin and morin in DNA solution: UV–vis dichroic (and mid-infrared) analysis explain the possible association between the biopolymer and a nucleophilic vegetable-dye. BBA 1997, 1336, 281–294. [Google Scholar] [CrossRef]
- Scanlon, S.E.; Scanlon, C.D.; Hegan, D.C.; Sulkowski, P.L.; Glazer, P.M. Nickel induces transcriptional down-regulation of DNA repair pathways in tumorigenic and non-tumorigenic lung cells. Carcinogenesis 2017, 38, 627–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ravichandran, R.; Rajendran, M.; Devapiriam, D. Antioxidant study of quercetin and their metal complex and determination of stability constant by spectrophotometry method. Food Chem. 2014, 146, 472–478. [Google Scholar] [CrossRef] [PubMed]
- Vaish, S.; Gupta, D.; Mehrotra, R.; Mehrotra, S.; Kumar Basantani, M. Glutathione S-transferase: A versatile protein family. Biotech 2020, 10, 321. [Google Scholar] [CrossRef]
- Kubczak, M.; Szustka, A.; Błoński, J.Z.; Gucký, T.; Misiewicz, M.; Krystof, V.; Robak, P.; Rogalińska, M. Dose and drug changes in chronic lymphocytic leukemia cell response in vitro: A comparison of standard therapy regimens with two novel cyclin-dependent kinase inhibitors. Mol. Med. Rep. 2019, 19, 3593–3603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, S.; Gao, N.; Zhang, Z.; Chen, G.; Budhraja, A.; Ke, Z.; Son, Y.; Wang, X.; Luo, J.; Shi, X. Quercetin Induces Tumor-Selective Apoptosis through Downregulation of Mcl-1 and Activation of Bax. Clin. Cancer Res. 2010, 16, 5679–5691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nguyen, L.T.; Lee, Y.H.; Sharma, A.R.; Park, J.B.; Jagga, B.; Sharma, G.; Lee, S.-S.; Nam, J.-S. Quercetin induces apoptosis and cell cycle arrest in triple-negative breast cancer cells through modulation of Foxo3a activity. Korean J. Physiol. Pharmacol. 2017, 21, 205–213. [Google Scholar] [CrossRef] [Green Version]
- Seo, H.S.; Ku, J.M.; Choi, H.-S.; Choi, Y.J.; Woo, J.-K.; Kim, M.; Kim, I.; Na, C.H.; Hur, H.; Jang, B.-H.; et al. Quercetin induces caspase-dependent extrinsic apoptosis through inhibition of signal transducer and activator of transcription 3 signaling in HER2-overexpressing BT-474 breast cancer cells. Oncol. Rep. 2016, 36, 31–42. [Google Scholar] [CrossRef] [Green Version]
- Yang, A.Y.; Kim, H.; Li, W.; Kong, A.N. Natural compound-derived epigenetic regulators targeting epigenetic readers, writers and erasers. Curr. Top. Med. Chem. 2016, 16, 697–713. [Google Scholar] [CrossRef]
- Xiao, X.; Shi, D.; Liu, L.; Wang, J.; Xie, X.; Kang, T.; Deng, W. Quercetin suppresses cyclooxygenase-2 expression and angiogenesis through inactivation of P300 signaling. PLoS ONE 2011, 6, e22934. [Google Scholar] [CrossRef]
- Lee, W.J.; Chen, Y.R.; Chen, Y.-R.; Tseng, B. Quercetin induces FasL-related apoptosis, in part, through promotion of histone H3 acetylation in human leukemia HL-60 cells. Oncol. Rep. 2011, 25, 583–591. [Google Scholar]
- Chen, T.; Zhang, X.; Zhu, G.; Liu, H.; Chen, J.; He, X. Quercetin inhibits TNF-α induced HUVECs apoptosis and inflammation via downregulating NF-kB and AP-1 signaling pathway in vitro. Medicine 2020, 99, e22241. [Google Scholar] [CrossRef] [PubMed]
- Kabir, T.; Tabassum, N.; Uddin, S.; Aziz, F.; Behl, T.; Mathew, B.; Rahman, H.; Akter, R.; Rauf, A.; Aleya, L. Therapeutic Potential of Polyphenols in the Management of Diabetic Neuropathy. Evid.-Based Complementary Altern. Med. 2021, 13, 9940169. [Google Scholar] [CrossRef]
- Kleemann, R.; Verschuren, L.; Morrison, M.; Zadelaar, S.; van Erk, M.J.; Wielinga, P.Y.; Kooistra, T. Anti-inflammatory, anti-proliferative and anti-atherosclerotic effects of quercetin in human in vitro and in vivo models. Atherosclerosis 2011, 218, 44–52. [Google Scholar] [CrossRef] [PubMed]
- Mu, C.; Jia, P.; Yan, Z.; Liu, X.; Li, X.; Liu, H. Quercetin induces cell cycle G1 arrest through elevating Cdk inhibitors p21 and p27 in human hepatoma cell line (HepG2). Methods Find Exp. Clin. Pharm. 2007, 29, 179–183. [Google Scholar] [CrossRef]
- Lai, W.W.; Hsu, S.C.; Chueh, F.S.; Chen, Y.-Y.; Yang, J.-S.; Lin, J.-P.; Lien, J.-C.; Tsai, C.-H.; Chung, J.-G. Quercetin inhibits migration and invasion of SAS human oral cancer cells through inhibition of NF-κB and matrix metalloproteinase-2/-9 signaling pathways. Anticancer Res. 2013, 33, 1941–1950. [Google Scholar] [PubMed]
- Tao, S.F.; He, H.F.; Chen, Q. Quercetin inhibits proliferation and invasion acts by up-regulating miR-146a in human breast cancer cells. Mol. Cell Biochem. 2015, 402, 93–100. [Google Scholar] [CrossRef]
- Zhijun, H.; Jie, C.; Yi, W.; Parhati, A.; Xiuli, Y.; Haitao, D. Quercetin suppresses proliferation and motility through modulating hippo pathway via upregulating mir-146a-5p in gastric cancer. J. Biomater. Tissue Eng. 2019, 9, 82–88. [Google Scholar]
- Uçar, E.Ö.; Şengelen, A.; Mertoğlu, E.; Pekmez, M.; Arda, N. Suppression of HSP70 expression by quercetin and its therapeutic potential against cancer. In Heat Shock Proteins; Springer: Basel, Switzerland, 2018; pp. 361–379. [Google Scholar]
- Zhong, Y.; Liao, J.; Hu, Y.; Wang, Y.; Sun, C.; Zhang, C.; Wang, G. PM2.5 upregulates microRNA-146a-3p and induces M1 polarization in RAW264.7 cells by targeting Sirtuin1. Int. J. Med. Sci. 2019, 16, 384–393. [Google Scholar] [CrossRef] [Green Version]
- Matos, H.R.; di Mascio, P.; Medeiros, M.H.G. Protective effect of lycopene on lipid peroxidation and oxidative DNA damage in cell culture. Arch. Biochem. Biophys. 2000, 383, 56–59. [Google Scholar] [CrossRef]
- Rizwan, M.; Rodriguez-Blanco, I.; Harbottle, A.; Birch Machin, M.A.; Watson, R.E.B.; Rhodes, L.E. Tomato paste rich in lycopene protects against cutaneous photodamage in humans in vivo: A randomized controlled trial. Br. J. Dermatol. 2011, 164, 154–162. [Google Scholar] [CrossRef] [PubMed]
- Ito, Y.; Kurata, M.; Hioki, R.; Suzuki, K.; Ochiai, J.; Aoki, K. Cancer mortality and serum levels of carotenoids, retinol, and tocopherol: A population-based follow-up study of inhabitants of a rural area of Japan. Asian Pac. J. Cancer Prev. 2005, 6, 10–15. [Google Scholar] [PubMed]
- Puah, B.-P.; Jalil, J.; Ali Attiq, A.; Kamisah, Y. New Insights into Molecular Mechanism behind Anti-Cancer Activities of Lycopene. Molecules 2021, 26, 3888. [Google Scholar] [CrossRef]
- Ben-Dor, A.; Steiner, M.; Gheberetal, L. Carotenoids activate the antioxidant response element transcription system. Mol. Cancer Ther. 2005, 4, 177–186. [Google Scholar]
- Lian, F.; Wang, X.-D. Enzymatic metabolites of lycopene induce Nrf2-mediated expression of phase II detoxifying/antioxidant enzymes in human bronchial epithelial cells. Int. J. Cancer 2008, 123, 1262–1268. [Google Scholar] [CrossRef] [Green Version]
- Itoh, K.; Tong, K.I.; Yamamoto, M. Molecular mechanism activating Nrf2-Keap1 pathway in regulation of adaptive response to electrophiles. Free Radic. Biol. Med. 2004, 36, 1208–1213. [Google Scholar] [CrossRef] [PubMed]
- Giovannucci, E. Insulin-like growth factor-I and binding protein-3 and risk of cancer. Insulin-like growth factor-I and binding protein-3 and risk of cancer. Horm. Res. 1999, 51, 34–41. [Google Scholar] [PubMed]
- Vrieling, A.; Voskuil, D.W.; Bonfrer, J.M.; Korse, C.M.; van Doorn, J.; Cats, A.; Depla, A.C.; Timmer, R.; Witteman, B.J.; van Leeuwen, F.E.; et al. Lycopene supplementation elevates circulating insulin-like growth factor binding protein-1 and -2 concentrations in persons at greater risk of colorectal cancer. Am. J. Clin. Nutr. 2007, 86, 1456–1462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, F.Y.; Cho, H.J.; Pai, M.H.; Chen, Y.H. Concomitant supplementation of lycopene and eicosapentaenoic acid inhibits the proliferation of human colon cancer cells. J. Nutr. Biochem. 2009, 20, 426–434. [Google Scholar] [CrossRef]
- Tang, Y.; Parmakhtiar, B.; Simoneau, A.R.; Xie, J.; Fruehauf, J.; Lilly, M.; Zi, X. Lycopene enhances docetaxel’s effect in castration-resistant prostate cancer associated with insulin-like growth factor I receptor levels. Neoplasia 2011, 13, 108–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, Y.O.; Hwang, E.-S.; Moon, T.W. The effect of lycopene on cell growth and oxidative DNA damage of Hep3B human hepatoma cells. Biofactors 2005, 23, 129–139. [Google Scholar] [CrossRef]
- Nahum, A.; Hirsch, K.; Danilenko, M.; Watts, C.K.; Prall, O.W.; Levy, J.; Sharoni, Y. Lycopene inhibition of cell cycle progression in breast and endometrial cancer cells is associated with reduction in cyclin D levels and retention of p27 Kip1 in the cyclin E-cdk2 complexes. Oncogene 2001, 20, 3428–3436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karas, M.; Amir, H.; Fishman, D.; Danilenko, M.; Segal, S.; Nahum, A.; Koifmann, A.; Giat, Y.; Levy, J.; Sharoni, Y. Lycopene interferes with cell cycle progression and insulin-like growth factor I signaling in mammary cancer cells. Nutr. Cancer 2000, 36, 101–111. [Google Scholar] [CrossRef]
- Palozza, P.; Colangelo, M.; Simone, R.; Catalano, A.; Boninsegna, A.; Lanza, P.; Monego, G.; Ranelletti, F.O. Lycopene induces cell growth inhibition by altering mevalonate pathway and Ras signaling in cancer cell lines. Carcinogenesis 2010, 31, 1813–1821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, F.Y.; Shih, C.J.; Cheng, L.H.; Ho, H.-J.; Chen, H.J. Lycopene inhibits growth of human colon cancer cells via suppression of the Akt signaling pathway. Mol. Nutr. Food Res. 2008, 52, 646–654. [Google Scholar] [CrossRef] [PubMed]
- Salman, H.; Bergman, M.; Djaldetti, M.; Bessler, H. Lycopene affects proliferation and apoptosis of four malignant cell lines. Biomed. Pharm. 2007, 61, 366–369. [Google Scholar] [CrossRef]
- Maru, G.B.; Hudlikar, R.R.; Kumar, G.; Gandhi, K.; Mahimka, M.B. Understanding the molecular mechanisms of cancer prevention by dietary phytochemicals: From experimental models to clinical trial. World J. Biol. Chem. 2016, 7, 88–99. [Google Scholar] [CrossRef] [PubMed]
- Velmurugan, B.; Mani, A.; Nagini, S. Combination of S allylcysteine and lycopene induces apoptosis by modulating Bcl-2, Bax, Bim and caspases during experimental gastric carcinogenesis. Eur. J. Cancer Prev. 2005, 4, 387–393. [Google Scholar] [CrossRef]
- Hantz, H.L.; Young, L.F.; Martin, K.R. Physiologically attainable concentrations of lycopene induce mitochondrial apoptosis in LNCaP human prostate cancer cells. Exp. Biol. Med. 2005, 230, 171–179. [Google Scholar] [CrossRef]
- Sahin, K.; Tuzcu, M.; Sahin, N.; Akdemir, F.; Ozercan, I.; Bayraktar, S.; Kucuk, O. Inhibitory effects of combination of lycopene and genistein on 7,12-Dimethyl benz(a)anthracene-induced breast cancer in rats. Nutr. Cancer 2011, 63, 1279–1286. [Google Scholar] [CrossRef] [PubMed]
- Feng, D.; Ling, W.-H.; Duan, R.-D. Lycopene suppresses LPS-induced NO and IL-6 production by inhibiting the activation of ERK, p38MAPK, and NF-κB in macrophages. Inflamm. Res. 2010, 59, 115–121. [Google Scholar] [CrossRef]
- Rafi, M.M.; Yadav, P.N.; Reyes, M. Lycopene inhibits LPS-induced proinflammatory mediator inducible nitric oxide synthase in mouse macrophage cells. J. Food Sci. 2007, 72, S069–S074. [Google Scholar] [CrossRef]
- Tang, F.Y.; Pai, M.H.; Wang, X.D. Consumption of lycopene inhibits the growth and progression of colon cancer in a mouse xenograft model. J. Agric. Food Chem. 2011, 59, 9011–9021. [Google Scholar] [CrossRef] [PubMed]
- Tapiero, H.; Townsend, D.M.; Tew, K.D. The role of carotenoids in the prevention of human pathologies. Biomed Pharmacother. 2004, 58, 100–110. [Google Scholar] [CrossRef]
- Langner, E.; Lemieszek, M.K.; Rzeski, W. Lycopene, sulforaphane, quercetin, and curcumin applied together show improved antiproliferative potential in colon cancer cells in vitro. J. Food Biochem. 2019, 43, e12802. [Google Scholar] [CrossRef] [PubMed]
- Lin, M.-C.; Wang, F.Y.; Kuo, Y.H.; Tang, F.Y. Cancer chemopreventive effects of lycopene: Suppression of MMP-7 expression and cell invasion in human colon cancer cells. J. Agric. Food Chem. 2011, 59, 11304–11318. [Google Scholar] [CrossRef] [PubMed]
- Huang, C.S.; Shih, M.K.; Chuang, C.H.; Hu, M.L. Lycopene inhibits cell migration and invasion and upregulates Nm23-H1 in a highly invasive hepatocarcinoma, SK-Hep-1 cells. J. Nutr. 2005, 135, 2119–2123. [Google Scholar] [CrossRef] [PubMed]
- Hwang, E.S.; Lee, H.J. Inhibitory effects of lycopene on the adhesion, invasion, and migration of SK-Hep1 human hepatoma cells. Exp. Biol. Med. 2006, 231, 322–327. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.; Chen, Y.; Fu, J.; Yang, Q.; Feng, L. High-throughput screening of lycopene-overproducing mutants of Blakeslea trispora by combining ARTP mutation with microtiter plate cultivation and transcriptional changes revealed by RNA-seq. Biochem. Engin. J. 2020, 161, 107664. [Google Scholar] [CrossRef]
- Alvi, S.S.; Ansari, I.A.; Khan, I.; Iqbal, J.; Khan, M.S. Potential role of lycopene in targeting proprotein convertase subtilisin/kexin type-9 to combat hypercholesterolemia. Free Radic. Biol. Med. 2017, 108, 384–403. [Google Scholar]
- Rogalińska, M.; Góralski, P.; Blonski, J.Z.; Robak, P.; Barciszewski, J.; Koceva-Chyła, A.; Piekarski, H.; Robak, T.; Kiliańska, Z.M. Personalized therapy tests for the monitoring of chronic lymphocytic leukemia development. Oncol. Lett. 2017, 13, 2079–2084. [Google Scholar] [CrossRef]
- Góralski, P.; Rogalińska, M.; Błoński, J.Z.; Pytel, E.; Robak, T.; Kilianska, Z.M.; Piekarski, H. The differences in thermal profiles between normal and leukemic cells exposed to anticancer drug evaluated by diffrrential scanning calorimetry. J. Therm. Anal. Calorim. 2014, 118, 1339–1344. [Google Scholar] [CrossRef] [Green Version]
- Rogalińska, M.; Blonski, J.Z.; Góralski, P.; Wawrzyniak, E.; Rogalska, A.; Robak, P.; Koceva-Chyła, A.; Piekarski, H.; Robak, T.; Kiliańska, Z.M. Personalized therapy tests for the monitoring of chronic lymphocytic leukemia development. Int. J. Oncol. 2015, 46, 1259–1267. [Google Scholar] [CrossRef]
- Howells, L.M.; Iwuji, C.O.O.; Irving, G.R.B.; Barber, S.; Walter, H.; Sidat, Z.; Griffin-Teall, N.; Singh, R.; Foreman, N.; Patel, S.R.; et al. Curcumin combined with FOLFOX chemotherapy is safe and tolerable in patients with metastatic colorectal cancer in a randomized phase IIa trial. J. Nutr. 2019, 149, 1133–1139. [Google Scholar] [CrossRef] [Green Version]
- Pastorelli, D.; Fabricio, A.S.C.; Giovanis, P.; D’Ippolito, S.; Fiduccia, P.; Soldà, C.; Buda, A.; Sperti, C.; Bardini, R.; Da Dalt, G.; et al. Phytosome complex of curcumin as complementary therapy of advanced pancreatic cancer improves safety and efficacy of gemcitabine: Results of a prospective phase II trial. Pharm. Res. 2018, 132, 72–79. [Google Scholar] [CrossRef] [PubMed]
- Basak, S.K.; Bera, A.; Yoon, A.J.; Morselli, M.; Jeong, C.; Tosevska, A.; Dong, T.S.; Eklund, M.; Russ, E.; Nasser, H.; et al. A randomized, phase 1, placebo-controlled trial of APG-157 in oral cancer demonstrates systemic absorption and an inhibitory effect on cytokines and tumor-associated microbes. Cancer 2020, 126, 1668–1682. [Google Scholar] [CrossRef] [PubMed]
- Choi, Y.H.; Han, D.H.; Kim, S.W.; Kim, M.J.; Sung, H.H.; Jeon, H.G.; Jeong, B.C.; Seo, S.I.; Jeon, S.S.; Lee, H.M.; et al. A randomized, double-blind, placebo-controlled trial to evaluate the role of curcumin in prostate cancer patients with intermittent androgen deprivation. Prostate 2019, 79, 614–621. [Google Scholar] [CrossRef] [PubMed]
- Dützmann, S.; Schiborr, C.; Kocher, A.; Pilatus, U.; Hattingen, E.; Weissenberger, J.; Geßler, F.; Quick-Weller, J.; Franz, K.; Seifert, V.; et al. Intratumoral concentrations and effects of orally administered micellar curcuminoids in glioblastoma patients. Nutr. Cancer 2016, 68, 943–948. [Google Scholar] [CrossRef] [PubMed]
- Indrawati, L.; Ascobat, P.; Bela, B.; Abdullah, M.; Surono, I.S. The effect of an Annona muricata leaf extract on nutritional status and cytotoxicity in colorectal cancer: A randomized controlled trial. Asia Pac. J. Clin. Nutr. 2017, 26, 606–612. [Google Scholar]
- Brenjian, S.; Moini, A.; Yamini, N.; Kashani, L.; Faridmojtahedi, M.; Bahramrezaie, M.; Khodarahmian, M.; Amidi, F. Resveratrol treatment in patients with polycystic ovary syndrome decreased pro-inflammatory and endoplasmic reticulum stress markers. Am. J. Reprod. Immunol. 2020, 83, e13186. [Google Scholar] [CrossRef] [PubMed]
- Bahramrezaie, M.; Amidi, F.; Aleyasin, A.; Saremi, A.; Aghahoseini, M.; Brenjian, S.; Khodarahmian, M.; Pooladi, A. Effects of resveratrol on VEGF & HIF1 genes expression in granulosa cells in the angiogenesis pathway and laboratory parameters of polycystic ovary syndrome: A triple-blind randomized clinical trial. J. Assist. Reprod. Genet. 2019, 36, 1701–1712. [Google Scholar]
- Kurkjian, C.; Kummar, S. Advances in the treatment of metastatic colorectal cancer. Am. J. Ther. 2009, 16, 412–420. [Google Scholar] [CrossRef] [PubMed]
- Singh, C.K.; Ndiaye, M.A.; Ahmad, N. Resveratrol and cancer: Challenges for clinical translation. BBA 2015, 1852, 1178–1185. [Google Scholar] [CrossRef] [Green Version]
- 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.; et al. Clinical pharmacology of resveratrol and its metabolites in colorectal cancer patients. Cancer Res. 2010, 70, 7392–7399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Howells, L.M.; Berry, D.P.; Elliott, P.J.; Jacobson, E.W.; Hoffmann, E.; Hegarty, B.; Brown, K.; Steward, W.P.; 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] [PubMed] [Green Version]
- Lazzeroni, M.; Guerrieri-Gonzaga, A.; Gandini, S.; Johansson, H.; Serrano, D.; Cazzaniga, M.; Aristarco, V.; Macis, D.; Mora, S.; Caldarella, P.; et al. A presurgical study of lecithin formulation of green tea extract in women with early breast cancer. Cancer Prev. Res. 2017, 10, 363–370. [Google Scholar] [CrossRef] [Green Version]
- Samavat, H.; Ursin, G.; Emory, T.H.; Lee, E.; Wang, R.; Torkelson, C.J.; Dostal, A.M.; Swenson, K.; Le, C.T.; Yang, C.S.; et al. A randomized controlled trial of green tea extract supplementation and mammographic density in postmenopausal women at increased risk of breast cancer. Cancer Prev. Res. 2017, 10, 710–718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Neetha, M.C.; Panchaksharappa, M.G.; Pattabhiramasastry, S.; Shivaprasad, N.V.; Venkatesh, U.G. Chemopreventive synergism between green tea extract and curcumin in patients with potentially malignant oral disorders: A double-blind, randomized preliminary study. J. Contemp. Dent. Pract. 2020, 21, 521–531. [Google Scholar] [CrossRef]
- Shin, C.M.; Lee, D.H.; Seo, A.Y.; Lee, H.J.; Kim, S.B.; Son, W.C.; Kim, Y.K.; Lee, S.J.; Park, S.H.; Kim, N.; et al. Green tea extracts for the prevention of metachronous colorectal polyps among patients who underwent endoscopic removal of colorectal adenomas: A randomized clinical trial. Clin. Nutr. 2018, 37, 452–458. [Google Scholar] [CrossRef] [PubMed]
- Lozanovski, V.J.; Houben, P.; Hinz, U.; Hackert, T.; Herr, I.; Schemmer, P. Pilot study evaluating broccoli sprouts in advanced pancreatic cancer (POUDER trial)—Study protocol for a randomized controlled trial. Trials 2014, 15, 204. [Google Scholar] [CrossRef] [Green Version]
- Ferry, D.R.; Smith, A.; Malkhandi, J.; Fyfe, D.W.; de Takats, P.G.; Anderson, D.; Baker, J.; Kerr, D.J. Phase I clinical trial of the flavonoid quercetin: Pharmacokinetics and evidence for in vivo tyrosine kinase inhibition. Clin. Cancer Res. 1996, 2, 659–668. [Google Scholar]
- Jatoi, A.; Burch, P.; Hillman, D.; Vanyo, J.M.; Dakhil, S.; Nikcevich, D.; Rowland, K.; Morton, R.; Flynn, P.J.; Young, C.; et al. North central cancer treatment group. A tomato-based, lycopene-containing intervention for androgen-independent prostate cancer: Results of a Phase II study from the North Central Cancer Treatment Group. Urology 2007, 69, 289–294. [Google Scholar] [CrossRef] [PubMed]
- Paur, I.; Lilleby, W.; Bøhn, S.K.; Hulander, E.; Klein, W.; Vlatkovic, L.; Axcrona, K.; Bolstad, N.; Bjøro, T.; Laake, P.; et al. Tomato-based randomized controlled trial in prostate cancer patients: Effect on PSA. Clin. Nutr. 2017, 36, 672–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lane, J.A.; Er, V.; Avery, K.N.L.; Horwood, J.; Cantwell, M.; Caro, G.P.; Crozier, A.; Smith, G.D.; Donovan, J.L.; Down, L.; et al. ProDiet: A phase II randomized placebo-controlled trial of green tea catechins and lycopene in men at increased risk of prostate cancer. Cancer Prev. Res. 2018, 11, 687–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beynon, R.A.; Richmond, R.C.; Santos Ferreira, D.L.; Ness, A.R.; May, M.; Smith, G.D.; Vincent, E.E.; Adams, C.; Ala-Korpela, M.; Würtz, P.; et al. Protec t study group; PRACTICAL consortium. Investigating the effects of lycopene and green tea on the metabolome of men at risk of prostate cancer: The ProDiet randomised controlled trial. Int. J. Cancer 2019, 144, 1918–1928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alfano, C.M.; Day, J.M.; Katz, M.L.; Herndon, J.E.; Bittoni, M.A.; Oliveri, J.M.; Donohue, K.; Paskett, E.D. Exercise and dietary change after diagnosis and cancer-related symptoms in long-term survivors of breast cancer: CALGB 79804. Psychological 2009, 18, 128–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kubczak, M.; Szustka, A.; Rogalińska, M. Molecular Targets of Natural Compounds with Anti-Cancer Properties. Int. J. Mol. Sci. 2021, 22, 13659. https://doi.org/10.3390/ijms222413659
Kubczak M, Szustka A, Rogalińska M. Molecular Targets of Natural Compounds with Anti-Cancer Properties. International Journal of Molecular Sciences. 2021; 22(24):13659. https://doi.org/10.3390/ijms222413659
Chicago/Turabian StyleKubczak, Małgorzata, Aleksandra Szustka, and Małgorzata Rogalińska. 2021. "Molecular Targets of Natural Compounds with Anti-Cancer Properties" International Journal of Molecular Sciences 22, no. 24: 13659. https://doi.org/10.3390/ijms222413659