The Role of Natural Flavonoids as Telomerase Inhibitors in Suppressing Cancer Growth
Abstract
:1. Introduction
2. Telomerase as a Potential Target for Cancer Therapy
3. Flavanols—Epigallocatechin Gallate (EGCG) and Epicatechin Gallate (ECG)
4. Flavones—Apigenin and Luteolin
4.1. Apigenin
4.2. Luteolin
5. Flavonols—Quercetin, Kaempferol, and Morin
5.1. Quercetin
5.2. Kaempferol
5.3. Morin
6. Combinatorial Studies with Flavonoids and MST312
7. Conclusions
8. Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Igney, F.H.; Krammer, P.H. Death and anti-death: Tumour resistance to apoptosis. Nat. Rev. Cancer 2002, 2, 277–288. [Google Scholar] [CrossRef] [PubMed]
- Popli, D.B.; Sircar, K.; Chowdhry, A. Telomerase: An exploration toward the end of cancer. Indian J. Dent. Res. 2017, 28, 574–584. [Google Scholar] [CrossRef] [PubMed]
- Chung, S.S.; Adekoya, D.; Enenmoh, I.; Clarke, O.; Wang, P.; Sarkyssian, M.; Wu, Y.; Vadgama, J.V. Salinomycin abolished STAT3 and STAT1 interactions and reduced telomerase activity in colorectal cancer cells. Anticancer Res. 2017, 37, 445–453. [Google Scholar] [CrossRef] [PubMed]
- Hiyama, E.; Hiyama, K. Telomere and telomerase in stem cells. Br. J. Cancer 2007, 96, 1020–1024. [Google Scholar] [CrossRef]
- Li, Y.; Tergaonkar, V. Telomerase reactivation in cancers: Mechanisms that govern transcriptional activation of the wild-type vs. mutant TERT promoters. Transcription 2016, 7, 44–49. [Google Scholar] [CrossRef]
- Khattar, E.; Tergaonkar, V. Transcriptional regulation of telomerase reverse transcriptase (TERT) by MYC. Front. Cell Dev. Biol. 2017, 5, 1. [Google Scholar] [CrossRef]
- Masutomi, K.; Possemato, R.; Wong, J.M.; Currier, J.L.; Tothova, Z.; Manola, J.B.; Ganesan, S.; Lansdorp, P.M.; Collins, K.; Hahn, W.C. The telomerase reverse transcriptase regulates chromatin state and DNA damage responses. Proc. Natl. Acad. Sci. USA 2005, 102, 8222–8227. [Google Scholar] [CrossRef]
- Akincilar, S.C.; Unal, B.; Tergaonkar, V. Reactivation of telomerase in cancer. Cell. Mol. Life Sci. 2016, 73, 1659–1670. [Google Scholar] [CrossRef]
- Xu, X.; Li, Y.; Bharath, S.R.; Ozturk, M.B.; Bowler, M.W.; Loo, B.Z.L.; Tergaonkar, V.; Song, H. Structural basis for reactivating the mutant TERT promoter by cooperative binding of p52 and ETS1. Nat. Commun. 2018, 9, 3183. [Google Scholar] [CrossRef]
- Kazemi-Lomedasht, F.; Rami, A.; Zarghami, N. Comparison of inhibitory effect of curcumin nanoparticles and free curcumin in human telomerase reverse transcriptase gene expression in breast cancer. Adv. Pharm. Bull. 2013, 3, 127–130. [Google Scholar] [CrossRef]
- Fernandes, S.G.; Dsouza, R.; Pandya, G.; Kirtonia, A.; Tergaonkar, V.; Lee, S.Y.; Garg, M.; Khattar, E. Role of telomeres and telomeric proteins in human malignancies and their therapeutic potential. Cancers 2020, 12, 1901. [Google Scholar] [CrossRef] [PubMed]
- Ozturk, M.B.; Li, Y.; Tergaonkar, V. Current insights to regulation and role of telomerase in human diseases. Antioxidants 2017, 6, 17. [Google Scholar] [CrossRef] [PubMed]
- Cancer Statistics—NCI. Available online: https://www.cancer.gov/about-cancer/understanding/statistics (accessed on 4 April 2023).
- Worldwide Cancer Data|World Cancer Research Fund International. Available online: https://www.wcrf.org/cancer-trends/worldwide-cancer-data/ (accessed on 4 April 2023).
- Ang, H.L.; Mohan, C.D.; Shanmugam, M.K.; Leong, H.C.; Makvandi, P.; Rangappa, K.S.; Bishayee, A.; Kumar, A.P.; Sethi, G. Mechanism of epithelial-mesenchymal transition in cancer and its regulation by natural compounds. Med. Res. Rev. 2023. [Google Scholar] [CrossRef] [PubMed]
- Tan, S.M.; Li, F.; Rajendran, P.; Kumar, A.P.; Hui, K.M.; Sethi, G. Identification of beta-escin as a novel inhibitor of signal transducer and activator of transcription 3/Janus-activated kinase 2 signaling pathway that suppresses proliferation and induces apoptosis in human hepatocellular carcinoma cells. J. Pharmacol. Exp. Ther. 2010, 334, 285–293. [Google Scholar] [CrossRef] [PubMed]
- Ahn, K.S.; Sethi, G.; Chaturvedi, M.M.; Aggarwal, B.B. Simvastatin, 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, suppresses osteoclastogenesis induced by receptor activator of nuclear factor-kappaB ligand through modulation of NF-kappaB pathway. Int. J. Cancer 2008, 123, 1733–1740. [Google Scholar] [CrossRef]
- Badrzadeh, F.; Akbarzadeh, A.; Zarghami, N.; Yamchi, M.R.; Zeighamian, V.; Tabatabae, F.S.; Taheri, M.; Kafil, H.S. Comparison between effects of free curcumin and curcumin loaded NIPAAm-MAA nanoparticles on telomerase and PinX1 gene expression in lung cancer cells. Asian Pac. J. Cancer Prev. 2014, 15, 8931–8936. [Google Scholar] [CrossRef]
- Ncube, B.; Van Staden, J. Tilting plant metabolism for improved metabolite biosynthesis and enhanced human benefit. Molecules 2015, 20, 12698–12731. [Google Scholar] [CrossRef]
- Seca, A.M.L.; Pinto, D. Plant secondary metabolites as anticancer agents: Successes in clinical trials and therapeutic application. Int. J. Mol. Sci. 2018, 19, 263. [Google Scholar] [CrossRef]
- Nabavi, S.M.; Šamec, D.; Tomczyk, M.; Milella, L.; Russo, D.; Habtemariam, S.; Suntar, I.; Rastrelli, L.; Daglia, M.; Xiao, J.; et al. Flavonoid biosynthetic pathways in plants: Versatile targets for metabolic engineering. Biotechnol. Adv. 2020, 38, 107316. [Google Scholar] [CrossRef]
- Liu, J.; Wang, X.; Yong, H.; Kan, J.; Jin, C. Recent advances in flavonoid-grafted polysaccharides: Synthesis, structural characterization, bioactivities and potential applications. Int. J. Biol. Macromol. 2018, 116, 1011–1025. [Google Scholar] [CrossRef]
- Sajeev, A.; Hegde, M.; Girisa, S.; Devanarayanan, T.N.; Alqahtani, M.S.; Abbas, M.; Sil, S.K.; Sethi, G.; Chen, J.T.; Kunnumakkara, A.B. Oroxylin A: A promising flavonoid for prevention and treatment of chronic diseases. Biomolecules 2022, 12, 1185. [Google Scholar] [CrossRef] [PubMed]
- Aggarwal, V.; Tuli, H.S.; Thakral, F.; Singhal, P.; Aggarwal, D.; Srivastava, S.; Pandey, A.; Sak, K.; Varol, M.; Khan, M.A.; et al. Molecular mechanisms of action of hesperidin in cancer: Recent trends and advancements. Exp. Biol. Med. (Maywood) 2020, 245, 486–497. [Google Scholar] [CrossRef] [PubMed]
- Rodríguez-García, C.; Sánchez-Quesada, C.; Gaforio, J.J. Dietary flavonoids as cancer chemopreventive agents: An updated review of human studies. Antioxidants 2019, 8, 137. [Google Scholar] [CrossRef] [PubMed]
- Yang, M.H.; Jung, S.H.; Um, J.Y.; Kumar, A.P.; Sethi, G.; Ahn, K.S. Daidzin targets epithelial-to-mesenchymal transition process by attenuating manganese superoxide dismutase expression and PI3K/Akt/mTOR activation in tumor cells. Life Sci. 2022, 295, 120395. [Google Scholar] [CrossRef]
- Yang, M.H.; Jung, S.H.; Chinnathambi, A.; Alahmadi, T.A.; Alharbi, S.A.; Sethi, G.; Ahn, K.S. Attenuation of STAT3 signaling cascade by daidzin can enhance the apoptotic potential of bortezomib against multiple myeloma. Biomolecules 2019, 10, 23. [Google Scholar] [CrossRef]
- Kopustinskiene, D.M.; Jakstas, V.; Savickas, A.; Bernatoniene, J. Flavonoids as anticancer agents. Nutrients 2020, 12, 457. [Google Scholar] [CrossRef]
- Tuli, H.S.; Kaur, J.; Vashishth, K.; Sak, K.; Sharma, U.; Choudhary, R.; Behl, T.; Singh, T.; Sharma, S.; Saini, A.K.; et al. Molecular mechanisms behind ROS regulation in cancer: A balancing act between augmented tumorigenesis and cell apoptosis. Arch. Toxicol. 2023, 97, 103–120. [Google Scholar] [CrossRef]
- Amic, D.; Davidović-Amić, D.; Beslo, D.; Trinajstić, N. Structure-radical scavenging activity relationships of flavonoids. Croat. Chem. Acta 2003, 76, 55–61. [Google Scholar]
- Xu, B.; Li, C.; Sung, C. Telomerase inhibitory effects of medicinal mushrooms and lichens, and their anticancer activity. Int. J. Med. Mushrooms 2014, 16, 17–28. [Google Scholar] [CrossRef]
- Xu, B.; Sung, C. Telomerase inhibitory effects and anti-proliferative properties of onion and other natural spices against cancer cells. Food Biosci. 2015, 10, 80–85. [Google Scholar] [CrossRef]
- Li, Y.; Cheng, H.S.; Chng, W.J.; Tergaonkar, V. Activation of mutant TERT promoter by RAS-ERK signaling is a key step in malignant progression of BRAF-mutant human melanomas. Proc. Natl. Acad. Sci. USA 2016, 113, 14402–14407. [Google Scholar] [CrossRef] [PubMed]
- Shanmugam, R.; Ozturk, M.B.; Low, J.L.; Akincilar, S.C.; Chua, J.Y.H.; Thangavelu, M.T.; Periyasamy, G.; DasGupta, R.; Tergaonkar, V. Genome-wide screens identify specific drivers of mutant hTERT promoters. Proc. Natl. Acad. Sci. USA 2022, 119, e2105171119. [Google Scholar] [CrossRef] [PubMed]
- Akıncılar, S.C.; Chua, J.Y.H.; Ng, Q.F.; Chan, C.H.T.; Eslami, S.Z.; Chen, K.; Low, J.L.; Arumugam, S.; Aswad, L.; Chua, C.; et al. Identification of mechanism of cancer-cell-specific reactivation of hTERT offers therapeutic opportunities for blocking telomerase specifically in human colorectal cancer. Nucleic Acids Res. 2023, 51, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Buseman, C.M.; Wright, W.E.; Shay, J.W. Is telomerase a viable target in cancer? Mutat. Res. 2012, 730, 90–97. [Google Scholar] [CrossRef] [PubMed]
- Khattar, E.; Kumar, P.; Liu, C.Y.; Akıncılar, S.C.; Raju, A.; Lakshmanan, M.; Maury, J.J.; Qiang, Y.; Li, S.; Tan, E.Y.; et al. Telomerase reverse transcriptase promotes cancer cell proliferation by augmenting tRNA expression. J. Clin. Investig. 2016, 126, 4045–4060. [Google Scholar] [CrossRef]
- Bryan, C.; Rice, C.; Hoffman, H.; Harkisheimer, M.; Sweeney, M.; Skordalakes, E. Structural basis of telomerase inhibition by the highly specific BIBR1532. Structure 2015, 23, 1934–1942. [Google Scholar] [CrossRef] [PubMed]
- Cruz, J.; Wojdyla, L.; Ivancich, M.; Puri, N. Targeting the telomere with T-Oligo, G-Quadruplex stabilizers, and tankyrase inhibitors. J. Cancer Sci. Ther. 2014, 6, 429–432. [Google Scholar] [CrossRef]
- Imetelstat Sodium in Treating Young Patients with Refractory or Recurrent Solid Tumors or Lymphoma. Available online: https://clinicaltrials.gov/ct2/show/NCT01273090 (accessed on 16 February 2023).
- Imetelstat in Combination With Paclitaxel (With or Without Bevacizumab) in Patients With Locally Recurrent or Metastatic Breast Cancer. Available online: https://clinicaltrials.gov/ct2/show/NCT01256762 (accessed on 16 February 2023).
- Open Label Study to Evaluate the Activity of Imetelstat in Patients With Essential Thrombocythemia or Polycythemia Vera. Available online: https://www.clinicaltrials.gov/ct2/show/NCT01243073 (accessed on 16 February 2023).
- Open Label Study With Imetelstat to Determine Effect of Imetelstat in Patients w/ Previously Treated Multiple Myeloma. Available online: https://clinicaltrials.gov/ct2/show/NCT01242930 (accessed on 16 February 2023).
- Imetelstat as Maintenance Therapy After Initial Induction Chemotherapy in Non-Small Cell Lung Cancer (NSCLC). Available online: https://clinicaltrials.gov/ct2/show/NCT01137968 (accessed on 16 February 2023).
- Kim, N.W.; Piatyszek, M.A.; Prowse, K.R.; Harley, C.B.; West, M.D.; Ho, P.L.; Coviello, G.M.; Wright, W.E.; Weinrich, S.L.; Shay, J.W. Specific association of human telomerase activity with immortal cells and cancer. Science 1994, 266, 2011–2015. [Google Scholar] [CrossRef]
- Sethi, G.; Ahn, K.S.; Sandur, S.K.; Lin, X.; Chaturvedi, M.M.; Aggarwal, B.B. Indirubin enhances tumor necrosis factor-induced apoptosis through modulation of nuclear factor-kappa B signaling pathway. J. Biol. Chem. 2006, 281, 23425–23435. [Google Scholar] [CrossRef]
- Ahn, K.S.; Sethi, G.; Aggarwal, B.B. Simvastatin potentiates TNF-alpha-induced apoptosis through the down-regulation of NF-kappaB-dependent antiapoptotic gene products: Role of IkappaBalpha kinase and TGF-beta-activated kinase-1. J. Immunol. 2007, 178, 2507–2516. [Google Scholar] [CrossRef]
- Ullah, A.; Munir, S.; Badshah, S.L.; Khan, N.; Ghani, L.; Poulson, B.G.; Emwas, A.H.; Jaremko, M. Important flavonoids and their role as a therapeutic agent. Molecules 2020, 25, 5243. [Google Scholar] [CrossRef] [PubMed]
- Gervasi, T.; Calderaro, A.; Barreca, D.; Tellone, E.; Trombetta, D.; Ficarra, S.; Smeriglio, A.; Mandalari, G.; Gattuso, G. Biotechnological applications and health-promoting properties of flavonols: An updated view. Int. J. Mol. Sci. 2022, 23, 1710. [Google Scholar] [CrossRef] [PubMed]
- Guterres, A.N.; Villanueva, J. Targeting telomerase for cancer therapy. Oncogene 2020, 39, 5811–5824. [Google Scholar] [CrossRef] [PubMed]
- Murkovic, M. Phenolic compounds: Occurrence, classes, and analysis. In Encyclopedia of Food and Health; Caballero, B., Finglas, P.M., Toldrá, F., Eds.; Academic Press: Oxford, UK, 2016; pp. 346–351. [Google Scholar] [CrossRef]
- Herrero, M.; Plaza, M.; Cifuentes, A.; Ibáñez, E. Extraction techniques for the determination of phenolic compounds in food. Compr. Sampl. Sample Prep. 2012, 4, 159–180. [Google Scholar] [CrossRef]
- 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]
- Shirakami, Y.; Shimizu, M. Possible mechanisms of green tea and its constituents against cancer. Molecules 2018, 23, 2284. [Google Scholar] [CrossRef]
- Meeran, S.M.; Patel, S.N.; Chan, T.H.; Tollefsbol, T.O. A novel prodrug of epigallocatechin-3-gallate: Differential epigenetic hTERT repression in human breast cancer cells. Cancer Prev. Res. 2011, 4, 1243–1254. [Google Scholar] [CrossRef]
- Moradzadeh, M.; Hosseini, A.; Erfanian, S.; Rezaei, H. Epigallocatechin-3-gallate promotes apoptosis in human breast cancer T47D cells through down-regulation of PI3K/AKT and Telomerase. Pharmacol. Rep. 2017, 69, 924–928. [Google Scholar] [CrossRef]
- Gurung, R.L.; Lim, S.N.; Low, G.K.; Hande, M.P. MST-312 alters telomere dynamics, gene expression profiles and growth in human breast cancer cells. J. Nutrigenet. Nutr. 2014, 7, 283–298. [Google Scholar] [CrossRef]
- Zhang, Y.; Han, G.; Fan, B.; Zhou, Y.; Zhou, X.; Wei, L.; Zhang, J. Green tea (-)-epigallocatechin-3-gallate down-regulates VASP expression and inhibits breast cancer cell migration and invasion by attenuating Rac1 activity. Eur. J. Pharmacol. 2009, 606, 172–179. [Google Scholar] [CrossRef]
- Fatemi, A.; Safa, M.; Kazemi, A. MST-312 induces G2/M cell cycle arrest and apoptosis in APL cells through inhibition of telomerase activity and suppression of NF-κB pathway. Tumor Biol. 2015, 36, 8425–8437. [Google Scholar] [CrossRef] [PubMed]
- Sadava, D.; Whitlock, E.; Kane, S.E. The green tea polyphenol, epigallocatechin-3-gallate inhibits telomerase and induces apoptosis in drug-resistant lung cancer cells. Biochem. Biophys. Res. Commun. 2007, 360, 233–237. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.; Zuo, J.; Wang, G. Epigallocatechin-3-gallate suppresses cell proliferation and promotes apoptosis in Ec9706 and Eca109 esophageal carcinoma cells. Oncol. Lett. 2017, 14, 4391–4395. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Yang, P.; Gao, F.; Yang, J.; Yao, K. Effects of epigallocatechin gallate on the proliferation and apoptosis of the nasopharyngeal carcinoma cell line CNE2. Exp. Ther. Med. 2014, 8, 1783–1788. [Google Scholar] [CrossRef]
- Yokoyama, M.; Noguchi, M.; Nakao, Y.; Pater, A.; Iwasaka, T. The tea polyphenol, (-)-epigallocatechin gallate effects on growth, apoptosis, and telomerase activity in cervical cell lines. Gynecol. Oncol. 2004, 92, 197–204. [Google Scholar] [CrossRef]
- Noguchi, M.; Yokoyama, M.; Watanabe, S.; Uchiyama, M.; Nakao, Y.; Hara, K.; Iwasaka, T. Inhibitory effect of the tea polyphenol, (-)-epigallocatechin gallate, on growth of cervical adenocarcinoma cell lines. Cancer Lett. 2006, 234, 135–142. [Google Scholar] [CrossRef]
- Yokoyama, M.; Noguchi, M.; Nakao, Y.; Ysunaga, M.; Yamasaki, F.; Iwasaka, T. Antiproliferative effects of the major tea polyphenol, (-)-epigallocatechin gallate and retinoic acid in cervical adenocarcinoma. Gynecol. Oncol. 2008, 108, 326–331. [Google Scholar] [CrossRef]
- Chen, H.; Landen, C.N.; Li, Y.; Alvarez, R.D.; Tollefsbol, T.O. Epigallocatechin gallate and sulforaphane combination treatment induce apoptosis in paclitaxel-resistant ovarian cancer cells through hTERT and Bcl-2 down-regulation. Exp. Cell Res. 2013, 319, 697–706. [Google Scholar] [CrossRef]
- Lin, S.C.; Li, W.C.; Shih, J.W.; Hong, K.F.; Pan, Y.R.; Lin, J.J. The tea polyphenols EGCG and EGC repress mRNA expression of human telomerase reverse transcriptase (hTERT) in carcinoma cells. Cancer Lett. 2006, 236, 80–88. [Google Scholar] [CrossRef]
- Shervington, A.; Pawar, V.; Menon, S.; Thakkar, D.; Patel, R. The sensitization of glioma cells to cisplatin and tamoxifen by the use of catechin. Mol. Biol. Rep. 2009, 36, 1181–1186. [Google Scholar] [CrossRef]
- Shukla, S.; Gupta, S. Chapter 41—Apigenin and cancer chemoprevention. In Bioactive Foods in Promoting Health; Watson, R.R., Preedy, V.R., Eds.; Academic Press: San Diego, CA, USA, 2010; pp. 663–689. [Google Scholar] [CrossRef]
- Kashyap, D.; Sharma, A.; Tuli, H.S.; Sak, K.; Garg, V.K.; Buttar, H.S.; Setzer, W.N.; Sethi, G. Apigenin: A natural bioactive flavone-type molecule with promising therapeutic function. J. Funct. Foods 2018, 48, 457–471. [Google Scholar] [CrossRef]
- Shields, M. Chapter 14—Chemotherapeutics. In Pharmacognosy; Badal, S., Delgoda, R., Eds.; Academic Press: Boston, MA, USA, 2017; pp. 295–313. [Google Scholar] [CrossRef]
- Yan, X.; Qi, M.; Li, P.; Zhan, Y.; Shao, H. Apigenin in cancer therapy: Anti-cancer effects and mechanisms of action. Cell Biosci. 2017, 7, 50. [Google Scholar] [CrossRef] [PubMed]
- Jayasooriya, R.G.; Kang, S.H.; Kang, C.H.; Choi, Y.H.; Moon, D.O.; Hyun, J.W.; Chang, W.Y.; Kim, G.Y. Apigenin decreases cell viability and telomerase activity in human leukemia cell lines. Food Chem. Toxicol. 2012, 50, 2605–2611. [Google Scholar] [CrossRef] [PubMed]
- Chakrabarti, M.; Banik, N.L.; Ray, S.K. Sequential hTERT knockdown and apigenin treatment inhibited invasion and proliferation and induced apoptosis in human malignant neuroblastoma SK-N-DZ and SK-N-BE2 cells. J. Mol. Neurosci. 2013, 51, 187–198. [Google Scholar] [CrossRef]
- Omar, S.H. Chapter 4—Biophenols: Impacts and prospects in anti-alzheimer drug discovery. In Discovery and Development of Neuroprotective Agents from Natural Products; Brahmachari, G., Ed.; Elsevier: Amsterdam, The Netherlands, 2018; pp. 103–148. [Google Scholar] [CrossRef]
- Shukla, R.; Pandey, V.; Vadnere, G.P.; Lodhi, S. Chapter 18—Role of flavonoids in management of inflammatory disorders. In Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases, 2nd ed.; Watson, R.R., Preedy, V.R., Eds.; Academic Press: Cambridge, MA, USA, 2019; pp. 293–322. [Google Scholar] [CrossRef]
- Selvi, R.B.; Swaminathan, A.; Chatterjee, S.; Shanmugam, M.K.; Li, F.; Ramakrishnan, G.B.; Siveen, K.S.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; et al. Inhibition of p300 lysine acetyltransferase activity by luteolin reduces tumor growth in head and neck squamous cell carcinoma (HNSCC) xenograft mouse model. Oncotarget 2015, 6, 43806–43818. [Google Scholar] [CrossRef]
- Tuli, H.S.; Garg, V.K.; Bhushan, S.; Uttam, V.; Sharma, U.; Jain, A.; Sak, K.; Yadav, V.; Lorenzo, J.M.; Dhama, K.; et al. Natural flavonoids exhibit potent anticancer activity by targeting microRNAs in cancer: A signature step hinting towards clinical perfection. Transl. Oncol. 2023, 27, 101596. [Google Scholar] [CrossRef]
- Wang, Z.; Zeng, M.; Wang, Z.; Qin, F.; Chen, J.; He, Z. Dietary luteolin: A narrative review focusing on its pharmacokinetic properties and effects on glycolipid metabolism. J. Agric. Food Chem. 2021, 69, 1441–1454. [Google Scholar] [CrossRef]
- Imran, M.; Rauf, A.; Abu-Izneid, T.; Nadeem, M.; Shariati, M.A.; Khan, I.A.; Imran, A.; Orhan, I.E.; Rizwan, M.; Atif, M.; et al. Luteolin, a flavonoid, as an anticancer agent: A review. Biomed. Pharmacother. 2019, 112, 108612. [Google Scholar] [CrossRef]
- Huang, L.; Jin, K.; Lan, H. Luteolin inhibits cell cycle progression and induces apoptosis of breast cancer cells through downregulation of human telomerase reverse transcriptase. Oncol. Lett. 2019, 17, 3842–3850. [Google Scholar] [CrossRef]
- Pai, A.; Lobo, R.; Basu-Mallik, S.; Jayashree, B.S. Extra precision docking studies of novel luteolin analogues for the inhibition of Tankyrase II: A theoretical-based approach toward novel cancer target. Thai J. Pharm. Sci. 2017, 41, 138–143. [Google Scholar]
- Williamson, G.; Day, A.J.; Plumb, G.W.; Couteau, D. Human metabolic pathways of dietary flavonoids and cinnamates. Biochem. Soc. Trans. 2000, 28, 16–22. [Google Scholar] [CrossRef] [PubMed]
- Popova, M.P.; Chinou, I.B.; Marekov, I.N.; Bankova, V.S. Terpenes with antimicrobial activity from Cretan propolis. Phytochemistry 2009, 70, 1262–1271. [Google Scholar] [CrossRef] [PubMed]
- Falcão, S.I.; Vale, N.; Gomes, P.; Domingues, M.R.; Freire, C.; Cardoso, S.M.; Vilas-Boas, M. Phenolic profiling of Portuguese propolis by LC-MS spectrometry: Uncommon propolis rich in flavonoid glycosides. Phytochem. Anal. 2013, 24, 309–318. [Google Scholar] [CrossRef] [PubMed]
- Lakhanpal, P.; Rai, D.K. Quercetin: A versatile flavonoid. Internet J. Med. Update 2007, 2, 20–35. [Google Scholar] [CrossRef]
- Lou, M.; Zhang, L.N.; Ji, P.G.; Feng, F.Q.; Liu, J.H.; Yang, C.; Li, B.F.; Wang, L. Quercetin nanoparticles induced autophagy and apoptosis through AKT/ERK/Caspase-3 signaling pathway in human neuroglioma cells: In vitro and in vivo. Biomed. Pharmacother. 2016, 84, 1–9. [Google Scholar] [CrossRef]
- Ren, K.W.; Li, Y.H.; Wu, G.; Ren, J.Z.; Lu, H.B.; Li, Z.M.; Han, X.W. Quercetin nanoparticles display antitumor activity via proliferation inhibition and apoptosis induction in liver cancer cells. Int. J. Oncol. 2017, 50, 1299–1311. [Google Scholar] [CrossRef] [PubMed]
- Naasani, I.; Seimiya, H.; Tsuruo, T. Telomerase inhibition, telomere shortening, and senescence of cancer cells by tea catechins. Biochem. Biophys. Res. Commun. 1998, 249, 391–396. [Google Scholar] [CrossRef] [PubMed]
- Avci, C.B.; Yilmaz, S.; Dogan, Z.O.; Saydam, G.; Dodurga, Y.; Ekiz, H.A.; Kartal, M.; Sahin, F.; Baran, Y.; Gunduz, C. Quercetin-induced apoptosis involves increased hTERT enzyme activity of leukemic cells. Hematology 2011, 16, 303–307. [Google Scholar] [CrossRef] [PubMed]
- Turgut Coşan, D.; Saydam, F.; Özbayer, C.; Doğaner, F.; Soyocak, A.; Güneş, H.V.; Değirmenci, İ.; Kurt, H.; Üstüner, M.C.; Bal, C. Impact of tannic acid on blood pressure, oxidative stress and urinary parameters in L-NNA-induced hypertensive rats. Cytotechnology 2015, 67, 97–105. [Google Scholar] [CrossRef]
- Nakayama, Y.; Sakamoto, H.; Satoh, K.; Yamamoto, T. Tamoxifen and gonadal steroids inhibit colon cancer growth in association with inhibition of thymidylate synthase, survivin and telomerase expression through estrogen receptor beta mediated system. Cancer Lett. 2000, 161, 63–71. [Google Scholar] [CrossRef]
- Choi, J.A.; Kim, J.Y.; Lee, J.Y.; Kang, C.M.; Kwon, H.J.; Yoo, Y.D.; Kim, T.W.; Lee, Y.S.; Lee, S.J. Induction of cell cycle arrest and apoptosis in human breast cancer cells by quercetin. Int. J. Oncol. 2001, 19, 837–844. [Google Scholar] [CrossRef] [PubMed]
- Kuo, S.M. Antiproliferative potency of structurally distinct dietary flavonoids on human colon cancer cells. Cancer Lett. 1996, 110, 41–48. [Google Scholar] [CrossRef] [PubMed]
- Gibellini, L.; Pinti, M.; Nasi, M.; Montagna, J.P.; De Biasi, S.; Roat, E.; Bertoncelli, L.; Cooper, E.L.; Cossarizza, A. Quercetin and cancer chemoprevention. Evid. Based Complement. Altern. Med. 2011, 2011, 591356. [Google Scholar] [CrossRef] [PubMed]
- Cui, Y.; Han, Y.; Yang, X.; Sun, Y.; Zhao, Y. Protective effects of quercetin and quercetin-5′,8-disulfonate against carbon tetrachloride-caused oxidative liver injury in mice. Molecules 2013, 19, 291–305. [Google Scholar] [CrossRef] [PubMed]
- Parvaresh, A.; Razavi, R.; Rafie, N.; Ghiasvand, R.; Pourmasoumi, M.; Miraghajani, M. Quercetin and ovarian cancer: An evaluation based on a systematic review. J. Res. Med. Sci. 2016, 21, 34. [Google Scholar] [CrossRef] [PubMed]
- Teiten, M.H.; Gaascht, F.; Dicato, M.; Diederich, M. Targeting the wingless signaling pathway with natural compounds as chemopreventive or chemotherapeutic agents. Curr. Pharm. Biotechnol. 2012, 13, 245–254. [Google Scholar] [CrossRef]
- Chen, S.F.; Nien, S.; Wu, C.H.; Liu, C.L.; Chang, Y.C.; Lin, Y.S. Reappraisal of the anticancer efficacy of quercetin in oral cancer cells. J. Chin. Med. Assoc. 2013, 76, 146–152. [Google Scholar] [CrossRef]
- Tang, S.N.; Singh, C.; Nall, D.; Meeker, D.; Shankar, S.; Srivastava, R.K. The dietary bioflavonoid quercetin synergizes with epigallocathechin gallate (EGCG) to inhibit prostate cancer stem cell characteristics, invasion, migration and epithelial-mesenchymal transition. J. Mol. Signal. 2010, 5, 14. [Google Scholar] [CrossRef]
- Ak, A.; Başaran, A.; Degirmenci, I.; Güneş, H.V.; Dıkmen, M. Evaluation of effects of quercetin (3,3′,4′,5,7-pentohidroxyflavon) on apoptosis and telomerase enzyme activity in Mcf7 And Nih-3t3 cell lines to compared with tamoxifen. Balk. Med. J. 2011, 28, 293–299. [Google Scholar] [CrossRef]
- Kuhar, M.; Imran, S.; Singh, N. Curcumin and quercetin combined with cisplatin to induce apoptosis in human laryngeal carcinoma Hep-2 cells through the mitochondrial pathway. J. Cancer Mol. 2007, 3, 121–128. [Google Scholar] [CrossRef]
- Zheng, D.S.; Chen, L.S. Triterpenoids from Ganoderma lucidum inhibit the activation of EBV antigens as telomerase inhibitors. Exp. Ther. Med. 2017, 14, 3273–3278. [Google Scholar] [CrossRef] [PubMed]
- Behjati, M.; Hashemi, M.; Kazemi, M.; Salehi, M.; Javanmard, S.H. Evaluation of energy balance on human telomerase reverse transcriptase (hTERT) alternative splicing by semi-quantitative RT-PCR in human umbilical vein endothelial cells. Adv. Biomed. Res. 2017, 6, 43. [Google Scholar] [CrossRef] [PubMed]
- Zamin, L.L.; Filippi-Chiela, E.C.; Dillenburg-Pilla, P.; Horn, F.; Salbego, C.; Lenz, G. Resveratrol and quercetin cooperate to induce senescence-like growth arrest in C6 rat glioma cells. Cancer Sci. 2009, 100, 1655–1662. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Ji, H.S.; Kang, J.H.; Shin, D.H.; Park, H.Y.; Choi, M.S.; Lee, C.H.; Lee, I.K.; Yun, B.S.; Jeong, T.S. Soy leaf extract containing kaempferol glycosides and pheophorbides improves glucose homeostasis by enhancing pancreatic β-cell function and suppressing hepatic lipid accumulation in db/db mice. J. Agric. Food Chem. 2015, 63, 7198–7210. [Google Scholar] [CrossRef] [PubMed]
- Bhagwat, S.; Haytowitz, D.B.; Holden, J.M. USDA Database For The Flavonoid Content Of Selected Foods, Release 3.1; US Department of Agriculture: Beltsville, MD, USA, 2014. [Google Scholar]
- Calderón-Montaño, J.M.; Burgos-Morón, E.; Pérez-Guerrero, C.; López-Lázaro, M. A review on the dietary flavonoid kaempferol. Mini-Rev. Med. Chem. 2011, 11, 298–344. [Google Scholar] [CrossRef] [PubMed]
- Neuhouser, M.L. Dietary flavonoids and cancer risk: Evidence from human population studies. Nutr. Cancer 2004, 50, 1–7. [Google Scholar] [CrossRef]
- Pei, J.; Chen, A.; Zhao, L.; Cao, F.; Ding, G.; Xiao, W. One-pot synthesis of hyperoside by a three-enzyme cascade using a UDP-galactose regeneration system. J. Agric. Food Chem. 2017, 65, 6042–6048. [Google Scholar] [CrossRef]
- Mishra, A.P.; Salehi, B.; Sharifi-Rad, M.; Pezzani, R.; Kobarfard, F.; Sharifi-Rad, J.; Nigam, M. Programmed cell death, from a cancer perspective: An overview. Mol. Diagn. Ther. 2018, 22, 281–295. [Google Scholar] [CrossRef]
- Marfe, G.; Tafani, M.; Indelicato, M.; Sinibaldi-Salimei, P.; Reali, V.; Pucci, B.; Fini, M.; Russo, M.A. Kaempferol induces apoptosis in two different cell lines via Akt inactivation, Bax and SIRT3 activation, and mitochondrial dysfunction. J. Cell. Biochem. 2009, 106, 643–650. [Google Scholar] [CrossRef]
- Imran, M.; Rauf, A.; Shah, Z.A.; Saeed, F.; Imran, A.; Arshad, M.U.; Ahmad, B.; Bawazeer, S.; Atif, M.; Peters, D.G.; et al. Chemo-preventive and therapeutic effect of the dietary flavonoid kaempferol: A comprehensive review. Phytother. Res. 2019, 33, 263–275. [Google Scholar] [CrossRef]
- Kim, K.Y.; Jang, W.Y.; Lee, J.Y.; Jun do, Y.; Ko, J.Y.; Yun, Y.H.; Kim, Y.H. Kaempferol activates G2-checkpoint of the cell cycle resulting in G2-arrest and mitochondria-dependent apoptosis in human acute leukemia jurkat T cells. J. Microbiol. Biotechnol. 2016, 26, 287–294. [Google Scholar] [CrossRef] [PubMed]
- Kim, B.; Jung, J.W.; Jung, J.; Han, Y.; Suh, D.H.; Kim, H.S.; Dhanasekaran, D.N.; Song, Y.S. PGC1α induced by reactive oxygen species contributes to chemoresistance of ovarian cancer cells. Oncotarget 2017, 8, 60299–60311. [Google Scholar] [CrossRef] [PubMed]
- Brown, J.; O’Prey, J.; Harrison, P.R. Enhanced sensitivity of human oral tumours to the flavonol, morin, during cancer progression: Involvement of the Akt and stress kinase pathways. Carcinogenesis 2003, 24, 171–177. [Google Scholar] [CrossRef]
- Manna, S.K.; Aggarwal, R.S.; Sethi, G.; Aggarwal, B.B.; Ramesh, G.T. Morin (3,5,7,2′,4′-Pentahydroxyflavone) abolishes nuclear factor-kappaB activation induced by various carcinogens and inflammatory stimuli, leading to suppression of nuclear factor-kappaB-regulated gene expression and up-regulation of apoptosis. Clin. Cancer Res. 2007, 13, 2290–2297. [Google Scholar] [CrossRef] [PubMed]
- Gupta, S.C.; Phromnoi, K.; Aggarwal, B.B. Morin inhibits STAT3 tyrosine 705 phosphorylation in tumor cells through activation of protein tyrosine phosphatase SHP1. Biochem. Pharmacol. 2013, 85, 898–912. [Google Scholar] [CrossRef]
- Fernandes, S.G.; Gala, K.; Khattar, E. Telomerase inhibitor MST-312 and quercetin synergistically inhibit cancer cell proliferation by promoting DNA damage. Transl. Oncol. 2023, 27, 101569. [Google Scholar] [CrossRef]
- Chung, S.S.; Oliva, B.; Dwabe, S.; Vadgama, J.V. Combination treatment with flavonoid morin and telomerase inhibitor MST-312 reduces cancer stem cell traits by targeting STAT3 and telomerase. Int. J. Oncol. 2016, 49, 487–498. [Google Scholar] [CrossRef]
Sr. No | Title | Study Type | Participants | Drug Used | Phase | Condition | Status | ClinicalTrials.gov Identifier | Reference |
---|---|---|---|---|---|---|---|---|---|
1 | A Phase 1 Study of Imetelstat, a Telomerase Inhibitor, in Children with Refractory or Recurrent Solid Tumors and Lymphomas | Single Group Assignment | 34 participants | Imetelstat sodium | I | Brain Tumor, Lymphoma, Lymphoproliferative Disorder, Small Intestine Cancer, Solid Tumor | Complete (October 2013) | NCT01273090 | [40] |
2 | A Randomized Phase II Study of Imetelstat (GRN163L) In Combination with Paclitaxel (With Or Without Bevacizumab) in Patients With Locally Recurrent Or Metastatic Breast Cancer | Randomized, Parallel Assignment | 166 participants | Imetelstat sodium (300 mg/m2), Bevacizumab (15 mg/kg), and Paclitaxel (90 mg/m2) | II | Locally Recurrent or Metastatic Breast Cancer | Complete (December 2012) | NCT01256762 | [41] |
3 | A Phase II Trial to Evaluate the Activity of Imetelstat (GRN163L) in Patients with Essential Thrombocythemia or Polycythemia Vera Who Require Cytoreduction and Have Failed or Are Intolerant to Previous Therapy or Who Refuse Standard Therapy | Single Group Assignment | 20 participants | Imetelstat (9.4 mg/kg) | II | Essential Thrombocythemia | Complete (April 2015) | NCT01243073 | [42] |
4 | A Phase II Trial to Determine the Effect of Imetelstat (GRN163L) on Patients with Previously Treated Multiple Myeloma | Non-Randomized, Single Group Assignment | 13 participants | Imetelstat (7.5 mg/kg, 9.4 mg/kg), Lenalidomide | II | Multiple Myeloma | Complete (November 2014) | NCT01242930 | [43] |
5 | A Randomized Phase II Study of Imetelstat as Maintenance Therapy After Initial Induction Chemotherapy for Advance Non-small Cell Lung Cancer (NSCLC) | Randomized, Parallel Assignment | 166 participants | Imetelstat (9.4 mg/kg) and Bevacizumab | II | NSCLC | Complete (September 2013) | NCT01137968 | [44] |
Flavonoid | Cell Line | Dose | Results | Reference |
---|---|---|---|---|
EGCG and pEGCG | MCF-7 and MDA-MB-231 breast cancer cell lines. MCF10A cell line (normal control) | Apoptosis induction and hTERT inhibition: EGCG (40 μmol/L) and pEGCG (20 μmol/L) Inhibition of cell proliferation: EGCG (60 μmol/L) pEGCG (40 μmol/L) | pEGCG demonstrated higher potency compared to EGCG in the inhibition of cell proliferation and apoptosis induction in breast cancer cell lines. Inhibition of hTERT was also shown in both cell lines. | [55] |
EGCG | T47D breast cancer cells | 80 µM | A significant decrease in hTERT gene expression causing apoptosis was observed. | [56] |
MST-312 (derivative of EGCG) | MCF-7 and MDA-MB-231 breast cancer cell lines | 0–10 µM | Reduction in telomerase activity, growth arrest, and induction of telomere dysfunction was observed in both cell lines, while reduced expression of TRF2 (telomere protective protein) in MDA-MB-231 cells. | [57] |
MST-312 | APL cells | 0.5,1, and 2 µM | Caspase mediated apoptosis, arrest in G2/M-phase of the cell cycle of APL cells. Along with telomerase inhibitory activity, NF-κB activity was also suppressed. Additionally, hTERT, Bcl-2, survivin, Mcl-1, and c-myc genes were downregulated. | [59] |
EGCG | SCLC cells (H69 and H69VP) | 70 µM | 50–60% Reduction in telomerase, 50 and 70% reduction in caspase 3 and 9, respectively, and block in the S-phase of the cell cycle was observed. | [60] |
EGCG | Eca109 and Ec9706 | 100, 200, or 300 mg/L | EGCG produced apoptosis, reduced the mitochondrial membrane potential, and raised the expression of caspase-3 and led to the inhibition of telomerase. | [61] |
EGCG | Nasopharyngeal carcinoma cell line CNE2 | 100, 200 µg/mL, | Prevented CNE2 cells from proliferating, caused cell cycle block, apoptosis of the cells was promoted, and downregulation of the mRNA and protein expression of hTERT as well as c-Myc protein. | [62] |
EGCG | HEC-18, HEC-18T, HEN-18, HEN-18S | 100 µM | Growth inhibition greater than 90% and induction of apoptosis was observed in HEC-18 and HEN-18. Telomerase was inhibited in all 4 cells. | [63] |
EGCG | OMC-4 and TMCC-1 | 50 and 100 µM | Growth and telomerase inhibition, induction of apoptosis and pKi-67 suppression was observed in both cell lines. | [64] |
EGCG and Retinoic Acid | HeLa and TMCC-1 | EGCG: 100 µM RA: 1 µM | Combination treatment caused inhibition of telomerase, induction of apoptosis and prevented cell proliferation. | [65] |
EGCG and Sulforaphane | SKOV3-ip1 and SKOV3TR-ip2 cells | 20 mmol/L EGCG and 10 mmol/L SFN | Combination treatment led to ovarian cancer cell inhibition, arrest in cell cycle phase G2/M and S, induction of apoptosis and DNA damage, reduction in hTERT and DNA methyltransferase 1 | [66] |
EGCG and EGC | H1299, OECM-1, SAS, WRO, SK-Hep-1, and Hep-3B cells | 10–40 µM | EGCG and EGC caused apoptosis and suppressed hTERT mRNA and promoter activity. | [67] |
EGCG, Cisplatin, and Tamoxifen | 1321N1 and U87-MG cells | EGCG (100 µM) Cisplatin (up to 50 µM) Tamoxifen (up to 20 µM) | Telomerase suppression activity was observed in both glioma cell lines when used in combination. | [68] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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
Parekh, N.; Garg, A.; Choudhary, R.; Gupta, M.; Kaur, G.; Ramniwas, S.; Shahwan, M.; Tuli, H.S.; Sethi, G. The Role of Natural Flavonoids as Telomerase Inhibitors in Suppressing Cancer Growth. Pharmaceuticals 2023, 16, 605. https://doi.org/10.3390/ph16040605
Parekh N, Garg A, Choudhary R, Gupta M, Kaur G, Ramniwas S, Shahwan M, Tuli HS, Sethi G. The Role of Natural Flavonoids as Telomerase Inhibitors in Suppressing Cancer Growth. Pharmaceuticals. 2023; 16(4):605. https://doi.org/10.3390/ph16040605
Chicago/Turabian StyleParekh, Neel, Ashish Garg, Renuka Choudhary, Madhu Gupta, Ginpreet Kaur, Seema Ramniwas, Moyad Shahwan, Hardeep Singh Tuli, and Gautam Sethi. 2023. "The Role of Natural Flavonoids as Telomerase Inhibitors in Suppressing Cancer Growth" Pharmaceuticals 16, no. 4: 605. https://doi.org/10.3390/ph16040605