[2]
Koehn, F.E.; Carter, G.T. The evolving role of natural products in drug discovery. Nat. Rev. Drug Discov., 2005, 4, 206-220.
[3]
Kashman, Y.; Groweiss, A.; Shmueli, J. Latrunculin, a new 2-thiazolidinone macrolide from the marine sponge latrunculiamagnifica. Tetrahedron Lett., 1980, 21, 3629-3632.
[4]
Spector, I.; Shochet, N.R.; Blasberger, D.; Kashman, Y. Latrunculins novel marine macrolides that disrupt microfilament organization and affect cell-growth 1. comparison with cytochalasin-D. Cell Motil. Cytoskeleton, 1989, 13, 127-144.
[5]
Yamazaki, D.; Kurisu, S.; Takenawa, T. Regulation of cancer cell motility through actin reorganization. Cancer Sci., 2005, 96, 379-386.
[6]
El Sayed, K.A.; Youssef, D.T.A.; Marchetti, D. Bioactive natural and semisynthetic latrunculins. J. Nat. Prod., 2006, 69, 219-223.
[7]
Longley, R.E.; McConnell, O.J.; Essich, E.; Harmody, D. Evaluation of marine sponge metabolites for cytotoxicity and signal transduction activity. J. Nat. Prod., 1993, 56, 915-920.
[8]
Konishi, H.; Kikuchi, S.; Ochiai, T.; Ikoma, H.; Kubota, T.; Ichikawa, D.; Fujiwara, H.; Okamoto, K.; Sakakura, C.; Sonoyama, T.; Kokuba, Y.; Sasaki, H.; Matsui, T.; Otsuji, E. Latrunculin a has a strong anticancer effect in a peritoneal dissemination model of human gastric cancer in mice. Anticancer Res., 2009, 29, 2091-2097.
[9]
Kobayashi, S.; Hidaka, S.; Kawamura, H.; Ozaki, M.; Hayase, Y.; Micacocidin, A. B and C, novel antimycoplasma agents from pseudomonassp. -I. taxonomy, fermentation, isolation, physicochemical properties and biological activities. J. Antibiot., 1998, 51, 323-327.
[10]
Carvalho, A.A.; Andrade, L.N.; de Sousa, E.B.V.; de Sousa, D.P. Antitumor phenylpropanoids found in essential oils. BioMed Res. Int., 2015, 2015, 21.
[11]
Lavecchia, A.; Giovanni, C. Virtual screening strategies in drug discovery: A critical review. Curr. Med. Chem., 2013, 20, 2839-2860.
[12]
Kumar, V.; Krishna, S.; Siddiqi, M.I. Virtual screening strategies: Recent advances in the identification and design of anti-cancer agents. Methods, 2015, 71, 64-70.
[13]
Schuster, D.; Wolber, G. Identification of bioactive natural products by pharmacophore-based virtual screening. Curr. Pharm. Des., 2010, 16, 1666-1681.
[14]
Costa, F.G.; Neto, B.R.S.; Gonçalves, R.L.; da Silva, R.A.; de-Oliveira, C.M.A.; Katoc, L.; Freitas, C.S.; Gianninid, M.J.S.M.; da-Silva, J.F.; Soares, C.M.A.; Pereira, M. Alkaloids as inhibitors of malate synthase from Paracoccidioides spp.: Receptor-ligand interaction-based virtual screening and molecular docking studies, antifungal activity, and the adhesion process. Antimicrob. Agents Ch, 2015, 59, 5581-5594.
[15]
de Araújo, R.; Guerra, F.; Lima, E.O.; de Simone, C.; Tavares, J.; Scotti, L.; Scotti, M.T.; de Aquino, T.; de Moura, R.O.; Mendonça, Junior, F.J.B.; Barbosa-Filho, J. Synthesis, Structure-Activity Relationships (SAR) and in silico studies of coumarin derivatives with antifungal activity. Int. J. Mol. Sci., 2013, 14, 1293-1309.
[16]
Félix, M.B.; de-Souza, E.R.; de-Lima, M.C.A.; Frade, D.A.G.; Serafim, V.L.; Rodrigues, K.A.F.; Néris, P.L.N.; Ribeiro, F.F.; Scotti, L.; Scotti, M.T.; de-Aquino, T.M.; Mendonça, Junior, F.J.B.; de Oliveira, M.R. Antileishmanial activity of new thiophene-indole hybrids: Design, synthesis, biological and cytotoxic evaluation, and chemometric studies. Bioorg. Med. Chem., 2016, 24, 3972-3977.
[17]
Ribeiro, F.F.; Mendonça, Junior, F.J.B.; da Silva, M.S.; Scotti, M.T.; Scotti, L. Computational and investigative study of flavonoids active against Typanosoma cruzi and Leishmania spp. Nat. Prod. Commun., 2015, 10, 917-920.
[18]
Chuang, C.; Cheng, T.; Leu, Y.; Chuang, K.; Tzou, S.; Chen, C. Discovery of akt kinase inhibitors through structure-based virtual screening and their evaluation as potential anticancer agents. Int. J. Mol. Sci., 2015, 16, 3202-3212.
[19]
Cebrian-Torrejón, G.; Domenech-Carbó, A.; Scotti, M.T.; Fournet, A.; Figadere, B.; Poupon, E. Experimental and theoretical study of possible correlation between the electrochemistry of canthin-6-one and the anti-proliferative activity against human cancer stem cells. J. Mol. Struct., 2015, 1102, 242-246.
[20]
Berman, H.M.; Westbrook, J.; Feng, Z.; Gililand, G.; Bhat, T.N.; Weissig, H. The protein data bank. Nucleic Acids Res., 2000, 28, 235-242.
[21]
Toropova, A.P.; Toropov, A.A. CORAL software: Prediction of carcinogenicity of drugs by means of the monte carlo method. Eur. J. Pharm. Sci., 2014, 52, 21-25.
[22]
Kar, S.K.; Roy, K. Development and validation of a robust QSAR model for prediction of carcinogenicity of drugs. Indian J. Biochem. Biophys., 2011, 48, 111-122.
[23]
Perumal, P.C.; Sowmya, S.; Pratibha, P.; Vidya, B.; Anusooriya, P.; Starlin, S.; Vasanth, R.; Sharmila, D.J.S.; Gopalakrishnan, V.K. Identification of novel PPARγ agonist from GC-MS analysis of ethanolic extract of Cayratia trifolia (L.): A computational molecular simulation studies. J. Appl. Pharm. Sci., 2014, 4, 6-11.
[24]
Schuster, D.; Wolber, G. Identification of bioactive natural products by pharmacophore-based virtual screening. Curr. Pharm. Des., 2010, 16, 1666-1681.
[25]
Souza, M.V.N.; Pinheiro, A.C.; Ferreira, M.L.; Gonçalves, R.S.B.; Lima, C.H.C. Produtos naturais em fase avançada de testes clínicos no tratamento contra o câncer. Fitos, 2007, 3(2), 25-42.
[26]
Bayala, B.; Bassole, I.H.N.; Scifo, R.; Gnoula, C.; Morel, L.; Lobaccaro, J.A.; Simpor, J. Anticancer activity of essential oils and their chemical components - A review. Am. J. Cancer Res., 2014, 4, 591-607.
[27]
Instituto Nacional de Câncer José Alencar Gomes da Silva (INCA). Estimativa | 2016 Incidência de câncer no Brasil; Ministério da Saúde, 2015, p. 122.
[28]
Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the last 25 years. J. Nat. Prod., 2007, 70, 461-477.
[29]
Singh, A.; Mhlongo, N.; Es Soliman, M. Anti-cancer glycosidase inhibitors from natural products: A computational and molecular modelling perspective. Anti-Cancer Agent. Med. Chem., 2015, 15(8), 933-946.
[30]
Lake, B. Synthesis & pharmacological investigation of 4-hydroxy coumarin derivatives & shown as anti-coagulant. Food Chem. Toxicol., 1999, 3, 412-423.
[31]
Lacy, A.; O’Kennedy, R. Studies on coumarins and coumarin-related compounds to determine their therapeutic role in the treatment of cancer. Curr. Pharm. Des., 2004, 10(30), 3797-3811.
[32]
Rohini, K.; Srikumar, P.S. In silico approach of anticancer activity of phytochemical coumarins against cancer target JNKS. Int. J. Pharm. Pharm. Sci., 2013, 5, 741-742.
[33]
Chilin, A.; Battistutta, R.; Bortolato, A.; Cozza, G.; Zanatta, S.; Poletto, G.; Mazzorana, M.; Zagotto, G.; Uriarte, E.; Guiotto, A.; Pinna, L.A.; Meggio, F.; Moro, S. Coumarin as attractive Casein Kinase 2 (CK2) inhibitor scaffold: An integrate approach to elucidate the putative binding motif and explain structure-activity relationships. J. Med. Chem., 2008, 51, 752-759.
[34]
Nolan, K.A.; Doncaster, J.R.; Dunstan, M.S.; Scott, K.A.; Frenkel, A.D.; Siegel, S.; Ross, D.; Barnes, J.; Levy, C.; Leys, C.; Whitehead, R.C.; Stratford, I.J.; Bryce, R.A. Synthesis and Biological evaluation of coumarin-based inhibitors of NAD(P)H: Quinone oxidoreductase-1 (NQO1). J. Med. Chem., 2009, 52, 7142-7156.
[35]
Zwergela, C.; Czepukojce, B.; Evain-Banad, E.; Xuf, Z.; Stazia, G.; Morig, M.; Patsilinakosa, A.; Maia, A.; Bottaa, B.; Ragnoa, R. Novel coumarin- and quinolinone-based polycycles as cell division cycle 25-A and -C phosphatases inhibitors induce proliferation arrest and apoptosis in cancer cells. Eur. J. Med. Chem., 2017, 134, 316-333.
[36]
Wagner, E.F.; Nebreda, A.R. Signal integration by JNK and p38 MAPK pathways in cancer development. Nat. Rev. Cancer, 2009, 9(8), 537-549.
[37]
Rotig, A.; Mollet, J.; Rio, M.; Munnuch, A. Infantile and pediatric quinone deficiency diseases. Mitochondrion, 2007, 7, S112-S121.
[38]
Babiaka, B.B.; Ntie-Kang, F.; Ndingkokhar, B.; Mbah, J.A.; Sipplc, W.; Yong, J.N. The chemistry and bioactivity of Southern African flora II: Flavonoids, quinones and minor compound classes. RSC Adv., 2015, 5, 57704-57720.
[39]
Siegel, D.; Gustafson, D.L.; Dehn, D.L.; Han, J.Y.; Boonchoong, P.; Berliner, L.J.; Ross, D. Mol. Pharmacol., 2004, 65, 1238-1247.
[40]
Colucci, M.A.; Reigan, P.; Siegel, D.; Chilloux, A.; Ross, D.; Moody, C. Synthesis and evaluation of 3-aryloxymethyl-1,2-
dimethylindole-4,7-diones as mechanism-based inhibitors of
NAD(P)H:quinone oxidoreductase 1 (NQO1) activity. J. Med. Chem., 2007, 50, 5780-5789.
[41]
Nolan, K.A.; Humphries, M.P.; Barnes, J.; Doncaster, J.R.; Caraher, M.C.; Tirelli, N.; Bryce, R.A.; Whitehead, R.C.; Stratford, I.J. Triazoloacridin-6-ones as novel inhibitors of the quinone oxidoreductases NQO1 and NQO2. Bioorg. Med. Chem., 2010, 18, 696-706.
[42]
Bian, J.; Qian, X.; Deng, B.; Xu, X.; Guo, X.; Wang, Y.; Li, X.; Sun, H.; You, Q.; Zhang, X. Discovery of NAD(P)H: Quinone oxidoreductase 1 (NQO1) inhibitors with novel chemical scaffolds by shape-based virtual screening combined with cascade dockin7. RSC Advances, 2015, 5(61), 49471-49474.
[43]
Morrison, H.; Jernstrom, B.; Nordenskjold, M.; Thor, H.; Orrenius, S. Induction of DNA damage by menadione (2-methyl-1,4-naphthoquinone) in primary cultures of rat hepatocytes. Biochem. Pharmacol., 1984, 33, 1763-1769.
[44]
Han, Y.; Shen, H.; Carr, B.I.; Wipf, P.; Lazo, J.S.; Pan, S.S. NAD(P)H: Quinone oxidoreductase-1-dependent and -independent cytotoxicity of potent quinone Cdc25 phosphatase inhibitors. J. Pharmacol. Exp. Ther., 2004, 309, 64-70.
[45]
Velišek, J.; Davidek, J.; Cejpek, K. Biosynthesis of food constituents: Natural pigments. Part 1-A review. Czech J. Food Sci., 2007, 25, 291-315.
[46]
Hasinoff, B.B.; Liang, H.; Wu, X.; Guziec, L.J.; Guziec, F.S., Jr; Marshall, K.; Yalowich, J.C. The structure-based design, synthesis and biological evaluation of DNA-binding bisintercalating bisanthrapyrazole anticancer compounds. Bioorg. Med. Chem., 2008, 16, 3959-3968.
[47]
Tamaian, R.; Niculescu, V.; Anghel, M. In silico predictions for improving permeability properties of principal anticancer anthracyclines through structural modification. Bulletin UASVM. Vet. Med., 2010, 67(1), 329-336.
[48]
Rudolph, J. Targeting Cdc25 Phosphatases in Cancer Therapy.In: Checkpoint Controls and Targets in Cancer Therapy; Siddik, Z.H., Ed.; Springer Science, LLC, 2009, pp. 261-269.
[49]
Lavecchia, A.; Giovanni, C.D.; Novellino, E. Expert Opin. Ther. Pat., 2010, 20, 405-425.
[50]
Cao, S.; Forster, C.; Brisson, M.; Lazo, J.S.; Kingston, D.G.I. Halenaquinone and xestoquinone derivatives, inhibitors of Cdc25B phosphatase from a Xestospongia sp. Bioorg. Med. Chem., 2005, 13, 999-1003.
[51]
Park, J.I.; Kwak, J.Y. The role of peroxisome proliferatoractivated receptors in colorectal cancer. PPAR Res., 2012, 1-2.
[52]
Lavecchia, A.; Di Giovanni, C.; Pesapane, A.; Montuori, N.; Ragno, P.; Martucci, N.M.; Masullo, M.; De Vendittis, E.; Novellino, E. Discovery of new inhibitors of Cdc25B dual specificity phosphatases by structure-based virtual screening. J. Med. Chem., 2012, 55, 4142-4158.
[53]
Ge, Y.S.; Han, Q.Q.; Duan, W.; Zhang, J.Q.; Chen, K.; Wan, J.J.; Liu, Y.; Liu, D. Discovery of Cdc25A lead inhibitors with novel chemotype by virtual screening: Application of pharmacophore modeling based on training set with unique limited components. ChemMedChem, 2017, 438-447.
[54]
Ham, S.W.; Carr, B.I. Cell division cycle 25 (Cdc25) phosphatase inhibitors as antitumor agents. Drug Des. Rev, 2004, 1, 123-132.
[55]
Contour-Galcera, M.; Sidhu, A.; G. Prévost, D. Bigg, B. Ducommun. Pharmacol. Ther, 2007, 115, 1-12.
[56]
Kumar, R.; Singh, M. Tannins: Their adverse role in ruminant nutrition. J. Agric. Food Chem., 1984, 32(3), 447-453.
[57]
Masoumi-Ardakani, Y.; Mandegary, A.; Esmaeilpour, K.; Najafipour, H.; Sharififar, F.; Pakravanan, M.; Ghazvini, H. Chemical
composition, anticonvulsant activity, and toxicity of essential oil
and methanolic extract of. Elettaria cardamomum. Plant. Med, 2016, 1482-1486.
[58]
Bhattacharjee, B.; Chatterje, J. Identification of proapoptopic, anti-inflammatory, antiproliferative, anti-invasive and anti-angiogenic targets of essential oils in cardamom by dual reverse virtual screening and binding pose analysis. J. Cancer Prev., 2013, 14(6), 3735-3742.
[59]
Pulliah, T. Encyclopaedia of World Medicinal Plants; New Delhi Regency Publication, 2006, p. 492.
[60]
Kumar, D.; Kumar, S.; Grupta, J.; Arva, R.; Grupta, A. A review on chemical and biological properties of Cayratia trifolia Linn. (Vitaceae). Pharmacogn. Rev., 2011, 5, 184-188.
[61]
Gour, K.; Patni, V. Gas chromatography-massa spectrometry (gcms-qt 2010) analysis of methanolic extracts of Cayratia trifolia (L.) Domin (family: vitaceae): A plant from semi-arid regions of rajasthan, India. J. Liq. Chromatogr. Relat. Technol., 2012, 35, 1616-1626.
[62]
Gupta, A.; Bhardwaj, A.; Gupta, J.; Bagchi, A. Anti-implantation activity of petroleum ether extract of leaves of Cayratia trifolia Linn. On female albino rat. Asian Pac. J. Trop. Biomed., 2012, 2, S197-S199.
[63]
Perumal, P.C.; Sowmya, S.; Pratibha, P.; Vidya, B.; Anusooriya, P.; Starlin, S.; Vasanth, R.; Sharmila, D.J.S.; Gopalakrishnan, V.K. Identification of novel PPARγ agonist from GC-MS analysis of
ethanolic extract of Cayratia trifolia (L.): A computational molecular
simulation studies. J. Appl. Pharm. Sci., 2014, 4, 006-011.
[64]
Park, J.I.; Kwak, J.Y. The role of peroxisome proliferator-activated receptors in colorectal cancer. PPAR Res., 2012, 1-12.
[65]
Sikka, S.; Chen, L.; Sethi, G. PremKumar, A. Targeting PPARγ signaling cascade for the prevention and treatment of prostate cancer. PPAR Res., 2012, 1-14.
[66]
Akbarpour, V.; Hemmati, K.; Sharifani, M. Physical and chemical properties of pomegranate (Punica granatum L) fruit in maturation stage. Am.-Eurasian J. Agric. Environ. Sci., 2009, 6, 411-416.
[67]
Middha, S.K.; Usha, T.; Pande, V. Pomegranate peel attenuates hyperglycemic effects of alloxan-induced diabetic rats. EXCLI J., 2014, 13, 223-224.
[68]
Putnik, P.; Kresoja, Ž.; Bosiljkov, T.; Režek Jambrak, A.; Barba, F.J.; Lorenzo, J.M.; Roohinejad, S.; Granato, D.; Žuntar, I.; Bursać Kovačević, D. Comparing the effects of thermal and non-thermal technologies on pomegranate juice quality: A review. Food Chem., 2019, 279, 150-161.
[69]
Usha, T.; Goyal, A.K.; Lubna, S.; Prashanth, H.P.; Mohan, T.M.; Pande, V.; Middha, S.K. Identification of anti-cancer targets of eco-friendly waste Punica granatum peel by dual reverse virtual screening and binding analysis. Asian Pac. J. Cancer Prev., 2014, 15.
[70]
Brandão, H.N.; David, J.P.; Couto, R.D.; Nascimento, J.A.P.; David, J.M. Química e farmacologia de quimioterápicos antineoplásicos derivados de plantas. Quim. Nova, 2010, 33, 1359-1369.
[71]
Verpoorte, R. Methods for the structure elucidation of alkaloids. J. Nat. Prod., 1986, 49, 1-25.
[72]
Cordell, G.A.; Quinn-Beattie, M.L.; Farnsworth, N.R. The potential of alkaloids in drug discovery. Phytother. Res., 2001, 15, 183-205.
[73]
Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the last 25 years. J. Nat. Prod., 2007, 70, 461-477.
[74]
Duflos, A.; Kruczynski, A.; Barret, J.M. Novel aspects of natural and modified vinca alkaloids. Curr. Med. Chem. Anticancer Agents, 2002, 2, 55-70.
[75]
Sarno, S.; Moro, S.; Meggio, F.; Zagotto, G.; Dal Ben, D.; Ghisellini, P.; Battistutta, R.; Zanotti, G.; Pinna, L.A. Toward the rational design of protein kinase casein kinase-2 inhibitors. Pharmacol. Ther., 2002, 93, 159-168.
[76]
Qiu, S.; Sun, H.; Zhang, A.H.; Xu, H.Y.; Yan, G.L.; Han, Y.; Wang, X.J. Natural alkaloids: Basic aspects, biological roles, and future perspectives. Chin. J. Nat. Med., 2014, 12, 401-406.
[77]
Konkimalla, V.B.; Suhas, V.L.; Chandra, N.R.; Gebhart, E.; Efferth, T. Diagnosis and therapy of oral squamous cell carcinoma. Expert Rev. Anticancer Ther., 2007, 7, 317-329.
[78]
El-Sayed, K.A.; McChesney, J.D.; Halim, A.F.; Zaghloul, A.M.; Lee, I.S. A study of alkaloids in Veratrum viride Aiton. Int. J. Pharmacogn, 1996, 34, 161-173.
[79]
Kupchan, S.M.; Zimmerman, J.H.; Afonso, A. The alkaloids and taxonomy of Veratrum and related genera. Lloydia, 1961, 24, 1-26.
[80]
Agrawal, P.K.; Srivastava, S.K.; Gaffield, W. Alkaloids. In: Pelletier
SW (ed) Chemical and biological perspectives, Springer, New
York. 1991, 43-296.
[81]
Honerjager, P. Cardioactive substances that prolong the open state of sodium channels. Rev. Physiol. Biochem. Pharmacol., 1982, 92, 1-74.
[82]
Gaffield, W.; Keeler, R.F. Implication of C-5, C-6 unsaturation asa key structural factor in steroidal alkaloid-induced mammalianteratogenesis. Experientia, 1993, 9, 922-924.
[83]
Gaffield, W.; Keeler, R.F. Steroidal alkaloid teratogens: Molecular probes for investigation of craniofacial malformations. J. Toxicol. Toxin Rev., 1996, 15, 303-326.
[84]
Zhang, F.; McLellan, J.S.; Ayala, A.M.; Leahy, D.J.; Linhardt, R.J. Kinetic and structural studies on interactions between heparin or heparan sulfate and proteins of the Hedgehog signaling pathway. Biochemistry, 2007, 46, 3933-3941.
[85]
Heller, E.; Hurchla, M.A.; Xiang, J.; Su, X.; Chen, S.; Schneider, J.; Joeng, K.; Vidal, M.; Goldberg, L.; Deng, H.; Hornick, M.C.; Prior, J.L.; Piwnica-Worms, D.; Long, F.; Cagan, R.; Weilbaecher, K.N. Hedgehog signaling inhibition blocks growth of resistant tumors through effects on tumor microenvironment. Cancer Res., 2012, 72, 897-907.
[86]
Bar, E.E.; Chaudhry, A.; Farah, M.H.; Eberhart, C.G. Hedgehog signaling promotes medulloblastoma survival via Bc/II. Am. J. Pathol., 2006, 170, 347-355.
[87]
Kiselyov, A.S. Small molecule drug discovery. Targeting the hedgehog signaling pathway with small molecules. Anticancer. Agents Med. Chem., 2006, 6, 445-449.
[88]
Sanchez, P.; Ruizi, A.A. In vivo inhibition of endogenous brain tumors through systemic interference of Hedgehog signaling in mice. Mech. Dev., 2005, 122, 223-230.
[89]
Lauth, M.; Bergstroem, A.; Shimokawa, T.; Toftgard, R. Inhibition of GLI-mediated transcription and tumor cell growth by smallmolecule antagonists. Proc. Natl. Acad. Sci. USA, 2007, 104(20), 8455-8460.
[90]
Khanfar, M.A.; El Sayed, K.A. The Veratrum alkaloids jervine, veratramine, and their analogues as prostate cancer migration and proliferation inhibitors: biological evaluation and pharmacophore modeling. Med. Chem., 2013, 22(10), 4775-4786.
[91]
Kozielewicz, P.; Paradowska, K. ErićIwona, S.; Zloh, W. Insights into mechanism of anticancer activity of pentacyclic oxindole alkaloids of Uncaria tomentosa by means of a computational reverse virtual screening and molecular docking approach. Monatsh. Chem., 2014, 145(7), 1201-1211.
[92]
Saraswati, S.; Kanuajia, P.K.; Kumar, S.; Kumar, R.; Alhaider, A.A. Tylophorine, a phenanthraindolizidine alkaloid isolated from Tylophora indica exerts antiangiogenic and antitumor activity by targeting vascular endothelial growth factor receptor 2-mediated angiogenesis. Mol. Cancer, 2013, 12, 82.
[93]
Zhang, F.; McLellan, J.S.; Ayala, A.M.; Leahy, D.J.; Linhardt, R.J. Kinetic and structural studies on interactions between heparin or heparan sulfate and proteins of the Hedgehog signaling pathway. Biochemistry, 2007, 46, 3933-3941.
[94]
Lee, M.D.; Antczak, C.; Li, Y.; Sirotnak, F.M.; Bornmann, W.G.; Scheinberg, D.A. A new human peptide deformylase inhibitable by actinonin. Biochem. Biophys. Res. Commun., 2003, 312, 309-315.
[95]
Potter, S.M.; Baum, J.A.; Teng, H.; Stillman, R.J.; Shay, N.F.; Erdman, Jr, J.W. Soy protein and isoflavones: their effects on blood lipids and bone density in postmenopausal women. Am. J. Clin. Nutr., 1998, 68, 1375-1379.
[96]
Havsteen, B.H. The biochemistry and medical significance of the flavonoids. Pharmacol. Ther., 2002, 96, 67-202.
[97]
Kinoshita, T.; Lepp, Z.; Kawai, Y.; Terao, J.; Chuman, H. An integrated database of flavonoids. Biofactors, 2006, 26, 179-188.
[98]
Scotti, L.; Mendonca, Junior, F.J.B.; Diogo, R.M.M.; da-Silva, M.S. PITTA, I.R.; Scotti, M.T. SAR, QSAR and docking of anticrrecancer flavonoids and variants: A review. Curr. Top. Med. Chem., 2013, 12, 2785-2809.
[99]
Liu, P.; Duan, J.A.; Hua, Y.Q.; Tang, Y.P.; Yao, X.; Su, S.L. J. Ethnopharmacol., 2011, 133(2), 591-597.
[100]
Su, S.L.; Yu, L.; Hua, Y.Q.; Duan, J.A.; Deng, H.S.; Tang, Y.P.; Lu, Y.; Ding, A.W. Screening and analyzing the potential bioactive components from Shaofu Zhuyu decoction, using human umbilical vein endothelial cell extraction and high-performance liquid chromatography coupled with mass spectrometry. Biomed. Chromatogr., 2008, 22(12), 1385-1392.
[101]
Liu, L.; Ma, H.; Tang, Y.; Chen, W.; Lu, Y.; Guo, J.; Duan, J.A. Discovery of estrogen receptor α modulators from natural compounds in Si-Wu-Tang series decoctions using estrogen-responsive MCF-7 breast cancer cells. Bioorg. Med. Chem. Lett., 2012, 22, 154-163.
[102]
Li, F.J.; Ye, L.; Lin, S.M.; Leung, L.K. Dietary flavones and flavonones display differential effects on aromatase (CYP19) transcription in the breast cancer cells MCF-7. Mol. Cell. Endocrinol., 2011, 344, 51-58.
[103]
Awasthi, M.; Singh, S.; Pandey, P.V.; Dwivedi, U.N. Molecular docking and 3DQSAR-based virtual screening of flavonoids as potential aromatase inhibitors against estrogen-dependent breast cancer. J. Biomol. Struct. Dyn., 2015, 33, 804-819.
[104]
Paoletta, S.; Steventon, G.B.; Wildeboer, D.; Ehrman, T.M.; Hylands, P.J.; Barlow, D.J. Screening of herbal constituents for aromatase inhibitory activity. Bioorg. Med. Chem., 2008, 16, 8466-8470.
[105]
Brueggemeier, R.W.; Hackett, J.C.; Diaz-Cruz, E.S. Aromatase inhibitors in the treatment of breast cancer. Endocr. Rev., 2005, 26, 331-345.
[106]
Xie, F.; Lang, Q.Y.; Zhou, M.; Zhang, H.X.; Zhang, Z.S.; Zhang, Y.F.; Wan, B.; Huang, Q.; Yu, L. The dietary flavonoid luteolin inhibits Aurora B kinase activity and blocks proliferation of cancer cells. Eur. J. Pharm. Sci., 2012, 46(5), 388-396.
[107]
Dar, A.A.; Goff, L.W.; Majid, S.; Berlin, J.; El-Rifai, W. Aurora kinase inhibitors–rising stars in cancer therapeutics? Mol. Cancer Ther., 2010, 9, 268-278.
[108]
Trott, O.; Olson, A.J. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem., 2010, 31, 455-461.
[109]
Shinde, V.; Dhalwal, K.; Paradkar, A.R.; Mahadik, K.R.; Kadam, S.S. Evaluation of in vitro antioxidant activity of human placental extract. Pharmacologyonline, 2006, 3, 172-179.
[110]
Schinella, G.R.; Tournier, H.A.; Prieto, J.M.; Mordujovich de Buschiazzo, P.; Rios, J.L. Antioxidant activity of anti-inflammatory plant extracts. Life Sci., 2002, 70, 1023-1033.
[111]
Deryugina, E.I.; James, P.Q. Chapter 2. Chick embryo chorioallantoic membrane models to quantify angiogenesis induced by inflammatory and tumor cells or purified effector molecules. Methods Enzymol., 2008, 444, 21-41.
[112]
Somani, Z.; Bhattacharjee, R.; Chodankar, M.; Abhilash, N. In vitro antioxidant, anti-inflammatory, inovo anti-angiogenic activities and virtual screening of phyto constituents of Chromolena odorata. Int. J. Curr. Res., 2014, 6, 7766-7771.
[113]
Rungsardthong, K.; Mares-Samano, S.; Penny, J. Virtual screening of ABCC1 transporter nucleotidebinding domains as a therapeutic target in multidrug resistant cancer. Bioinformation, 2012, 8, 907-911.
[114]
Dong, X.; Zhou, X.; Jing, H.; Chen, J.; Liu, T.; Yang, B.; He, Q.; Hu, Y. Pharmacophore identification, virtual screening and biological evaluation of prenylated flavonoids derivatives as PKB/Akt1 inhibitors. Eur. J. Med. Chem., 2011, 46, 5949-5958.
[115]
Chappell, J. Biochemistry and molecular biology of the isoprenoid biosynthetic pathway in plants. Annu. Rev. Plant Biol., 1995, 46, 521-547.
[116]
Mcmorris, T.C. Discovery and development of sesquiterpenoid derived hydroxymethylacylfulvene: A new anticancer drug. Bioorg. Med. Chem., 1999, 7, 881-886.
[117]
Cowan, M.M. Plant products as antimicrobial agents. Clin. Microbiol. Rev., 1999, 12(4), 564-582.
[118]
Konstantinopoulou, M.; Karioti, A.; Skaltsas, S.; Skaltsa, H. Sesquiterpene lactones from anthemisa ltissima and their anti Helicobacter pylori activity. J. Nat. Prod., 2003, 66, 699-702.
[119]
Hamzaa, R.; Osman, N. Using of coffee and cardamom mixture to ameliorate oxidative stress induced in γ-irradiated rats. Biochem. Anal. Biochem., 2012, 1, 113-119.
[120]
Moteki, H.; Hibasami, H.; Yamada, Y.; Katsuzaki, H.; Imai, K.; Komiya, T. Specific induction of apoptosis by 1,8-cineole in two human leukemia cell lines, but not a in human stomach cancer cell line. Oncol. Rep., 2002, 9, 757-760.
[121]
Ernst, P.B.; Gold, B.D. The disease spectrum of Helicobacter pylori: The immunopathogenesis of gastroduodenal ulcer and gastric cancer. Annu. Rev. Microbiol., 2000, 54, 615-640.
[122]
Isik, A.; Okan, I.; Firat, D.; Yilmaz, B.; Akcakaya, A.; Sahin, M. A new prognostic strategy for gastric carcinoma: Albumin level and metastatic lymph node ratio. Minerva Chir., 2014, 69, 147-153.
[123]
Ahmad, A.; Govil, Y.; Frank, B.B. Gastric mucosa-associated lymphoid tissuelymphoma. Am. J. Gastroenterol., 2003, 98, 975-986.
[124]
Isik, A.; Alimoglu, O.; Okan, I.; Bas, G.; Turgut, H.; Sahin, M. Dieulafoy lesion in the stomach. Case Rep. Gastroenterol., 2008, 2(3), 469-473.
[125]
Legrain, P.; Strosberg, D. Protein interaction domain mapping for the selection of validated targets and lead compounds in the anti-infectious area. Curr. Pharm. Des., 2002, 8, 1189-1198.
[126]
Cremades, N.; Bueno, M.; Toja, M.; Sancho, J. Towards a new therapeutic target: Helicobacter pylori flavodoxin. Biophys. Chem., 2005, 115, 267-276.
[127]
Dawood, M.; Fatima, N.; Mumtaz, A.; Rehman, S.; Shazadi, I.; Mahmood, Q.; Muhammad, S.A. Molecular docking studies of sesquiterpenoids against Helicobacter pylori peptide deformylase. Br. J. Pharm. Res., 2016, 10(3), 1-7.
[128]
Mazel, D.; Pochet, S.; Marliere, P. Genetic characterization of polypeptide deformylase, a distinctive enzyme of eubacterial translation. EMBO J., 1994, 13(4), 914-923.
[129]
Lee, M.D.; Antczak, C.; Li, Y.; Sirotnak, F.M.; Bornmann, W.G.; Scheinberg, D.A. A new human peptide deformylase inhibitable by actinonin. Biochem. Biophys. Res. Commun., 2003, 312, 309-315.
[130]
Serero, A.; Giglione, C.; Sardini, A.; Martinez-Sanz, J.; Meinnel, T. An unusual peptide deformylase features in the human mitochondrial N-terminal methionine excision pathway. J. Biol. Chem., 2003, 278, 52953-52963.
[131]
Zhang, Y.S.; Talalay, P.; Cho, C.G.; Posner, G.H. A major inducer of anticarcinogenic protective enzymes from broccoli - isolation and elucidation of structure. Proc. Natl. Acad. Sci. USA, 1992, 89, 2399-2403.
[132]
Scotti, L.; Scotti, M.T.; Ishiki, H.M.; Ferreira, M.J.P.; Emerenciano, V.P.; Menezes, C.M.D.; Ferreira, E.I. Quantitative elucidation of the structure-bitterness relationship of cynaropicrin and grosheimin derivatives. Food Chem., 2007, 105, 77-83.
[133]
Schmidt, T.J. Structure-activity relationships of sesquiterpene lactones. Stud. Nat. Prod. Chem, 2006, 33, 309-392.
[134]
Schmidt, T.J.; Heilmann, J. Quantitative structure-cytotoxicity relationships of sesquiterpene lactones derived from partial charge (Q)-based fractional accessible surface area descriptors (Q_frASAs) quant. Struct.-. Act. Relationships, 2002, 21, 276-287.
[135]
Molecular Operating Environment (MOE), 2013.08; Chemical
Computing Group ULC, 1010 Sherbooke St. West, Suite #910,
Montreal, QC, Canada, H3A 2R7. 2017.
[136]
Scotti, M.T.; Fernandes, M.B.; Ferreira, M.J.P.; Emerenciano, V.P. Quantitative structure-activity relationship of sesquiterpene lactones with cytotoxic activity. Bioorg. Med. Chem., 2007, 15, 2927-2934.
[137]
Fernandes, M.B.; Scotti, M.T.; Ferreira, M.J.P.; Emerenciano, V.P. Use of self-organizing maps and molecular descriptors to predict the cytotoxic activity of sesquiterpene lactones. Eur. J. Med. Chem., 2008, 43, 2197-2205.
[139]
Schomburg, C.; Schuehly, W.; Da-Costa, F.B.; Klempnauer, K.H.; Schmidt, T.J. Natural sesquiterpene lactones as inhibitors of Myb-dependent gene expression: Structure-activity relationships. Eur. J. Med. Chem., 2013, 63, 313-320.
[140]
Horiuchi, D.; Huskey, N.E.; Kusdra, L.; Wohlbold, L.; Merrick, K.A. Chemical-genetic analysis of cyclin dependent kinase 2 function reveals an important role in cellular transformation by multiple oncogenic pathways. Proc. Natl. Acad. Sci. USA, 2012, 109, E1091-E1027.
[141]
Ganatra, S.H.; Suchak, A.S. Inhibition studies of naturally occurring terpene based compounds with cyclin-dependent kinase 2 enzyme. J. Comput. Sci. Syst. Biol., 2012, 5, 68-73.
[142]
Harrigan, G.G.; Bolzani, V.S.; Gunatilaka, L.; Kingston, D.G.I. Kaurane and trachylobane diterpenes from Xylopia aethiopica. Phytochemistry, 1994, 36, 109-113.
[143]
Wang, L.; Li, D.; Wang, C.; Zhang, Y.; Xu, J. Recent progress in the development of natural ent-kaurane diterpenoids with anti-tumor activity. Mini Rev. Med. Chem., 2011, 11, 910-919.
[144]
Sul, Y.H.; Lee, M.S.; Cha, E.Y.; Thuong, P.T.; Khoi, N.M.; Song, I.S. An ent-kaurane diterpenoid from croton tonkinensis induces apoptosis by regulating AMP-activated protein kinase in SK-HEP1 human hepatocellular carcinoma cells. Biol. Pharm. Bull., 2013, 36, 158-164.
[145]
Hueso-Falcón, I.; Girón, N.; Velasco, P.; Amaro-Luis, J.M.; Ravelo, A.G.; De las Heras, B.; Hortelano, S.; Esteves-Braun, A. Synthesis and induction of apoptosis signaling pathway of ent-kaurane derivatives. Bioorg. Med. Chem., 2010, 18, 1724-1735.
[146]
Kar, S.; Palit, S.; Ball, W.B.; Das, P.K. Carnosic acid modulates Akt/IKK/NF-KB signaling by PP2A and induces intrinsic and extrinsic pathway mediated apoptosis in human prostate carcinoma PC-3 cells. Apoptosis, 2012, 17, 735-747.
[147]
Wu, D.G.; Yu, P.; Li, J.W.; Jiang, P.; Sun, J.; Wang, H.Z.; Zhang, L.D.; Wen, M.B.; Bie, P. Apigenin potentiates the growth inhibitory effects by IKK-β-mediated NF-kB activation in pancreatic cancer cells. Toxicol. Lett., 2014, 224, 157-164.
[148]
Yingkun, N.; Zhenyu, W.; Jing, L.; Xiuyun, L.; Huimin, Y. Stevioside protects LPS-induced acute lung injury in mice. Inflammation, 2012, 36, 242-250.
[149]
Xiang, Q.; Liu, Q.; Xu, L.; Qiao, Y.; Wang, Y.; Liu, X. Carnosic acid protects biomolecules from free radical-mediated oxidative damage in vitro. Food Sci. Biotechnol., 2013, 22, 1381-1388.
[150]
Scotti, L.; Ishiki, H.; Mendonça, Junior, F.J.B.; Santos, P.F.; Tavares, J.F. SILVA, M.S.; Scotti, M.T. Theoretical research into anticancer activity of diterpenes isolated from the paraiban flora. Nat. Prod. Commun., 2014, 9, 911-914.
[151]
Scotti, L.; Scotti, M.T.; Pasqualotto, K.F.M.; Bolzani, V.S.; Ferreira, E.I. Molecular physicochemical parameters predicting antioxidant activity of Brazilian natural products. Braz. J. Pharmacog, 2009, 19, 908-913.
[152]
Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. Autodock4 and AutoDockTools4: Automated docking with selective receptor flexiblity. J. Comput. Chem., 2009, 16, 2785-2791.
[153]
Scotti, L.; Ferreira, E.I.; Silva, M.S.; Scotti, M.T. Chemometric studies on natural products as potential inhibitors of the NADH oxidase from Trypanosoma cruzi using the VolSurf approach. Molecules, 2010, 15, 7363-7377.
[154]
Sharaf, M.A.; Illman, D.L.; Kowalski, B.R. Chemometrics; John Wiley & Sons: New York, 1986, pp. 1-336.
[155]
Ooms, F. Molecular modeling and computer aided drug design. Examples of their applications in medicinal chemistry. Curr. Med. Chem., 2000, 7, 141-158.
[156]
Scotti, L.; Scotti, M.T.; Ishiki, H.; Mendonça, Junior, F.J.B.; Santos, P.F.; Tavares, J.F.; Silva, M.S. Prediction of anticancer activity of diterpenes isolated from the Paraiban flora through a PLS model and molecular surfaces. Nat. Prod. Commun., 2014, 9, 609-612.