Review Article

白藜芦醇:治疗黑色素瘤的新药物?

卷 28, 期 4, 2021

发表于: 12 December, 2019

页: [687 - 711] 页: 25

弟呕挨: 10.2174/0929867326666191212101225

价格: $65

摘要

黑素瘤是最具生命威胁和侵袭性的皮肤恶性肿瘤。黑色素瘤的发病率稳步上升。转移性黑色素瘤对标准的抗黑色素瘤治疗(如化疗)具有极大的耐药性,并且转移性黑色素瘤患者的5年生存率低于10%。凋亡、血管生成和自噬在黑色素瘤病理生理学中的作用已被证实。因此,寻找能够提高受试者生活质量、减少治疗耐药性和不良反应的辅助治疗方法迫在眉睫。白藜芦醇是一种多酚成分,存在于葡萄和一些植物中,具有抗癌特性,因为它可以诱导肿瘤细胞凋亡,并抗血管生成剂防止转移。然而,还需要进行更多的临床试验来证明白藜芦醇的功效。在此,我们首次总结了白藜芦醇在黑色素瘤中的抗癌活性。

关键词: 黑色素瘤,白藜芦醇,治疗,天然化合物,细胞凋亡,血管生成

[1]
Houghton, A.N.; Polsky, D. Focus on melanoma. Cancer Cell, 2002, 2(4), 275-278.
[http://dx.doi.org/10.1016/S1535-6108(02)00161-7] [PMID: 12398891]
[2]
Singh, S.; Zafar, A.; Khan, S.; Naseem, I. Towards therapeutic advances in melanoma management: An overview. Life Sci., 2017, 174, 50-58.
[http://dx.doi.org/10.1016/j.lfs.2017.02.011] [PMID: 28238718]
[3]
Tong, L.X.; Young, L.C. Nutrition: the future of melanoma prevention? J. Am. Acad. Dermatol., 2014, 71(1), 151-160.
[http://dx.doi.org/10.1016/j.jaad.2014.01.910] [PMID: 24656410]
[4]
Mirzaei, H.; Sahebkar, A.; Avan, A.; Jaafari, M.R.; Salehi, R.; Salehi, H.; Baharvand, H.; Rezaei, A.; Hadjati, J.; Pawelek, J.M.; Mirzaei, H.R. Application of mesenchymal stem cells in melanoma: a potential therapeutic strategy for delivery of targeted agents. Curr. Med. Chem., 2016, 23(5), 455-463.
[http://dx.doi.org/10.2174/0929867323666151217122033] [PMID: 26674785]
[5]
Zapas, J.L.; Coley, H.C.; Beam, S.L.; Brown, S.D.; Jablonski, K.A.; Elias, E.G. The risk of regional lymph node metastases in patients with melanoma less than 1.0 mm thick: recommendations for sentinel lymph node biopsy. J. Am. Coll. Surg., 2003, 197(3), 403-407.
[http://dx.doi.org/10.1016/S1072-7515(03)00432-0] [PMID: 12946795]
[6]
Negroiu, G.; Dwek, R.A.; Petrescu, S.M. Tyrosinase-related protein-2 and -1 are trafficked on distinct routes in B16 melanoma cells. Biochem. Biophys. Res. Commun., 2005, 328(4), 914-921.
[http://dx.doi.org/10.1016/j.bbrc.2005.01.040] [PMID: 15707965]
[7]
Mirzaei, H.; Salehi, H.; Oskuee, R.K.; Mohammadpour, A.; Mirzaei, H.R.; Sharifi, M.R.; Salarinia, R.; Darani, H.Y.; Mokhtari, M.; Masoudifar, A.; Sahebkar, A.; Salehi, R.; Jaafari, M.R. The therapeutic potential of human adipose-derived mesenchymal stem cells producing CXCL10 in a mouse melanoma lung metastasis model. Cancer Lett., 2018, 419, 30-39.
[http://dx.doi.org/10.1016/j.canlet.2018.01.029] [PMID: 29331419]
[8]
Leung, A.M.; Hari, D.M.; Morton, D.L. Surgery for distant melanoma metastasis. Cancer J., 2012, 18(2), 176-184.
[http://dx.doi.org/10.1097/PPO.0b013e31824bc981] [PMID: 22453019]
[9]
Mosca, P.J.; Teicher, E.; Nair, S.P.; Pockaj, B.A. Can surgeons improve survival in stage IV melanoma? J. Surg. Oncol., 2008, 97(5), 462-468.
[http://dx.doi.org/10.1002/jso.20950] [PMID: 18270974]
[10]
Trinh, V.A. Current management of metastatic melanoma. Am. J. Health Syst. Pharm., 2008, 65(24)(Suppl. 9), S3-S8.
[http://dx.doi.org/10.2146/ajhp080460] [PMID: 19052264]
[11]
Pautu, V.; Leonetti, D.; Lepeltier, E.; Clere, N.; Passirani, C. Nanomedicine as a potent strategy in melanoma tumor microenvironment. Pharmacol. Res., 2017, 126, 31-53.
[http://dx.doi.org/10.1016/j.phrs.2017.02.014] [PMID: 28223185]
[12]
Kuske, M.; Westphal, D.; Wehner, R.; Schmitz, M.; Beissert, S.; Praetorius, C.; Meier, F. Immunomodulatory effects of BRAF and MEK inhibitors: Implications for Melanoma therapy. Pharmacol. Res., 2018, 136, 151-159.
[http://dx.doi.org/10.1016/j.phrs.2018.08.019] [PMID: 30145328]
[13]
Khan, N.; Khan, M.K.; Almasan, A.; Singh, A.D.; Macklis, R. The evolving role of radiation therapy in the management of malignant melanoma. Int. J. Radiat. Oncol. Biol. Phys., 2011, 80(3), 645-654.
[http://dx.doi.org/10.1016/j.ijrobp.2010.12.071]
[14]
Siegel, R.; Ward, E.; Brawley, O.; Jemal, A. Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J. Clin., 2011, 61(4), 212-236.
[http://dx.doi.org/10.3322/caac.20121] [PMID: 21685461]
[15]
Nicholl, M.B.; Elashoff, D.; Takeuchi, H.; Morton, D.L.; Hoon, D.S. Molecular upstaging based on paraffin-embedded sentinel lymph nodes: ten-year follow-up confirms prognostic utility in melanoma patients. Ann. Surg., 2011, 253(1), 116-122.
[http://dx.doi.org/10.1097/SLA.0b013e3181fca894] [PMID: 21135695]
[16]
Balkwill, F.; Mantovani, A. Inflammation and cancer: back to Virchow? Lancet, 2001, 357(9255), 539-545.
[http://dx.doi.org/10.1016/S0140-6736(00)04046-0] [PMID: 11229684]
[17]
Vidal-Vanaclocha, F. The prometastatic microenvironment of the liver. Cancer Microenviron., 2008, 1(1), 113-129.
[http://dx.doi.org/10.1007/s12307-008-0011-6] [PMID: 19308690]
[18]
Mirzaei, H.R.; Sahebkar, A.; Salehi, R.; Nahand, J.S.; Karimi, E.; Jaafari, M.R.; Mirzaei, H. Boron neutron capture therapy: moving toward targeted cancer therapy. J. Cancer Res. Ther., 2016, 12(2), 520-525.
[http://dx.doi.org/10.4103/0973-1482.176167] [PMID: 27461603]
[19]
Cianciosi, D.; Varela-Lopez, A.; Forbes-Hernandez, T.Y.; Gasparrini, M.; Afrin, S.; Reboredo-Rodriguez, P.; Zhang, J.; Quiles, J.L.; Nabavi, S.F.; Battino, M.; Giampieri, F. Targeting molecular pathways in cancer stem cells by natural bioactive compounds. Pharmacol. Res., 2018, 135, 150-165.
[http://dx.doi.org/10.1016/j.phrs.2018.08.006] [PMID: 30103002]
[20]
Banik, K.; Ranaware, A.M.; Deshpande, V.; Nalawade, S.P.; Padmavathi, G.; Bordoloi, D.; Sailo, B.L.; Shanmugam, M.K.; Fan, L.; Arfuso, F.; Sethi, G.; Kunnumakkara, A.B. Honokiol for cancer therapeutics: a traditional medicine that can modulate multiple oncogenic targets. Pharmacol. Res., 2019, 144, 192-209.
[http://dx.doi.org/10.1016/j.phrs.2019.04.004] [PMID: 31002949]
[21]
Asensi, M.; Ortega, A.; Mena, S.; Feddi, F.; Estrela, J.M. Natural polyphenols in cancer therapy. Crit. Rev. Clin. Lab. Sci., 2011, 48(5-6), 197-216.
[http://dx.doi.org/10.3109/10408363.2011.631268] [PMID: 22141580]
[22]
Pal, H.C.; Hunt, K.M.; Diamond, A.; Elmets, C.A.; Afaq, F.; Afaq, F. Phytochemicals for the management of melanoma. Mini Rev. Med. Chem., 2016, 16(12), 953-979.
[http://dx.doi.org/10.2174/1389557516666160211120157] [PMID: 26864554]
[23]
Mirzaei, H.; Naseri, G.; Rezaee, R.; Mohammadi, M.; Banikazemi, Z.; Mirzaei, H.R.; Salehi, H.; Peyvandi, M.; Pawelek, J.M.; Sahebkar, A. Curcumin: a new candidate for melanoma therapy? Int. J. Cancer, 2016, 139(8), 1683-1695.
[http://dx.doi.org/10.1002/ijc.30224] [PMID: 27280688]
[24]
Mirzaei, H.; Khoi, M.J.; Azizi, M.; Goodarzi, M. Can curcumin and its analogs be a new treatment option in cancer therapy? Cancer Gene Ther., 2016, 23(11), 410.
[http://dx.doi.org/10.1038/cgt.2016.47] [PMID: 27853147]
[25]
Hesari, A.; Azizian, M.; Sheikhi, A.; Nesaei, A.; Sanaei, S.; Mahinparvar, N.; Derakhshani, M.; Hedayt, P.; Ghasemi, F.; Mirzaei, H. Chemopreventive and therapeutic potential of curcumin in esophageal cancer: Current and future status. Int. J. Cancer, 2019, 144(6), 1215-1226.
[http://dx.doi.org/10.1002/ijc.31947]] [PMID: 30362511]
[26]
Mehta, R.G.; Pezzuto, J.M. Discovery of cancer preventive agents from natural products: from plants to prevention. Curr. Oncol. Rep., 2002, 4(6), 478-486.
[http://dx.doi.org/10.1007/s11912-002-0059-2] [PMID: 12354359 ]
[27]
Rivière, C.; Pawlus, A.D.; Mérillon, J-M. Natural stilbenoids: distribution in the plant kingdom and chemotaxonomic interest in Vitaceae. Nat. Prod. Rep., 2012, 29(11), 1317-1333.
[http://dx.doi.org/10.1039/c2np20049j] [PMID: 23014926]
[28]
Thiel, G.; Ulrich, M.; Mukaida, N.; Rössler, O.G. Resveratrol stimulation induces interleukin-8 gene transcription via NF-κB. Pharmacol. Res., 2018, 134, 238-245.
[http://dx.doi.org/10.1016/j.phrs.2018.07.003] [PMID: 30018026]
[29]
Bradamante, S.; Barenghi, L.; Villa, A. Cardiovascular protective effects of resveratrol. Cardiovasc. Drug Rev., 2004, 22(3), 169-188.
[http://dx.doi.org/10.1111/j.1527-3466.2004.tb00139.x] [PMID: 15492766]
[30]
Jang, M.; Cai, L.; Udeani, G.O.; Slowing, K.V.; Thomas, C.F.; Beecher, C.W.; Fong, H.H.; Farnsworth, N.R.; Kinghorn, A.D.; Mehta, R.G.; Moon, R.C.; Pezzuto, J.M. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science, 1997, 275(5297), 218-220.
[http://dx.doi.org/10.1126/science.275.5297.218] [PMID: 8985016]
[31]
Sinha, K.; Chaudhary, G.; Gupta, Y.K. Protective effect of resveratrol against oxidative stress in middle cerebral artery occlusion model of stroke in rats. Life Sci., 2002, 71(6), 655-665.
[http://dx.doi.org/10.1016/S0024-3205(02)01691-0] [PMID: 12072154]
[32]
Falchetti, R.; Fuggetta, M.P.; Lanzilli, G.; Tricarico, M.; Ravagnan, G. Effects of resveratrol on human immune cell function. Life Sci., 2001, 70(1), 81-96.
[http://dx.doi.org/10.1016/S0024-3205(01)01367-4] [PMID: 11764009]
[33]
Gao, X.; Xu, Y.X.; Janakiraman, N.; Chapman, R.A.; Gautam, S.C. Immunomodulatory activity of resveratrol: suppression of lymphocyte proliferation, development of cell-mediated cytotoxicity, and cytokine production. Biochem. Pharmacol., 2001, 62(9), 1299-1308.
[http://dx.doi.org/10.1016/S0006-2952(01)00775-4] [PMID: 11705464]
[34]
Chen, J-C.; Chen, Y.; Lin, J-H.; Wu, J-M.; Tseng, S-H. Resveratrol suppresses angiogenesis in gliomas: evaluation by color Doppler ultrasound. Anticancer Res., 2006, 26(2A), 1237-1245.
[PMID: 16619530]
[35]
Bishayee, A. Cancer prevention and treatment with resveratrol: from rodent studies to clinical trials. Cancer Prev. Res. (Phila.), 2009, 2(5), 409-418.
[http://dx.doi.org/10.1158/1940-6207.CAPR-08-0160] [PMID: 19401532]
[36]
Delmas, D.; Rébé, C.; Micheau, O.; Athias, A.; Gambert, P.; Grazide, S.; Laurent, G.; Latruffe, N.; Solary, E. Redistribution of CD95, DR4 and DR5 in rafts accounts for the synergistic toxicity of resveratrol and death receptor ligands in colon carcinoma cells. Oncogene, 2004, 23(55), 8979-8986.
[http://dx.doi.org/10.1038/sj.onc.1208086] [PMID: 15480430]
[37]
Fulda, S.; Debatin, K.M. Sensitization for anticancer drug-induced apoptosis by the chemopreventive agent resveratrol. Oncogene, 2004, 23(40), 6702-6711.
[http://dx.doi.org/10.1038/sj.onc.1207630] [PMID: 15273734]
[38]
Baur, J.A.; Sinclair, D.A. Therapeutic potential of resveratrol: the in vivo evidence. Nat. Rev. Drug Discov., 2006, 5(6), 493-506.
[http://dx.doi.org/10.1038/nrd2060] [PMID: 16732220]
[39]
Yang, S.; Irani, K.; Heffron, S.E.; Jurnak, F.; Meyskens, F.L. Jr. Alterations in the expression of the apurinic/apyrimidinic endonuclease-1/redox factor-1 (APE/Ref-1) in human melanoma and identification of the therapeutic potential of resveratrol as an APE/Ref-1 inhibitor. Mol. Cancer Ther., 2005, 4(12), 1923-1935.
[http://dx.doi.org/10.1158/1535-7163.MCT-05-0229] [PMID: 16373707]
[40]
Morris, J.C. Resveratrol, thyroid cancer, and iodide: drink up? Thyroid, 2010, 20(2), 125-126.
[http://dx.doi.org/10.1089/thy.2010.1617] [PMID: 20151819]
[41]
Casanova, F.; Quarti, J.; da Costa, D.C.; Ramos, C.A.; da Silva, J.L.; Fialho, E. Resveratrol chemosensitizes breast cancer cells to melphalan by cell cycle arrest. J. Cell. Biochem., 2012, 113(8), 2586-2596.
[http://dx.doi.org/10.1002/jcb.24134] [PMID: 22415970]
[42]
Tyagi, A.; Gu, M.; Takahata, T.; Frederick, B.; Agarwal, C.; Siriwardana, S.; Agarwal, R.; Sclafani, R.A. Resveratrol selectively induces DNA Damage, independent of Smad4 expression, in its efficacy against human head and neck squamous cell carcinoma. Clin. Cancer Res., 2011, 17(16), 5402-5411.
[http://dx.doi.org/10.1158/1078-0432.CCR-11-1072] [PMID: 21705453]
[43]
Miki, H.; Uehara, N.; Kimura, A.; Sasaki, T.; Yuri, T.; Yoshizawa, K.; Tsubura, A. Resveratrol induces apoptosis via ROS-triggered autophagy in human colon cancer cells. Int. J. Oncol., 2012, 40(4), 1020-1028.
[http://dx.doi.org/10.3892/ijo.2012.1325] [PMID: 22218562]
[44]
Surh, Y.J.; Hurh, Y.J.; Kang, J.Y.; Lee, E.; Kong, G.; Lee, S.J. Resveratrol, an antioxidant present in red wine, induces apoptosis in human promyelocytic leukemia (HL-60) cells. Cancer Lett., 1999, 140(1-2), 1-10.
[http://dx.doi.org/10.1016/S0304-3835(99)00039-7] [PMID: 10403535]
[45]
Shishodia, S.; Aggarwal, B.B. Resveratrol: a polyphenol for all seasons. Oxid. Stress and Dis., 2006, 20, 1.
[46]
Bhardwaj, A.; Sethi, G.; Vadhan-Raj, S.; Bueso-Ramos, C.; Takada, Y.; Gaur, U.; Nair, A.S.; Shishodia, S.; Aggarwal, B.B. Resveratrol inhibits proliferation, induces apoptosis, and overcomes chemoresistance through down-regulation of STAT3 and nuclear factor-kappaB-regulated antiapoptotic and cell survival gene products in human multiple myeloma cells. Blood, 2007, 109(6), 2293-2302.
[http://dx.doi.org/10.1182/blood-2006-02-003988] [PMID: 17164350]
[47]
Wu, Z.; Liu, B.; Cailing, E.; Liu, J.; Zhang, Q.; Liu, J.; Chen, N.; Chen, R.; Zhu, R. Resveratrol inhibits the proliferation of human melanoma cells by inducing G1/S cell cycle arrest and apoptosis. Mol. Med. Rep., 2015, 11(1), 400-404.
[http://dx.doi.org/10.3892/mmr.2014.2716] [PMID: 25333673]
[48]
Kim, M.Y. Nitric oxide triggers apoptosis in A375 human melanoma cells treated with capsaicin and resveratrol. Mol. Med. Rep., 2012, 5(2), 585-591.
[http://dx.doi.org/10.3892/mmr.2011.688]] [PMID: 22143933 ]
[49]
Ahsan, H.; Hadi, S.M. Strand scission in DNA induced by curcumin in the presence of Cu(II). Cancer Lett., 1998, 124(1), 23-30.
[http://dx.doi.org/10.1016/S0304-3835(97)00442-4] [PMID: 9500187]
[50]
Singh, S.; Asad, S.F.; Ahmad, A.; Khan, N.U.; Hadi, S.M. Oxidative DNA damage by capsaicin and dihydrocapsaicin in the presence of Cu(II). Cancer Lett., 2001, 169(2), 139-146.
[http://dx.doi.org/10.1016/S0304-3835(01)00544-4] [PMID: 11431102]
[51]
Rahman, A. Shahabuddin; Hadi, S.M.; Parish, J.H. Complexes involving quercetin, DNA and Cu(II). Carcinogenesis, 1990, 11(11), 2001-2003.
[http://dx.doi.org/10.1093/carcin/11.11.2001] [PMID: 2225332]
[52]
Darby, J.R.T.; Mohd Dollah, M.H.B.; Regnault, T.R.H.; Williams, M.T.; Morrison, J.L. Systematic review: impact of resveratrol exposure during pregnancy on maternal and fetal outcomes in animal models of human pregnancy complications-Are we ready for the clinic? Pharmacol. Res., 2019, 144, 264-278.
[http://dx.doi.org/10.1016/j.phrs.2019.04.020] [PMID: 31029765]
[53]
Haghighatdoost, F.; Hariri, M. Effect of resveratrol on lipid profile: An updated systematic review and meta-analysis on randomized clinical trials. Pharmacol. Res., 2018, 129, 141-150.
[http://dx.doi.org/10.1016/j.phrs.2017.12.033] [PMID: 29305228]
[54]
Juan, M.E.; Alfaras, I.; Planas, J.M. Colorectal cancer chemoprevention by trans-resveratrol. Pharmacol. Res., 2012, 65(6), 584-591.
[http://dx.doi.org/10.1016/j.phrs.2012.03.010] [PMID: 22465196]
[55]
Ignatowicz, E.; Baer-Dubowska, W. Resveratrol, a natural chemopreventive agent against degenerative diseases. Pol. J. Pharmacol., 2001, 53(6), 557-569.
[PMID: 11985329 ]
[56]
Soleas, G.J.; Angelini, M.; Grass, L.; Diamandis, E.P.; Goldberg, D.M. Methods in enzymology; Elsevier, 2001, Vol. 335, pp. 145-154.
[http://dx.doi.org/10.1016/s0076-6879(01)35239-4 ] [PMID: 11400363]
[57]
Soleas, G.J.; Diamandis, E.P.; Goldberg, D.M. Wine as a biological fluid: history, production, and role in disease prevention. J. Clin. Lab. Anal., 1997, 11(5), 287-313.
[http://dx.doi.org/10.1002/(SICI)1098-2825(1997)11:-5<287:AID-JCLA6>3.0.CO;2-4] [PMID: 9292395]
[58]
Renaud, S.; de Lorgeril, M. Wine, alcohol, platelets and the French paradox for coronary heart disease. Lancet, 1992, 339(8808), 1523-1526.
[http://dx.doi.org/10.1016/0140-6736(92)91277-F] [PMID: 1351198]
[59]
Providencia, R. Cardiovascular protection from alcoholic drinks: scientific basis of the French Paradox. Rev. Port. Cardiol., 2006, 25(11), 1043-1058.
[PMID: 17274460]
[60]
Maulik, N. Reactive oxygen species drives myocardial angiogenesis? Antioxidants & redox signaling, , 2006, 8(11-12), 2161-2168.
[http://dx.doi.org/ 10.1089/ars.2006.8.2161]
[61]
Truong, V.L.; Jun, M.; Jeong, W.S. Role of resveratrol in regulation of cellular defense systems against oxidative stress. Biofactors, 2018, 44(1), 36-49.
[http://dx.doi.org/10.1002/biof.1399] [PMID: 29193412]
[62]
Delmas, D.; Jannin, B.; Latruffe, N. Resveratrol: preventing properties against vascular alterations and ageing. Mol. Nutr. Food Res., 2005, 49(5), 377-395.
[http://dx.doi.org/10.1002/mnfr.200400098] [PMID: 15830334]
[63]
Holme, A.L.; Pervaiz, S. Resveratrol in cell fate decisions. J. Bioenerg. Biomembr., 2007, 39(1), 59-63.
[http://dx.doi.org/10.1007/s10863-006-9053-y] [PMID: 17308975]
[64]
Martín, A.R.; Villegas, I.; Sánchez-Hidalgo, M.; de la Lastra, C.A. The effects of resveratrol, a phytoalexin derived from red wines, on chronic inflammation induced in an experimentally induced colitis model. Br. J. Pharmacol., 2006, 147(8), 873-885.
[http://dx.doi.org/10.1038/sj.bjp.0706469] [PMID: 16474422]
[65]
Martín, A.R.; Villegas, I.; La Casa, C.; de la Lastra, C.A. Resveratrol, a polyphenol found in grapes, suppresses oxidative damage and stimulates apoptosis during early colonic inflammation in rats. Biochem. Pharmacol., 2004, 67(7), 1399-1410.
[http://dx.doi.org/10.1016/j.bcp.2003.12.024] [PMID: 15013856]
[66]
de la Lastra, C.A.; Villegas, I. Resveratrol as an anti-inflammatory and anti-aging agent: mechanisms and clinical implications. Mol. Nutr. Food Res., 2005, 49(5), 405-430.
[http://dx.doi.org/10.1002/mnfr.200500022] [PMID: 15832402]
[67]
Aziz, M.H.; Kumar, R.; Ahmad, N. Cancer chemoprevention by resveratrol: in vitro and in vivo studies and the underlying mechanisms. (review) Int. J. Oncol., 2003, 23(1), 17-28.
[http://dx.doi.org/10.3892/ijo.23.1.17] [PMID: 12792772]
[68]
Somoza, V. MAGIC-OL Resveratrol. Mol. Nutr. Food Res., 2005, 49(5), 373-373.
[http://dx.doi.org/10.1002/mnfr.200590020] [PMID: 15880559]
[69]
Cal, C.; Garban, H.; Jazirehi, A.; Yeh, C.; Mizutani, Y.; Bonavida, B. Resveratrol and cancer: chemoprevention, apoptosis, and chemo-immunosensitizing activities. Curr. Med. Chem. Anticancer Agents, 2003, 3(2), 77-93.
[http://dx.doi.org/10.2174/1568011033353443] [PMID: 12678904]
[70]
Ahmad, K.A.; Clement, M.V.; Pervaiz, S. Pro-oxidant activity of low doses of resveratrol inhibits hydrogen peroxide-induced apoptosis. Ann. N. Y. Acad. Sci., 2003, 1010, 365-373.
[http://dx.doi.org/10.1196/annals.1299.067] [PMID: 15033754]
[71]
Martinez, J.; Moreno, J.J. Effect of resveratrol, a natural polyphenolic compound, on reactive oxygen species and prostaglandin production. Biochem. Pharmacol., 2000, 59(7), 865-870.
[http://dx.doi.org/10.1016/S0006-2952(99)00380-9] [PMID: 10718345]
[72]
Gusman, J.; Malonne, H.; Atassi, G. A reappraisal of the potential chemopreventive and chemotherapeutic properties of resveratrol. Carcinogenesis, 2001, 22(8), 1111-1117.
[http://dx.doi.org/10.1093/carcin/22.8.1111] [PMID: 11470738]
[73]
Ozgová, S.; Hermánek, J.; Gut, I. Different antioxidant effects of polyphenols on lipid peroxidation and hydroxyl radicals in the NADPH-, Fe-ascorbate- and Fe-microsomal systems. Biochem. Pharmacol., 2003, 66(7), 1127-1137.
[http://dx.doi.org/10.1016/S0006-2952(03)00425-8] [PMID: 14505792]
[74]
Gatouillat, G.; Balasse, E.; Joseph-Pietras, D.; Morjani, H.; Madoulet, C. Resveratrol induces cell-cycle disruption and apoptosis in chemoresistant B16 melanoma. J. Cell. Biochem., 2010, 110(4), 893-902.
[http://dx.doi.org/10.1002/jcb.22601] [PMID: 20564188]
[75]
Bhattacharya, S.; Darjatmoko, S.R.; Polans, A.S. Resveratrol modulates the malignant properties of cutaneous melanoma through changes in the activation and attenuation of the antiapoptotic protooncogenic protein Akt/PKB. Melanoma Res., 2011, 21(3), 180-187.
[http://dx.doi.org/10.1097/CMR.0b013e3283456dfc] [PMID: 21407133]
[76]
van Ginkel, P.R.; Darjatmoko, S.R.; Sareen, D.; Subramanian, L.; Bhattacharya, S.; Lindstrom, M.J.; Albert, D.M.; Polans, A.S. Resveratrol inhibits uveal melanoma tumor growth via early mitochondrial dysfunction. Invest. Ophthalmol. Vis. Sci., 2008, 49(4), 1299-1306.
[http://dx.doi.org/10.1167/iovs.07-1233] [PMID: 18385041]
[77]
Szekeres, T.; Saiko, P.; Fritzer-Szekeres, M.; Djavan, B.; Jäger, W. Chemopreventive effects of resveratrol and resveratrol derivatives. Ann. N. Y. Acad. Sci., 2011, 1215, 89-95.
[http://dx.doi.org/10.1111/j.1749-6632.2010.05864.x] [PMID: 21261645]
[78]
Bhatia, S.; Tykodi, S.S.; Thompson, J.A. Treatment of metastatic melanoma: an overview. Oncology (Williston Park), 2009, 23(6), 488-496.
[PMID: 19544689]
[79]
Sporn, M.B.; Dunlop, N.M.; Newton, D.L.; Smith, J.M. Prevention of chemical carcinogenesis by vitamin A and its synthetic analogs (retinoids). Fed. Proc., 1976, 35(6), 1332-1338.
[PMID: 770206]
[80]
Yuan, Y.; Xue, X.; Guo, R.B.; Sun, X.L.; Hu, G. Resveratrol enhances the antitumor effects of temozolomide in glioblastoma via ROS-dependent AMPK-TSC-mTOR signaling pathway. CNS Neurosci. Ther., 2012, 18(7), 536-546.
[http://dx.doi.org/10.1111/j.1755-5949.2012.00319.x] [PMID: 22530672]
[81]
Iwuchukwu, O.F.; Tallarida, R.J.; Nagar, S. Resveratrol in combination with other dietary polyphenols concomitantly enhances antiproliferation and UGT1A1 induction in Caco-2 cells. Life Sci., 2011, 88(23-24), 1047-1054.
[http://dx.doi.org/10.1016/j.lfs.2011.03.016] [PMID: 21466813]
[82]
Harper, C.E.; Cook, L.M.; Patel, B.B.; Wang, J.; Eltoum, I.A.; Arabshahi, A.; Shirai, T.; Lamartiniere, C.A. Genistein and resveratrol, alone and in combination, suppress prostate cancer in SV-40 tag rats. Prostate, 2009, 69(15), 1668-1682.
[http://dx.doi.org/10.1002/pros.21017] [PMID: 19670229]
[83]
Singh, C.K.; George, J.; Ahmad, N. Resveratrol-based combinatorial strategies for cancer management. Ann. N. Y. Acad. Sci., 2013, 1290, 113-121.
[http://dx.doi.org/10.1111/nyas.12160] [PMID: 23855473]
[84]
Olson, S.Y.; Garbán, H.J. Regulation of apoptosis-related genes by nitric oxide in cancer. Nitric Oxide, 2008, 19(2), 170-176.
[http://dx.doi.org/10.1016/j.niox.2008.04.005] [PMID: 18460349]
[85]
Williams, E.L.; Djamgoz, M.B. Nitric oxide and metastatic cell behaviour. BioEssays, 2005, 27(12), 1228-1238.
[http://dx.doi.org/10.1002/bies.20324] [PMID: 16299735]
[86]
Vogelstein, B.; Lane, D.; Levine, A.J. Surfing the p53 network. Nature, 2000, 408(6810), 307-310.
[http://dx.doi.org/10.1038/35042675] [PMID: 11099028]
[87]
Athar, M.; Back, J.H.; Kopelovich, L.; Bickers, D.R.; Kim, A.L. Multiple molecular targets of resveratrol: Anti-carcinogenic mechanisms. Arch. Biochem. Biophys., 2009, 486(2), 95-102.
[http://dx.doi.org/10.1016/j.abb.2009.01.018] [PMID: 19514131]
[88]
Piotrowska, H.; Kucinska, M.; Murias, M. Biological activity of piceatannol: leaving the shadow of resveratrol. Mutat. Res., 2012, 750(1), 60-82.
[http://dx.doi.org/10.1016/j.mrrev.2011.11.001] [PMID: 22108298]
[89]
Zhao, H.; Han, L.; Jian, Y.; Ma, Y.; Yan, W.; Chen, X.; Xu, H.; Li, L. Resveratrol induces apoptosis in human melanoma cell through negatively regulating Erk/PKM2/Bcl-2 axis. OncoTargets Ther., 2018, 11, 8995-9006.
[http://dx.doi.org/10.2147/OTT.S186247] [PMID: 30588012]
[90]
Heo, J.R.; Kim, S.M.; Hwang, K.A.; Kang, J.H.; Choi, K.C. Resveratrol induced reactive oxygen species and endoplasmic reticulum stress mediated apoptosis, and cell cycle arrest in the A375SM malignant melanoma cell line. Int. J. Mol. Med., 2018, 42(3), 1427-1435.
[http://dx.doi.org/10.3892/ijmm.2018.3732] [PMID: 29916532]
[91]
Rojas, C.; Pan-Castillo, B.; Valls, C.; Pujadas, G.; Garcia-Vallve, S.; Arola, L.; Mulero, M. Resveratrol enhances palmitate-induced ER stress and apoptosis in cancer cells. PLoS One, 2014, 9(12)e113929
[http://dx.doi.org/10.1371/journal.pone.0113929] [PMID: 25436452]
[92]
Marciniak, S.J.; Yun, C.Y.; Oyadomari, S.; Novoa, I.; Zhang, Y.; Jungreis, R.; Nagata, K.; Harding, H.P.; Ron, D. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev., 2004, 18(24), 3066-3077.
[http://dx.doi.org/10.1101/gad.1250704] [PMID: 15601821]
[93]
Rozpedek, W.; Pytel, D.; Mucha, B.; Leszczynska, H.; Diehl, J.A.; Majsterek, I. The role of the PERK/eIF2α/ATF4/CHOP signaling pathway in tumor progression during endoplasmic reticulum stress. Curr. Mol. Med., 2016, 16(6), 533-544.
[http://dx.doi.org/10.2174/1566524016666160523143937] [PMID: 27211800]
[94]
Yen, Y-P.; Tsai, K-S.; Chen, Y-W.; Huang, C-F.; Yang, R-S.; Liu, S-H. Arsenic induces apoptosis in myoblasts through a reactive oxygen species-induced endoplasmic reticulum stress and mitochondrial dysfunction pathway. Arch. Toxicol., 2012, 86(6), 923-933.
[http://dx.doi.org/10.1007/s00204-012-0864-9] [PMID: 22622864]
[95]
Kim, E.K.; Choi, E-J. Pathological roles of MAPK signaling pathways in human diseases. Biochimica et Biophysica Acta (BBA)-. Molecular Basis of Disease, 2010, 1802(4), 396-405.
[http://dx.doi.org/10.1016/j.bbadis.2009.12.009]
[96]
Birben, E.; Sahiner, U.M.; Sackesen, C.; Erzurum, S.; Kalayci, O. Oxidative stress and antioxidant defense. World Allergy Organ. J., 2012, 5(1), 9-19.
[http://dx.doi.org/10.1097/WOX.0b013e3182439613]] [PMID: 23268465]
[97]
Wang, X-J.; Sun, Z.; Villeneuve, N.F.; Zhang, S.; Zhao, F.; Li, Y.; Chen, W.; Yi, X.; Zheng, W.; Wondrak, G.T.; Wong, P.K.; Zhang, D.D. Nrf2 enhances resistance of cancer cells to chemotherapeutic drugs, the dark side of Nrf2. Carcinogenesis, 2008, 29(6), 1235-1243.
[http://dx.doi.org/10.1093/carcin/bgn095] [PMID: 18413364]
[98]
Menegon, S.; Columbano, A.; Giordano, S. The dual roles of NRF2 in cancer. Trends Mol. Med., 2016, 22(7), 578-593.
[http://dx.doi.org/10.1016/j.molmed.2016.05.002] [PMID: 27263465]
[99]
Rachakonda, G.; Sekhar, K.R.; Jowhar, D.; Samson, P.C.; Wikswo, J.P.; Beauchamp, R.D.; Datta, P.K.; Freeman, M.L. Increased cell migration and plasticity in Nrf2-deficient cancer cell lines. Oncogene, 2010, 29(25), 3703-3714.
[http://dx.doi.org/10.1038/onc.2010.118] [PMID: 20440267]
[100]
Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol., 2013, 53, 401-426.
[http://dx.doi.org/10.1146/annurev-pharmtox-011112-140320] [PMID: 23294312]
[101]
Moon, E.J.; Giaccia, A. Dual roles of NRF2 in tumor prevention and progression: possible implications in cancer treatment. Free Radic. Biol. Med., 2015, 79, 292-299.
[http://dx.doi.org/10.1016/j.freeradbiomed.2014.11.009] [PMID: 25458917]
[102]
Karin, M. Nuclear factor-kappaB in cancer development and progression. Nature, 2006, 441(7092), 431-436.
[http://dx.doi.org/10.1038/nature04870] [PMID: 16724054]
[103]
Ivanov, V.N.; Bhoumik, A.; Ronai, Z. Death receptors and melanoma resistance to apoptosis. Oncogene, 2003, 22(20), 3152-3161.
[http://dx.doi.org/10.1038/sj.onc.1206456] [PMID: 12789291]
[104]
Aggarwal, B.B.; Bhardwaj, A.; Aggarwal, R.S.; Seeram, N.P.; Shishodia, S.; Takada, Y. Role of resveratrol in prevention and therapy of cancer: preclinical and clinical studies. Anticancer Res., 2004, 24(5A), 2783-2840.
[PMID: 15517885]
[105]
Shankar, S.; Singh, G.; Srivastava, R.K. Chemoprevention by resveratrol: molecular mechanisms and therapeutic potential. Front. Biosci., 2007, 12(12), 4839-4854.
[http://dx.doi.org/10.2741/2432] [PMID: 17569614]
[106]
Dörrie, J.; Gerauer, H.; Wachter, Y.; Zunino, S.J. Resveratrol induces extensive apoptosis by depolarizing mitochondrial membranes and activating caspase-9 in acute lymphoblastic leukemia cells. Cancer Res., 2001, 61(12), 4731-4739.
[PMID: 11406544]
[107]
Niles, R.M.; McFarland, M.; Weimer, M.B.; Redkar, A.; Fu, Y-M.; Meadows, G.G. Resveratrol is a potent inducer of apoptosis in human melanoma cells. Cancer Lett., 2003, 190(2), 157-163.
[http://dx.doi.org/10.1016/S0304-3835(02)00676-6] [PMID: 12565170]
[108]
Weidner, N.; Semple, J.P.; Welch, W.R.; Folkman, J. Tumor angiogenesis and metastasis--correlation in invasive breast carcinoma. N. Engl. J. Med., 1991, 324(1), 1-8.
[http://dx.doi.org/10.1056/NEJM199101033240101] [PMID: 1701519]
[109]
Weidner, N. Angiogenesis as a predictor of clinical outcome in cancer patients. Hum. Pathol., 2000, 31(4), 403-405.
[http://dx.doi.org/10.1053/hp.2000.6724] [PMID: 10821484]
[110]
Neufeld, G.; Cohen, T.; Gengrinovitch, S.; Poltorak, Z. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J., 1999, 13(1), 9-22.
[http://dx.doi.org/10.1096/fasebj.13.1.9] [PMID: 9872925]
[111]
Volm, M.; Koomägi, R.; Mattern, J. Prognostic value of vascular endothelial growth factor and its receptor Flt-1 in squamous cell lung cancer. Int. J. Cancer, 1997, 74(1), 64-68.
[http://dx.doi.org/10.1002/(SICI)10970215(19970220)-74:1<64:AID-IJC11>3.0.CO;2-I] [PMID: 9036871]
[112]
Pelletier, F.; Bermont, L.; Puzenat, E.; Blanc, D.; Cairey-Remonnay, S.; Mougin, C.; Laurent, R.; Humbert, P.; Aubin, F. Circulating vascular endothelial growth factor in cutaneous malignant melanoma. Br. J. Dermatol., 2005, 152(4), 685-689.
[http://dx.doi.org/10.1111/j.1365-2133.2005.06507.x] [PMID: 15840099]
[113]
Sánchez-Puig, N.; Veprintsev, D.B.; Fersht, A.R. Binding of natively unfolded HIF-1alpha ODD domain to p53. Mol. Cell, 2005, 17(1), 11-21.
[http://dx.doi.org/10.1016/j.molcel.2004.11.019] [PMID: 15629713]
[114]
Hansson, L.O.; Friedler, A.; Freund, S.; Rudiger, S.; Fersht, A.R. Two sequence motifs from HIF-1alpha bind to the DNA-binding site of p53. Proc. Natl. Acad. Sci. USA, 2002, 99(16), 10305-10309.
[http://dx.doi.org/10.1073/pnas.122347199] [PMID: 12124396]
[115]
Graeber, T.G.; Peterson, J.F.; Tsai, M.; Monica, K.; Fornace, A.J., Jr; Giaccia, A.J. Hypoxia induces accumulation of p53 protein, but activation of a G1-phase checkpoint by low-oxygen conditions is independent of p53 status. Mol. Cell. Biol., 1994, 14(9), 6264-6277.
[http://dx.doi.org/10.1128/MCB.14.9.6264] [PMID: 8065358]
[116]
Tokunaga, T.; Nakamura, M.; Oshika, Y.; Tsuchida, T.; Kazuno, M.; Fukushima, Y.; Kawai, K.; Abe, Y.; Kijima, H.; Yamazaki, H.; Tamaoki, N.; Ueyama, Y. Alterations in tumour suppressor gene p53 correlate with inhibition of thrombospondin-1 gene expression in colon cancer cells. Virchows Arch., 1998, 433(5), 415-418.
[http://dx.doi.org/10.1007/s004280050268]
[117]
Iruela-Arispe, M.L.; Lombardo, M.; Krutzsch, H.C.; Lawler, J.; Roberts, D.D. Inhibition of angiogenesis by thrombospondin-1 is mediated by 2 independent regions within the type 1 repeats. Circulation, 1999, 100(13), 1423-1431.
[http://dx.doi.org/10.1161/01.CIR.100.13.1423] [PMID: 10500044]
[118]
Sid, B.; Sartelet, H.; Bellon, G.; El Btaouri, H.; Rath, G.; Delorme, N.; Haye, B.; Martiny, L. Thrombospondin 1: a multifunctional protein implicated in the regulation of tumor growth. Crit. Rev. Oncol. Hematol., 2004, 49(3), 245-258.
[http://dx.doi.org/10.1016/j.critrevonc.2003.09.009] [PMID: 15036264]
[119]
Grant, S.W.; Kyshtoobayeva, A.S.; Kurosaki, T.; Jakowatz, J.; Fruehauf, J.P. Mutant p53 correlates with reduced expression of thrombospondin-1, increased angiogenesis, and metastatic progression in melanoma. Cancer Detect. Prev., 1998, 22(3), 185-194.
[http://dx.doi.org/10.1046/j.1525-1500.1998.0oa18.x] [PMID: 9618039]
[120]
Cao, Z.; Fang, J.; Xia, C.; Shi, X.; Jiang, B.H. Trans-3,4,5′-Trihydroxystibene inhibits hypoxia-inducible factor 1alpha and vascular endothelial growth factor expression in human ovarian cancer cells. Clin. Cancer Res., 2004, 10(15), 5253-5263.
[http://dx.doi.org/10.1158/1078-0432.CCR-03-0588] [PMID: 15297429]
[121]
Wu, H.; Liang, X.; Fang, Y.; Qin, X.; Zhang, Y.; Liu, J. Resveratrol inhibits hypoxia-induced metastasis potential enhancement by restricting hypoxia-induced factor-1 alpha expression in colon carcinoma cells. Biomed. Pharmacother., 2008, 62(9), 613-621.
[http://dx.doi.org/10.1016/j.biopha.2008.06.036] [PMID: 18674879]
[122]
Trapp, V.; Parmakhtiar, B.; Papazian, V.; Willmott, L.; Fruehauf, J.P. Anti-angiogenic effects of resveratrol mediated by decreased VEGF and increased TSP1 expression in melanoma-endothelial cell co-culture. Angiogenesis, 2010, 13(4), 305-315.
[http://dx.doi.org/10.1007/s10456-010-9187-8] [PMID: 20927579]
[123]
Ivanov, V.N.; Partridge, M.A.; Johnson, G.E.; Huang, S.X.; Zhou, H.; Hei, T.K. Resveratrol sensitizes melanomas to TRAIL through modulation of antiapoptotic gene expression. Exp. Cell Res., 2008, 314(5), 1163-1176.
[http://dx.doi.org/10.1016/j.yexcr.2007.12.012] [PMID: 18222423]
[124]
Fulda, S.; Debatin, K-M. Sensitization for tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis by the chemopreventive agent resveratrol. Cancer Res., 2004, 64(1), 337-346.
[http://dx.doi.org/10.1158/0008-5472.CAN-03-1656] [PMID: 14729643]
[125]
Wang, M.; Yu, T.; Zhu, C.; Sun, H.; Qiu, Y.; Zhu, X.; Li, J. Resveratrol triggers protective autophagy through the ceramide/Akt/mTOR pathway in melanoma B16 cells. Nutr. Cancer, 2014, 66(3), 435-440.
[http://dx.doi.org/10.1080/01635581.2013.878738] [PMID: 24579778]
[126]
Osmond, G.W.; Augustine, C.K.; Zipfel, P.A.; Padussis, J.; Tyler, D.S. Enhancing melanoma treatment with resveratrol. J. Surg. Res., 2012, 172(1), 109-115.
[http://dx.doi.org/10.1016/j.jss.2010.07.033] [PMID: 20855085]
[127]
Fang, Y.; Bradley, M.J.; Cook, K.M.; Herrick, E.J.; Nicholl, M.B. A potential role for resveratrol as a radiation sensitizer for melanoma treatment. J. Surg. Res., 2013, 183(2), 645-653.
[http://dx.doi.org/10.1016/j.jss.2013.02.037] [PMID: 23522452]
[128]
Lei, M.J.; Dong, Y.; Sun, C.X.; Zhang, X.H. Resveratrol inhibits proliferation, promotes differentiation and melanogenesis in HT-144 melanoma cells through inhibition of MEK/ERK kinase pathway. Microb. Pathog., 2017, 111, 410-413.
[http://dx.doi.org/10.1016/j.micpath.2017.09.029] [PMID: 28919486]
[129]
Lee, T.H.; Seo, J.O.; Baek, S-H.; Kim, S.Y. Inhibitory effects of resveratrol on melanin synthesis in ultraviolet B-induced pigmentation in Guinea pig skin. Biomol. Ther. (Seoul), 2014, 22(1), 35-40.
[http://dx.doi.org/10.4062/biomolther.2013.081] [PMID: 24596619]
[130]
Salado, C.; Olaso, E.; Gallot, N.; Valcarcel, M.; Egilegor, E.; Mendoza, L.; Vidal-Vanaclocha, F. Resveratrol prevents inflammation-dependent hepatic melanoma metastasis by inhibiting the secretion and effects of interleukin-18. J. Transl. Med., 2011, 9(1), 59.
[http://dx.doi.org/10.1186/1479-5876-9-59] [PMID: 21569399]
[131]
Luo, H.; Umebayashi, M.; Doi, K.; Morisaki, T.; Shirasawa, S.; Tsunoda, T. Resveratrol overcomes cellular resistance to vemurafenib through dephosphorylation of akt in BRAF-mutated melanoma cells. Anticancer Res., 2016, 36(7), 3585-3589.
[PMID: 27354627]
[132]
Junco, J.J.; Mancha-Ramirez, A.; Malik, G.; Wei, S-J.; Kim, D.J.; Liang, H.; Slaga, T.J. Ursolic acid and resveratrol synergize with chloroquine to reduce melanoma cell viability. Melanoma Res., 2015, 25(2), 103-112.
[http://dx.doi.org/10.1097/CMR.0000000000000137] [PMID: 25647735]
[133]
Chen, M-C.; Chang, W-W.; Kuan, Y-D.; Lin, S-T.; Hsu, H-C.; Lee, C-H. Resveratrol inhibits LPS-induced epithelial-mesenchymal transition in mouse melanoma model. Innate Immun., 2012, 18(5), 685-693.
[http://dx.doi.org/10.1177/1753425912436589] [PMID: 22344225]
[134]
Cheng, Y.J.; Chang, M.Y.; Chang, W.W.; Wang, W.K.; Liu, C.F.; Lin, S.T.; Lee, C.H. Resveratrol enhances chemosensitivity in mouse melanoma model through connexin 43 upregulation. Environ. Toxicol., 2015, 30(8), 877-886.
[http://dx.doi.org/10.1002/tox.21952] [PMID: 24449132]
[135]
Chen, Y-J.; Chen, Y-Y.; Lin, Y-F.; Hu, H-Y.; Liao, H-F. Resveratrol inhibits alpha-melanocyte-stimulating hormone signaling, viability and invasiveness in melanoma cells. Evid. Based Complement. Alternat. Med., 2013, 2013632121
[http://dx.doi.org/10.1155/2013/632121] [PMID: 23762150]
[136]
Habibie, H.; Yokoyama, S.; Abdelhamed, S.; Awale, S.; Sakurai, H.; Hayakawa, Y.; Saiki, I. Survivin suppression through STAT3/β-catenin is essential for resveratrol-induced melanoma apoptosis. Int. J. Oncol., 2014, 45(2), 895-901.
[http://dx.doi.org/10.3892/ijo.2014.2480] [PMID: 24946930]
[137]
Menicacci, B.; Laurenzana, A.; Chillà, A.; Margheri, F.; Peppicelli, S.; Tanganelli, E.; Fibbi, G.; Giovannelli, L.; Del Rosso, M.; Mocali, A. Chronic resveratrol treatment inhibits MRC5 fibroblast SASP-related protumoral effects on melanoma cells. J. Gerontol. A Biol. Sci. Med. Sci., 2017, 72(9), 1187-1195.
[http://dx.doi.org/10.1093/gerona/glw336] [PMID: 28329136]
[138]
Lee, S.H.; Koo, B.S.; Park, S.Y.; Kim, Y.M. Anti-angiogenic effects of resveratrol in combination with 5-fluorouracil on B16 murine melanoma cells. Mol. Med. Rep., 2015, 12(2), 2777-2783.
[http://dx.doi.org/10.3892/mmr.2015.3675] [PMID: 25936796]
[139]
Sim, D.Y.; Sohng, J.K.; Jung, H.J. Anticancer activity of 7,8-dihydroxyflavone in melanoma cells via downregulation of α-MSH/cAMP/MITF pathway. Oncol. Rep., 2016, 36(1), 528-534.
[http://dx.doi.org/10.3892/or.2016.4825] [PMID: 27220989]
[140]
Lee, H.; Zhang, P.; Herrmann, A.; Yang, C.; Xin, H.; Wang, Z.; Hoon, D.S.; Forman, S.J.; Jove, R.; Riggs, A.D.; Yu, H. Acetylated STAT3 is crucial for methylation of tumor-suppressor gene promoters and inhibition by resveratrol results in demethylation. Proc. Natl. Acad. Sci. USA, 2012, 109(20), 7765-7769.
[http://dx.doi.org/10.1073/pnas.1205132109] [PMID: 22547799]
[141]
Guan, H.; Singh, N.P.; Singh, U.P.; Nagarkatti, P.S.; Nagarkatti, M. Resveratrol prevents endothelial cells injury in high-dose interleukin-2 therapy against melanoma. PLoS One, 2012, 7(4)e35650
[http://dx.doi.org/10.1371/journal.pone.0035650] [PMID: 22532866]
[142]
Wu, F.; Cui, L. Resveratrol suppresses melanoma by inhibiting NF-κB/miR-221 and inducing TFG expression. Arch. Dermatol. Res., 2017, 309(10), 823-831.
[http://dx.doi.org/10.1007/s00403-017-1784-6] [PMID: 28936555]
[143]
Yang, Z.; Yang, S.; Misner, B.J.; Chiu, R.; Liu, F.; Meyskens, F.L. Jr Nitric oxide initiates progression of human melanoma via a feedback loop mediated by apurinic/apyrimidinic endonuclease-1/redox factor-1, which is inhibited by resveratrol. Mol. Cancer Ther., 2008, 7(12), 3751-3760.
[http://dx.doi.org/10.1158/1535-7163.MCT-08-0562] [PMID: 19074850]
[144]
Fuggetta, M.P.; D’Atri, S.; Lanzilli, G.; Tricarico, M.; Cannavò, E.; Zambruno, G.; Falchetti, R.; Ravagnan, G. In vitro antitumour activity of resveratrol in human melanoma cells sensitive or resistant to temozolomide. Melanoma Res., 2004, 14(3), 189-196.
[http://dx.doi.org/10.1097/01.cmr.0000130007.54508.b2] [PMID: 15179187]
[145]
Hsieh, T.C.; Wang, Z.; Hamby, C.V.; Wu, J.M. Inhibition of melanoma cell proliferation by resveratrol is correlated with upregulation of quinone reductase 2 and p53. Biochem. Biophys. Res. Commun., 2005, 334(1), 223-230.
[http://dx.doi.org/10.1016/j.bbrc.2005.06.073] [PMID: 15993843]
[146]
Yang, S.; Meyskens, F.L. Jr Alterations in activating protein 1 composition correlate with phenotypic differentiation changes induced by resveratrol in human melanoma. Mol. Pharmacol., 2005, 67(1), 298-308.
[http://dx.doi.org/10.1124/mol.104.006023] [PMID: 15492115]
[147]
Ogas, T.; Kondratyuk, T.P.; Pezzuto, J.M. Resveratrol analogs: promising chemopreventive agents. Ann. N. Y. Acad. Sci., 2013, 1290, 21-29.
[http://dx.doi.org/10.1111/nyas.12196] [PMID: 23855462]
[148]
Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E. Jr.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos., 2004, 32(12), 1377-1382.
[http://dx.doi.org/10.1124/dmd.104.000885] [PMID: 15333514]
[149]
Neves, A.R.; Lucio, M.; Lima, J.L.; Reis, S. Resveratrol in medicinal chemistry: a critical review of its pharmacokinetics, drug-delivery, and membrane interactions. Curr. Med. Chem., 2012, 19(11), 1663-1681.
[http://dx.doi.org/10.2174/092986712799945085] [PMID: 22257059]
[150]
Szekeres, T.; Fritzer-Szekeres, M.; Saiko, P.; Jäger, W. Resveratrol and resveratrol analogues--structure-activity relationship. Pharm. Res., 2010, 27(6), 1042-1048.
[http://dx.doi.org/10.1007/s11095-010-0090-1] [PMID: 20232118]
[151]
Savio, M.; Coppa, T.; Bianchi, L.; Vannini, V.; Maga, G.; Forti, L.; Cazzalini, O.; Lazzè, M.C.; Perucca, P.; Prosperi, E.; Stivala, L.A. The resveratrol analogue 4,4′-dihydroxy-trans-stilbene inhibits cell proliferation with higher efficiency but different mechanism from resveratrol. Int. J. Biochem. Cell Biol., 2009, 41(12), 2493-2502.
[http://dx.doi.org/10.1016/j.biocel.2009.08.005] [PMID: 19679195]
[152]
Murias, M.; Jäger, W.; Handler, N.; Erker, T.; Horvath, Z.; Szekeres, T.; Nohl, H.; Gille, L. Antioxidant, prooxidant and cytotoxic activity of hydroxylated resveratrol analogues: structure-activity relationship. Biochem. Pharmacol., 2005, 69(6), 903-912.
[http://dx.doi.org/10.1016/j.bcp.2004.12.001] [PMID: 15748702]
[153]
Li, X-Z.; Wei, X.; Zhang, C-J.; Jin, X-L.; Tang, J-J.; Fan, G-J.; Zhou, B. Hypohalous acid-mediated halogenation of resveratrol and its role in antioxidant and antimicrobial activities. Food Chem., 2012, 135(3), 1239-1244.
[http://dx.doi.org/10.1016/j.foodchem.2012.05.043] [PMID: 22953849]
[154]
Nawaz, W.; Zhou, Z.; Deng, S.; Ma, X.; Ma, X.; Li, C.; Shu, X. Therapeutic versatility of resveratrol derivatives. Nutrients, 2017, 9(11), 1188.
[http://dx.doi.org/10.3390/nu9111188] [PMID: 29109374]
[155]
D’Mello, S.A.; Finlay, G.J.; Baguley, B.C.; Askarian-Amiri, M.E. Signaling Pathways in Melanogenesis. Int. J. Mol. Sci., 2016, 17(7)E1144
[http://dx.doi.org/10.3390/ijms17071144] [PMID: 27428965]
[156]
Choi, J.; Bae, S.J.; Ha, Y.M.; No, J.K.; Lee, E.K.; Lee, J.S.; Song, S.; Lee, H.; Suh, H.; Yu, B.P.; Chung, H.Y. A newly synthesized, potent tyrosinase inhibitor: 5-(6-hydroxy-2-naphthyl)-1,2,3-benzenetriol. Bioorg. Med. Chem. Lett., 2010, 20(16), 4882-4884.
[http://dx.doi.org/10.1016/j.bmcl.2010.06.087] [PMID: 20619644]
[157]
Kosuru, R.; Rai, U.; Prakash, S.; Singh, A.; Singh, S. Promising therapeutic potential of pterostilbene and its mechanistic insight based on preclinical evidence. Eur. J. Pharmacol., 2016, 789, 229-243.
[http://dx.doi.org/10.1016/j.ejphar.2016.07.046] [PMID: 27475678]
[158]
Estrela, J.M.; Ortega, A.; Mena, S.; Rodriguez, M.L.; Asensi, M. Pterostilbene: biomedical applications. Crit. Rev. Clin. Lab. Sci., 2013, 50(3), 65-78.
[http://dx.doi.org/10.3109/10408363.2013.805182] [PMID: 23808710]
[159]
Remsberg, C.M.; Yáñez, J.A.; Ohgami, Y.; Vega-Villa, K.R.; Rimando, A.M.; Davies, N.M. Pharmacometrics of pterostilbene: preclinical pharmacokinetics and metabolism, anticancer, antiinflammatory, antioxidant and analgesic activity. Phytother. Res., 2008, 22(2), 169-179.
[http://dx.doi.org/10.1002/ptr.2277] [PMID: 17726731]
[160]
Lin, H.S.; Yue, B.D.; Ho, P.C. Determination of pterostilbene in rat plasma by a simple HPLC-UV method and its application in pre-clinical pharmacokinetic study. Biomed. Chromatogr., 2009, 23(12), 1308-1315.
[http://dx.doi.org/10.1002/bmc.1254] [PMID: 19488981]
[161]
Maccario, C.; Savio, M.; Ferraro, D.; Bianchi, L.; Pizzala, R.; Pretali, L.; Forti, L.; Stivala, L.A. The resveratrol analog 4,4′-dihydroxy-trans-stilbene suppresses transformation in normal mouse fibroblasts and inhibits proliferation and invasion of human breast cancer cells. Carcinogenesis, 2012, 33(11), 2172-2180.
[http://dx.doi.org/10.1093/carcin/bgs244] [PMID: 22828135]
[162]
Saha, B.; Pai, G.B.; Subramanian, M.; Gupta, P.; Tyagi, M.; Patro, B.S.; Chattopadhyay, S. Resveratrol analogue, trans-4,4′-dihydroxystilbene (DHS), inhibits melanoma tumor growth and suppresses its metastatic colonization in lungs. Biomed. Pharmacother., 2018, 107, 1104-1114.
[http://dx.doi.org/10.1016/j.biopha.2018.08.085]
[163]
Du, M.; Zhang, Z.; Gao, T. Piceatannol induced apoptosis through up-regulation of microRNA-181a in melanoma cells. Biol. Res., 2017, 50(1), 36.
[http://dx.doi.org/10.1186/s40659-017-0141-8] [PMID: 29041990]
[164]
Larrosa, M.; Tomás-Barberán, F.A.; Espín, J.C. The grape and wine polyphenol piceatannol is a potent inducer of apoptosis in human SK-Mel-28 melanoma cells. Eur. J. Nutr., 2004, 43(5), 275-284.
[http://dx.doi.org/10.1007/s00394-004-0471-5] [PMID: 15309446]
[165]
Wong, Y.; Osmond, G.; Brewer, K.I.; Tyler, D.S.; Andrus, M.B. Synthesis of 4′-ester analogs of resveratrol and their evaluation in malignant melanoma and pancreatic cell lines. Bioorg. Med. Chem. Lett., 2010, 20(3), 1198-1201.
[http://dx.doi.org/10.1016/j.bmcl.2009.12.006] [PMID: 20022501]
[166]
Saha, B.; Pai, G.B.; Subramanian, M.; Gupta, P.; Tyagi, M.; Patro, B.S.; Chattopadhyay, S. Resveratrol analogue, trans-4,4′-dihydroxystilbene (DHS), inhibits melanoma tumor growth and suppresses its metastatic colonization in lungs. Biomed. Pharmacother., 2018, 107, 1104-1114.
[http://dx.doi.org/10.1016/j.biopha.2018.08.085] [PMID: 30257322]
[167]
Androutsopoulos, V.P.; Fragiadaki, I.; Tosca, A. Activation of ERK1/2 is required for the antimitotic activity of the resveratrol analogue 3,4,5,4′-tetramethoxystilbene (DMU-212) in human melanoma cells. Exp. Dermatol., 2015, 24(8), 632-634.
[http://dx.doi.org/10.1111/exd.12721] [PMID: 25865632]
[168]
Bae, S.J.; Ha, Y.M.; Kim, J-A.; Park, J.Y.; Ha, T.K.; Park, D.; Chun, P.; Park, N.H.; Moon, H.R.; Chung, H.Y. A novel synthesized tyrosinase inhibitor: (E)-2-((2,4-dihydroxyphenyl)diazenyl)phenyl 4-methylbenzene-sulfonate as an azo-resveratrol analog. Biosci. Biotechnol. Biochem., 2013, 77(1), 65-72.
[http://dx.doi.org/10.1271/bbb.120547] [PMID: 23291747]
[169]
Moriyama, H.; Moriyama, M.; Ninomiya, K.; Morikawa, T.; Hayakawa, T. Inhibitory effects of oligostilbenoids from the bark of Shorea roxburghii on malignant melanoma cell growth: Implications for novel topical anticancer candidates. Biol. Pharm. Bull., 2016, 39(10), 1675-1682.
[http://dx.doi.org/10.1248/bpb.b16-00420] [PMID: 27725445]
[170]
Nath, L.R.; Kumar, S.N.; Das, A.A.; Nambisan, B.; Shabna, A.; Mohandas, C.; Anto, R.J. In vitro evaluation of the antioxidant, 3,5-dihydroxy-4-ethyl-trans-stilbene (DETS) isolated from bacillus cereus as a potent candidate against malignant melanoma. Front. Microbiol., 2016, 7, 452.
[http://dx.doi.org/10.3389/fmicb.2016.00452] [PMID: 27148169]
[171]
Androutsopoulos, V.P.; Fragiadaki, I.; Spandidos, D.A.; Tosca, A. The resveratrol analogue, 3,4,5,4′ trans-tetramethoxystilbene, inhibits the growth of A375 melanoma cells through multiple anticancer modes of action. Int. J. Oncol., 2016, 49(4), 1305-1314.
[http://dx.doi.org/10.3892/ijo.2016.3635] [PMID: 27498704]
[172]
Liu, Q.; Kim, C.; Jo, Y.H.; Kim, S.B.; Hwang, B.Y.; Lee, M.K. Synthesis and biological evaluation of resveratrol derivatives as melanogenesis inhibitors. Molecules, 2015, 20(9), 16933-16945.
[http://dx.doi.org/10.3390/molecules200916933] [PMID: 26393543]
[173]
Nivelle, L.; Hubert, J.; Courot, E.; Borie, N.; Renault, J-H.; Nuzillard, J-M.; Harakat, D.; Clément, C.; Martiny, L.; Delmas, D.; Jeandet, P.; Tarpin, M. Cytotoxicity of labruscol, a new resveratrol dimer produced by grapevine cell suspensions, on human skin melanoma cancer cell line HT-144. Molecules, 2017, 22(11), 1940.
[http://dx.doi.org/10.3390/molecules22111940] [PMID: 29120391]
[174]
Osmond, G.W.; Masko, E.M.; Tyler, D.S.; Freedland, S.J.; Pizzo, S. In vitro and in vivo evaluation of resveratrol and 3, 5-dihydroxy-4′-acetoxy-trans-stilbene in the treatment of human prostate carcinoma and melanoma. J. Surg. Res., 2013, 179(1), e141-e148.
[http://dx.doi.org/10.1016/j.jss.2012.02.057] [PMID: 22482756]
[175]
Morris, V.L.; Toseef, T.; Nazumudeen, F.B.; Rivoira, C.; Spatafora, C.; Tringali, C.; Rotenberg, S.A. Anti-tumor properties of cis-resveratrol methylated analogs in metastatic mouse melanoma cells. Mol. Cell. Biochem., 2015, 402(1-2), 83-91.
[http://dx.doi.org/10.1007/s11010-014-2316-8] [PMID: 25567208]
[176]
Park, J.; Park, J.H.; Suh, H-J.; Lee, I.C.; Koh, J.; Boo, Y.C. Effects of resveratrol, oxyresveratrol, and their acetylated derivatives on cellular melanogenesis. Arch. Dermatol. Res., 2014, 306(5), 475-487.
[http://dx.doi.org/10.1007/s00403-014-1440-3] [PMID: 24414332]
[177]
Park, S.; Seok, J.K.; Kwak, J.Y.; Choi, Y-H.; Hong, S.S.; Suh, H-J.; Park, W.; Boo, Y.C. Anti-melanogenic effects of resveratryl triglycolate, a novel hybrid compound derived by esterification of resveratrol with glycolic acid. Arch. Dermatol. Res., 2016, 308(5), 325-334.
[http://dx.doi.org/10.1007/s00403-016-1644-9] [PMID: 27059716]
[178]
Yoon, H-S.; Hyun, C-G.; Lee, N-H.; Park, S-S.; Shin, D-B. Comparative depigmentation effects of resveratrol and its two methyl analogues in α-melanocyte stimulating hormone-triggered b16/f10 murine melanoma cells. Prev. Nutr. Food Sci., 2016, 21(2), 155-159.
[http://dx.doi.org/10.3746/pnf.2016.21.2.155] [PMID: 27390733]
[179]
Chen, X.; Li, W.; Xu, C.; Wang, J.; Zhu, B.; Huang, Q.; Chen, D.; Sheng, J.; Zou, Y.; Lee, Y.M.; Tan, R.; Shen, P.; Wong, Y.K.; Lin, Q.; Wang, J.; Hua, Z. Comparative profiling of analog targets: a case study on resveratrol for mouse melanoma metastasis suppression. Theranostics, 2018, 8(13), 3504-3516.
[http://dx.doi.org/10.7150/thno.24336] [PMID: 30026862]
[180]
Benlloch, M.; Obrador, E.; Valles, S.L.; Rodriguez, M.L.; Sirerol, J.A.; Alcácer, J.; Pellicer, J.A.; Salvador, R.; Cerdá, C.; Sáez, G.T. Pterostilbene decreases the antioxidant defenses of aggressive cancer cells in vivo: a physiological glucocorticoids-and Nrf2-dependent mechanism. Antioxidants & redox signaling, , 2016, 24(17), 974-990.
[http://dx.doi.org/10.1089/ars.2015.6437]
[181]
Mena, S.; Rodríguez, M.L.; Ponsoda, X.; Estrela, J.M.; Jäättela, M.; Ortega, A.L. Pterostilbene-induced tumor cytotoxicity: a lysosomal membrane permeabilization-dependent mechanism. PLoS One, 2012, 7(9)e44524
[http://dx.doi.org/10.1371/journal.pone.0044524] [PMID: 22957077]
[182]
Nivelle, L.; Aires, V.; Rioult, D.; Martiny, L.; Tarpin, M.; Delmas, D. Molecular analysis of differential antiproliferative activity of resveratrol, epsilon viniferin and labruscol on melanoma cells and normal dermal cells. . Food Chem. Toxicol., 2018, 116(8), 323-334.
[http://dx.doi.org/10.1016/j.fct.2018.04.043]
[183]
Larrosa, M.; Tomás-Barberán, F.A.; Espín, J.C. Grape polyphenol resveratrol and the related molecule 4-hydroxystilbene induce growth inhibition, apoptosis, S-phase arrest, and upregulation of cyclins A, E, and B1 in human SK-Mel-28 melanoma cells. J. Agric. Food Chem., 2003, 51(16), 4576-4584.
[http://dx.doi.org/10.1021/jf030073c] [PMID: 14705880]
[184]
Amri, A.; Chaumeil, J.C.; Sfar, S.; Charrueau, C. Administration of resveratrol: What formulation solutions to bioavailability limitations? J. Control. Release, 2012, 158(2), 182-193.
[http://dx.doi.org/10.1016/j.jconrel.2011.09.083] [PMID: 21978644]
[185]
Davidov-Pardo, G.; McClements, D.J. Nutraceutical delivery systems: resveratrol encapsulation in grape seed oil nanoemulsions formed by spontaneous emulsification. Food Chem., 2015, 167, 205-212.
[http://dx.doi.org/10.1016/j.foodchem.2014.06.082] [PMID: 25148980]
[186]
Rius, C.; Abu-Taha, M.; Hermenegildo, C.; Piqueras, L.; Cerda-Nicolas, J.M.; Issekutz, A.C.; Estan, L.; Cortijo, J.; Morcillo, E.J.; Orallo, F.; Sanz, M.J. Trans- but not cis-resveratrol impairs angiotensin-II-mediated vascular inflammation through inhibition of NF-kappaB activation and peroxisome proliferator-activated receptor-gamma upregulation. J. Immunol., 2010, 185(6), 3718-3727.
[http://dx.doi.org/10.4049/jimmunol.1001043] [PMID: 20709957]
[187]
Camont, L.; Cottart, C.H.; Rhayem, Y.; Nivet-Antoine, V.; Djelidi, R.; Collin, F.; Beaudeux, J.L.; Bonnefont-Rousselot, D. Simple spectrophotometric assessment of the trans-/cis-resveratrol ratio in aqueous solutions. Anal. Chim. Acta, 2009, 634(1), 121-128.
[http://dx.doi.org/10.1016/j.aca.2008.12.003] [PMID: 19154820]
[188]
Planas, J.M.; Alfaras, I.; Colom, H.; Juan, M.E. The bioavailability and distribution of trans-resveratrol are constrained by ABC transporters. Arch. Biochem. Biophys., 2012, 527(2), 67-73.
[http://dx.doi.org/10.1016/j.abb.2012.06.004] [PMID: 22750234]
[189]
Radko, Y.; Christensen, K.B.; Christensen, L.P. Semi-preparative isolation of dihydroresveratrol-3-O-β-d-glucuronide and four resveratrol conjugates from human urine after oral intake of a resveratrol-containing dietary supplement. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci., 2013, 930, 54-61.
[http://dx.doi.org/10.1016/j.jchromb.2013.05.002] [PMID: 23727867]
[190]
Ruotolo, R.; Calani, L.; Fietta, E.; Brighenti, F.; Crozier, A.; Meda, C.; Maggi, A.; Ottonello, S.; Del Rio, D. Anti-estrogenic activity of a human resveratrol metabolite. Nutr. Metab. Cardiovasc. Dis., 2013, 23(11), 1086-1092.
[http://dx.doi.org/10.1016/j.numecd.2013.01.002] [PMID: 23465317]
[191]
Paolino, D.; Cosco, D.; Muzzalupo, R.; Trapasso, E.; Picci, N.; Fresta, M. Innovative bola-surfactant niosomes as topical delivery systems of 5-fluorouracil for the treatment of skin cancer. Int. J. Pharm., 2008, 353(1-2), 233-242.
[http://dx.doi.org/10.1016/j.ijpharm.2007.11.037] [PMID: 18191509]
[192]
González-Paredes, A.; Manconi, M.; Caddeo, C.; Ramos-Cormenzana, A.; Monteoliva-Sánchez, M.; Fadda, A.M. Archaeosomes as carriers for topical delivery of betamethasone dipropionate: in vitro skin permeation study. J. Liposome Res., 2010, 20(4), 269-276.
[http://dx.doi.org/10.3109/08982100903402962] [PMID: 19954402]
[193]
Marianecci, C.; Rinaldi, F.; Mastriota, M.; Pieretti, S.; Trapasso, E.; Paolino, D.; Carafa, M. Anti-inflammatory activity of novel ammonium glycyrrhizinate/niosomes delivery system: human and murine models. J. Control. Release, 2012, 164(1), 17-25.
[http://dx.doi.org/10.1016/j.jconrel.2012.09.018] [PMID: 23041542]
[194]
Sinico, C.; Fadda, A.M. Vesicular carriers for dermal drug delivery. Expert Opin. Drug Deliv., 2009, 6(8), 813-825.
[http://dx.doi.org/10.1517/17425240903071029] [PMID: 19569979]
[195]
Elsayed, M.M.; Abdallah, O.Y.; Naggar, V.F.; Khalafallah, N.M. Deformable liposomes and ethosomes as carriers for skin delivery of ketotifen. Pharmazie, 2007, 62(2), 133-137.
[PMID: 17341034]
[196]
Ainbinder, D.; Paolino, D.; Fresta, M.; Touitou, E. Drug delivery applications with ethosomes. J. Biomed. Nanotechnol., 2010, 6(5), 558-568.
[http://dx.doi.org/10.1166/jbn.2010.1152] [PMID: 21329048]
[197]
Cevc, G.; Vierl, U. Nanotechnology and the transdermal route: A state of the art review and critical appraisal. J. Control. Release, 2010, 141(3), 277-299.
[http://dx.doi.org/10.1016/j.jconrel.2009.10.016] [PMID: 19850095]
[198]
El Zaafarany, G.M.; Awad, G.A.; Holayel, S.M.; Mortada, N.D. Role of edge activators and surface charge in developing ultradeformable vesicles with enhanced skin delivery. Int. J. Pharm., 2010, 397(1-2), 164-172.
[http://dx.doi.org/10.1016/j.ijpharm.2010.06.034] [PMID: 20599487]
[199]
Benson, H.A. Transfersomes for transdermal drug delivery. Expert Opin. Drug Deliv., 2006, 3(6), 727-737.
[http://dx.doi.org/10.1517/17425247.3.6.727] [PMID: 17076595]
[200]
Cosco, D.; Paolino, D.; Maiuolo, J.; Marzio, L.D.; Carafa, M.; Ventura, C.A.; Fresta, M. Ultradeformable liposomes as multidrug carrier of resveratrol and 5-fluorouracil for their topical delivery. Int. J. Pharm., 2015, 489(1-2), 1-10.
[http://dx.doi.org/10.1016/j.ijpharm.2015.04.056] [PMID: 25899287]
[201]
Parveen, S.; Misra, R.; Sahoo, S.K. Nanoparticles: a boon to drug delivery, therapeutics, diagnostics and imaging. Nanomedicine (Lond.), 2012, 8(2), 147-166.
[http://dx.doi.org/10.1016/j.nano.2011.05.016] [PMID: 21703993]
[202]
Mattos, A.C.; Altmeyer, C.; Tominaga, T.T.; Khalil, N.M.; Mainardes, R.M. Polymeric nanoparticles for oral delivery of 5-fluorouracil: formulation optimization, cytotoxicity assay and pre-clinical pharmacokinetics study. Eur. J. Pharm. Sci., 2016, 84, 83-91.
[http://dx.doi.org/10.1016/j.ejps.2016.01.012] [PMID: 26775869]
[203]
Feng, S.S. Nanoparticles of biodegradable polymers for new-concept chemotherapy. Expert Rev. Med. Devices, 2004, 1(1), 115-125.
[http://dx.doi.org/10.1586/17434440.1.1.115] [PMID: 16293015]
[204]
Figueiró, F.; Bernardi, A.; Frozza, R.L.; Terroso, T.; Zanotto-Filho, A.; Jandrey, E.H.; Moreira, J.C.; Salbego, C.G.; Edelweiss, M.I.; Pohlmann, A.R.; Guterres, S.S.; Battastini, A.M. Resveratrol-loaded lipid-core nanocapsules treatment reduces in vitro and in vivo glioma growth. J. Biomed. Nanotechnol., 2013, 9(3), 516-526.
[http://dx.doi.org/10.1166/jbn.2013.1547] [PMID: 23621009]
[205]
Sanna, V.; Siddiqui, I.A.; Sechi, M.; Mukhtar, H. Resveratrol-loaded nanoparticles based on poly(epsilon-caprolactone) and poly(D,L-lactic-co-glycolic acid)-poly(ethylene glycol) blend for prostate cancer treatment. Mol. Pharm., 2013, 10(10), 3871-3881.
[http://dx.doi.org/10.1021/mp400342f] [PMID: 23968375]
[206]
Carletto, B.; Berton, J.; Ferreira, T.N.; Dalmolin, L.F.; Paludo, K.S.; Mainardes, R.M.; Farago, P.V.; Favero, G.M. Resveratrol-loaded nanocapsules inhibit murine melanoma tumor growth. Colloids Surf. B Biointerfaces, 2016, 144, 65-72.
[http://dx.doi.org/10.1016/j.colsurfb.2016.04.001] [PMID: 27070053]
[207]
Blanco, E.; Shen, H.; Ferrari, M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat. Biotechnol., 2015, 33(9), 941-951.
[http://dx.doi.org/10.1038/nbt.3330] [PMID: 26348965]
[208]
Leibowitz-Amit, R.; Sidi, Y.; Avni, D. Aberrations in the micro-RNA biogenesis machinery and the emerging roles of micro-RNAs in the pathogenesis of cutaneous malignant melanoma. Pigment Cell Melanoma Res., 2012, 25(6), 740-757.
[http://dx.doi.org/10.1111/pcmr.12018] [PMID: 22958787]
[209]
Lujambio, A.; Lowe, S.W. The microcosmos of cancer. Nature, 2012, 482(7385), 347-355.
[http://dx.doi.org/10.1038/nature10888] [PMID: 22337054]
[210]
Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol., 2014, 15(8), 509-524.
[http://dx.doi.org/10.1038/nrm3838] [PMID: 25027649]
[211]
Li, J.; Chen, J.; Wang, S.; Li, P.; Zheng, C.; Zhou, X.; Tao, Y.; Chen, X.; Sun, L.; Wang, A.; Cao, K.; Tang, S.; Zhou, J. Blockage of transferred exosome-shuttled miR-494 inhibits melanoma growth and metastasis. J. Cell. Physiol., 2019. Epub ahead of print
[http://dx.doi.org/10.1002/jcp.28234] [PMID: 30723916]
[212]
Ushio, N.; Rahman, M.M.; Maemura, T.; Lai, Y.C.; Iwanaga, T.; Kawaguchi, H.; Miyoshi, N.; Momoi, Y.; Miura, N. Identification of dysregulated microRNAs in canine malignant melanoma. Oncol. Lett., 2019, 17(1), 1080-1088.
[http://dx.doi.org/10.3892/ol.2018.9692] [PMID: 30655868]
[213]
Weber, C.E.; Luo, C.; Hotz-Wagenblatt, A.; Gardyan, A.; Kordaß, T.; Holland-Letz, T.; Osen, W.; Eichmüller, S.B. miR-339-3p is a tumor suppressor in melanoma. Cancer Res., 2016, 76(12), 3562-3571.
[http://dx.doi.org/10.1158/0008-5472.CAN-15-2932] [PMID: 27197185]
[214]
Mattia, G.; Errico, M.C.; Felicetti, F.; Petrini, M.; Bottero, L.; Tomasello, L.; Romania, P.; Boe, A.; Segnalini, P.; Di Virgilio, A.; Colombo, M.P.; Carè, A. Constitutive activation of the ETS-1-miR-222 circuitry in metastatic melanoma. Pigment Cell Melanoma Res., 2011, 24(5), 953-965.
[http://dx.doi.org/10.1111/j.1755-148X.2011.00881.x] [PMID: 21711453]
[215]
Quintavalle, C.; Garofalo, M.; Zanca, C.; Romano, G.; Iaboni, M.; del Basso De Caro, M.; Martinez-Montero, J.C.; Incoronato, M.; Nuovo, G.; Croce, C.M.; Condorelli, G. miR-221/222 overexpession in human glioblastoma increases invasiveness by targeting the protein phosphate PTPμ. Oncogene, 2012, 31(7), 858-868.
[http://dx.doi.org/10.1038/onc.2011.280] [PMID: 21743492]
[216]
Babapoor, S.; Wu, R.; Kozubek, J.; Auidi, D.; Grant-Kels, J.M.; Dadras, S.S. Identification of microRNAs associated with invasive and aggressive phenotype in cutaneous melanoma by next-generation sequencing. Lab. Invest., 2017, 97(6), 636-648.
[http://dx.doi.org/10.1038/labinvest.2017.5] [PMID: 28218741]
[217]
Garofalo, M.; Quintavalle, C.; Romano, G.; Croce, C.M.; Condorelli, G. miR221/222 in cancer: their role in tumor progression and response to therapy. Curr. Mol. Med., 2012, 12(1), 27-33.
[http://dx.doi.org/10.2174/156652412798376170] [PMID: 22082479]
[218]
Panday, A.; Inda, M.E.; Bagam, P.; Sahoo, M.K.; Osorio, D.; Batra, S. Transcription factor NF-κB: an update on intervention strategies. Arch. Immunol. Ther. Exp. (Warsz.), 2016, 64(6), 463-483.
[http://dx.doi.org/10.1007/s00005-016-0405-y] [PMID: 27236331]
[219]
Mobley, A.K.; Braeuer, R.R.; Kamiya, T.; Shoshan, E.; Bar-Eli, M. Driving transcriptional regulators in melanoma metastasis. Cancer Metastasis Rev., 2012, 31(3-4), 621-632.
[http://dx.doi.org/10.1007/s10555-012-9358-8] [PMID: 22684365]
[220]
Bassères, D.S.; Ebbs, A.; Levantini, E.; Baldwin, A.S. Requirement of the NF-κB subunit p65/RelA for K-Ras–induced lung tumorigenesis. Cancer Res., 2010, 70(9), 3537-3546.
[http://dx.doi.org/10.1158/0008-5472.CAN-09-4290] [PMID: 20406971]
[221]
Gallagher, S.J.; Mijatov, B.; Gunatilake, D.; Gowrishankar, K.; Tiffen, J.; James, W.; Jin, L.; Pupo, G.; Cullinane, C.; McArthur, G.A.; Tummino, P.J.; Rizos, H.; Hersey, P. Control of NF-kB activity in human melanoma by bromodomain and extra-terminal protein inhibitor I-BET151. Pigment Cell Melanoma Res., 2014, 27(6), 1126-1137.
[http://dx.doi.org/10.1111/pcmr.12282] [PMID: 24924589]
[222]
Zehavi, L.; Schayek, H.; Jacob-Hirsch, J.; Sidi, Y.; Leibowitz-Amit, R.; Avni, D. MiR-377 targets E2F3 and alters the NF-kB signaling pathway through MAP3K7 in malignant melanoma. Mol. Cancer, 2015, 14(1), 68.
[http://dx.doi.org/10.1186/s12943-015-0338-9] [PMID: 25889255]
[223]
Giles, K.M.; Brown, R.A.; Ganda, C.; Podgorny, M.J.; Candy, P.A.; Wintle, L.C.; Richardson, K.L.; Kalinowski, F.C.; Stuart, L.M.; Epis, M.R.; Haass, N.K.; Herlyn, M.; Leedman, P.J. microRNA-7-5p inhibits melanoma cell proliferation and metastasis by suppressing RelA/NF-κB. Oncotarget, 2016, 7(22), 31663-31680.
[http://dx.doi.org/10.18632/oncotarget.9421] [PMID: 27203220]
[224]
Ren, Z.; Wang, L.; Cui, J.; Huoc, Z.; Xue, J.; Cui, H.; Mao, Q.; Yang, R. Resveratrol inhibits NF-kB signaling through suppression of p65 and IkappaB kinase activities. Pharmazie, 2013, 68(8), 689-694.
[PMID: 24020126]
[225]
Takeuchi, S.; Masuda, C.; Maebayashi, H.; Tooyama, I. Immunohistochemical mapping of TRK-fused gene products in the rat brainstem. Acta Histochem. Cytochem., 2012, 45(1), 57-64.
[http://dx.doi.org/10.1267/ahc.11051] [PMID: 22489105]
[226]
Dutton-Regester, K.; Aoude, L.G.; Nancarrow, D.J.; Stark, M.S.; O’Connor, L.; Lanagan, C.; Pupo, G.M.; Tembe, V.; Carter, C.D.; O’Rourke, M.; Scolyer, R.A.; Mann, G.J.; Schmidt, C.W.; Herington, A.; Hayward, N.K. Identification of TFG (TRK-fused gene) as a putative metastatic melanoma tumor suppressor gene. Genes Chromosomes Cancer, 2012, 51(5), 452-461.
[http://dx.doi.org/10.1002/gcc.21932] [PMID: 22250051]
[227]
Tili, E.; Michaille, J.J.; Gandhi, V.; Plunkett, W.; Sampath, D.; Calin, G.A. miRNAs and their potential for use against cancer and other diseases. Future Oncol., 2007, 3(5), 521-537.
[http://dx.doi.org/10.2217/14796694.3.5.521] [PMID: 17927518]
[228]
Otsuka, K.; Yamamoto, Y.; Ochiya, T. Regulatory role of resveratrol, a microRNA-controlling compound, in HNRNPA1 expression, which is associated with poor prognosis in breast cancer. Oncotarget, 2018, 9(37), 24718-24730.
[http://dx.doi.org/10.18632/oncotarget.25339] [PMID: 29872500]
[229]
Wu, H.; Wang, Y.; Wu, C.; Yang, P.; Li, H.; Li, Z. Resveratrol induces cancer cell apoptosis through MiR-326/PKM2-mediated ER stress and mitochondrial fission. J. Agric. Food Chem., 2016, 64(49), 9356-9367.
[http://dx.doi.org/10.1021/acs.jafc.6b04549] [PMID: 27960279]
[230]
Karimi Dermani, F.; Saidijam, M.; Amini, R.; Mahdavinezhad, A.; Heydari, K.; Najafi, R. Resveratrol inhibits proliferation, invasion, and epithelial-mesenchymal transition by increasing miR-200c expression in HCT-116 colorectal cancer cells. J. Cell. Biochem., 2017, 118(6), 1547-1555.
[http://dx.doi.org/10.1002/jcb.25816] [PMID: 27918105]
[231]
Yang, S.; Li, W.; Sun, H.; Wu, B.; Ji, F.; Sun, T.; Chang, H.; Shen, P.; Wang, Y.; Zhou, D. Resveratrol elicits anti-colorectal cancer effect by activating miR-34c-KITLG in vitro and in vivo. BMC Cancer, 2015, 15, 969.
[http://dx.doi.org/10.1186/s12885-015-1958-6] [PMID: 26674205]
[232]
Zhou, W.; Wang, S.; Ying, Y.; Zhou, R.; Mao, P. miR-196b/miR-1290 participate in the antitumor effect of resveratrol via regulation of IGFBP3 expression in acute lymphoblastic leukemia. Oncol. Rep., 2017, 37(2), 1075-1083.
[http://dx.doi.org/10.3892/or.2016.5321] [PMID: 28000876]
[233]
Dhar, S.; Kumar, A.; Rimando, A.M.; Zhang, X.; Levenson, A.S. Resveratrol and pterostilbene epigenetically restore PTEN expression by targeting oncomiRs of the miR-17 family in prostate cancer. Oncotarget, 2015, 6(29), 27214-27226.
[http://dx.doi.org/10.18632/oncotarget.4877] [PMID: 26318586]
[234]
Yang, S.F.; Lee, W.J.; Tan, P.; Tang, C.H.; Hsiao, M.; Hsieh, F.K.; Chien, M.H. Upregulation of miR-328 and inhibition of CREB-DNA-binding activity are critical for resveratrol-mediated suppression of matrix metalloproteinase-2 and subsequent metastatic ability in human osteosarcomas. Oncotarget, 2015, 6(5), 2736-2753.
[http://dx.doi.org/10.18632/oncotarget.3088] [PMID: 25605016]
[235]
Ren, X.; Bai, X.; Zhang, X.; Li, Z.; Tang, L.; Zhao, X.; Li, Z.; Ren, Y.; Wei, S.; Wang, Q.; Liu, C.; Ji, J. Quantitative nuclear proteomics identifies that miR-137-mediated EZH2 reduction regulates resveratrol-induced apoptosis of neuroblastoma cells. Mol. Cell. Proteomics, 2015, 14(2), 316-328.
[http://dx.doi.org/10.1074/mcp.M114.041905] [PMID: 25505154]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy