메뉴 건너뛰기




Volumn 21, Issue 12, 2012, Pages 1801-1818

Terpenoids: Natural products for cancer therapy

Author keywords

Cancer; Mechanisms; Natural products; Terpenoids; Traditional Chinese medicine

Indexed keywords

ALISOL B; ANDROGRAPHOLIDE; ANTINEOPLASTIC AGENT; ARTEMISININ; CANTHARIDIN; CELASTROL; CUCURBITACIN; DIHYDROARTEMISININ; LIMONENE; LYCOPENE; NATURAL PRODUCT; ORIDONIN; PACHYMIC ACID; PSEUDOLARIC ACID B; TANSHINONE IIA; TERPENOID DERIVATIVE; TRIPTOLIDE; UNCLASSIFIED DRUG;

EID: 84868685907     PISSN: 13543784     EISSN: 17447658     Source Type: Journal    
DOI: 10.1517/13543784.2012.727395     Document Type: Review
Times cited : (261)

References (198)
  • 1
    • 0042844744 scopus 로고    scopus 로고
    • Natural products as sources of new drugs over the period 1981-2002
    • Newman DJ, Cragg GM, Snader KM. Natural products as sources of new drugs over the period 1981-2002. J Nat Prod 2003;66:1022-37
    • (2003) J Nat Prod , vol.66 , pp. 1022-1037
    • Newman, D.J.1    Cragg, G.M.2    Snader, K.M.3
  • 2
    • 34250785114 scopus 로고    scopus 로고
    • The function of terpene natural products in the natural world
    • Gershenzon J, Dudareva N. The function of terpene natural products in the natural world. Nat Chem Biol 2007;3:408-14
    • (2007) Nat Chem Biol , vol.3 , pp. 408-414
    • Gershenzon, J.1    Dudareva, N.2
  • 3
    • 84873856866 scopus 로고    scopus 로고
    • Development of artemisinin compounds for cancer treatment
    • [Epub ahead of print]
    • Lai HC, Singh NP, Sasaki T. et al. Development of artemisinin compounds for cancer treatment. Invest New Drugs 2012. [Epub ahead of print]
    • (2012) Invest New Drugs
    • Lai, H.C.1    Singh, N.P.2    Sasaki, T.3
  • 5
    • 35348929897 scopus 로고    scopus 로고
    • D-Limonene: Safety and clinical applications
    • Sun J. D-Limonene: safety and clinical applications. Altern Med Rev 2007;12:259-64
    • (2007) Altern Med Rev , vol.12 , pp. 259-264
    • Sun, J.1
  • 6
    • 67649484490 scopus 로고    scopus 로고
    • D-Limonene sensitizes docetaxel-induced cytotoxicity in human prostate cancer cells: Generation of reactive oxygen species and induction of apoptosis
    • Rabi T, Bishayee A. D-Limonene sensitizes docetaxel-induced cytotoxicity in human prostate cancer cells: generation of reactive oxygen species and induction of apoptosis. J Carcinog 2009;8:9
    • (2009) J Carcinog , vol.8 , pp. 9
    • Rabi, T.1    Bishayee, A.2
  • 7
    • 0031834303 scopus 로고    scopus 로고
    • Phase i and pharmacokinetic study of D-limonene in patients with advanced cancer. Cancer Research Campaign Phase I/II Clinical Trials Committee
    • Vigushin DM, Poon GK, Boddy A, et al. Phase I and pharmacokinetic study of D-limonene in patients with advanced cancer. Cancer Research Campaign Phase I/II Clinical Trials Committee. Cancer Chemother Pharmacol 1998;42:111-17
    • (1998) Cancer Chemother Pharmacol , vol.42 , pp. 111-117
    • Vigushin, D.M.1    Poon, G.K.2    Boddy, A.3
  • 8
    • 0019941618 scopus 로고
    • The mechanism of cyclic monoterpene inhibition of hepatic 3-hydroxy-3-methylglutaryl coenzyme A reductase in vivo in the rat
    • Clegg RJ, Middleton B, Bell GD, et al. The mechanism of cyclic monoterpene inhibition of hepatic 3-hydroxy-3-methylglutaryl coenzyme A reductase in vivo in the rat. J Biol Chem 1982;257:2294-9
    • (1982) J Biol Chem , vol.257 , pp. 2294-2299
    • Clegg, R.J.1    Middleton, B.2    Bell, G.D.3
  • 9
    • 0028300763 scopus 로고
    • Modulation of the mevalonate pathway and cell growth by pravastatin and d-limonene in a human hepatoma cell line (Hep G2)
    • Kawata S, Nagase T, Yamasaki E, et al. Modulation of the mevalonate pathway and cell growth by pravastatin and d-limonene in a human hepatoma cell line (Hep G2). Br J Cancer 1994;69:1015-20
    • (1994) Br J Cancer , vol.69 , pp. 1015-1020
    • Kawata, S.1    Nagase, T.2    Yamasaki, E.3
  • 10
    • 0035427196 scopus 로고    scopus 로고
    • Inhibition by d-limonene of experimental hepatocarcinogenesis in Sprague-Dawley rats does not involve p21(ras) plasma membrane association
    • Kaji I, Tatsuta M, Iishi H, et al. Inhibition by d-limonene of experimental hepatocarcinogenesis in Sprague-Dawley rats does not involve p21(ras) plasma membrane association. Int J Cancer 2001;93:441-4
    • (2001) Int J Cancer , vol.93 , pp. 441-444
    • Kaji, I.1    Tatsuta, M.2    Iishi, H.3
  • 11
    • 33845590531 scopus 로고    scopus 로고
    • Induction of apoptosis by d-limonene is mediated by a caspase-dependent mitochondrial death pathway in human leukemia cells
    • Ji J, Zhang L, Wu YY, et al. Induction of apoptosis by d-limonene is mediated by a caspase-dependent mitochondrial death pathway in human leukemia cells. Leuk Lymphoma 2006;47:2617-24
    • (2006) Leuk Lymphoma , vol.47 , pp. 2617-2624
    • Ji, J.1    Zhang, L.2    Wu, Y.Y.3
  • 12
    • 67649817034 scopus 로고    scopus 로고
    • The effects of cantharidin and cantharidin derivates on tumour cells
    • Liu D, Chen Z. The effects of cantharidin and cantharidin derivates on tumour cells. Anticancer Agents Med Chem 2009;9:392-6
    • (2009) Anticancer Agents Med Chem , vol.9 , pp. 392-396
    • Liu, D.1    Chen, Z.2
  • 13
    • 0034618205 scopus 로고    scopus 로고
    • Anhydride modified cantharidin analogues: Synthesis inhibition of protein phosphatases 1 and 2A and anticancer activity
    • McCluskey A, Bowyer MC, Collins E, et al. Anhydride modified cantharidin analogues: synthesis, inhibition of protein phosphatases 1 and 2A and anticancer activity. Bioorg Med Chem Lett 2000;10:1687-90
    • (2000) Bioorg Med Chem Lett , vol.10 , pp. 1687-1690
    • McCluskey, A.1    Bowyer, M.C.2    Collins, E.3
  • 14
    • 0014741374 scopus 로고
    • Studies on the mechanism of skin tumor promotion
    • Hennings H, Boutwell RK. Studies on the mechanism of skin tumor promotion. Cancer Res 1970;30:312-20
    • (1970) Cancer Res , vol.30 , pp. 312-320
    • Hennings, H.1    Boutwell, R.K.2
  • 15
    • 0037054929 scopus 로고    scopus 로고
    • Effector mechanisms of norcantharidin-induced mitotic arrest and apoptosis in human hepatoma cells
    • Chen YN, Chen JC, Yin SC, et al. Effector mechanisms of norcantharidin-induced mitotic arrest and apoptosis in human hepatoma cells. Int J Cancer 2002;100:158-65
    • (2002) Int J Cancer , vol.100 , pp. 158-165
    • Chen, Y.N.1    Chen, J.C.2    Yin, S.C.3
  • 16
    • 33646751454 scopus 로고    scopus 로고
    • Cantharidin-induced cytotoxicity and cyclooxygenase 2 expression in human bladder carcinoma cell line
    • Huan SK, Lee HH, Liu DZ, et al. Cantharidin-induced cytotoxicity and cyclooxygenase 2 expression in human bladder carcinoma cell line. Toxicology 2006;223:136-43
    • (2006) Toxicology , vol.223 , pp. 136-143
    • Huan, S.K.1    Lee, H.H.2    Liu, D.Z.3
  • 17
    • 2342535824 scopus 로고    scopus 로고
    • Roles of p38 and JNK mitogen-activated protein kinase pathways during cantharidin-induced apoptosis in U937 cells
    • Huh JE, Kang KS, Chae C, et al. Roles of p38 and JNK mitogen-activated protein kinase pathways during cantharidin-induced apoptosis in U937 cells. Biochem Pharmacol 2004;67:1811-18
    • (2004) Biochem Pharmacol , vol.67 , pp. 1811-1818
    • Huh, J.E.1    Kang, K.S.2    Chae, C.3
  • 18
    • 0027217027 scopus 로고
    • Cantharidin, another natural toxin that inhibits the activity of serine/threonine protein phosphatases types 1 and 2A
    • Honkanen RE. Cantharidin, another natural toxin that inhibits the activity of serine/threonine protein phosphatases types 1 and 2A. FEBS Lett 1993;330:283-6
    • (1993) FEBS Lett , vol.330 , pp. 283-286
    • Honkanen, R.E.1
  • 19
    • 0036208532 scopus 로고    scopus 로고
    • Anticancer activity and protein phosphatase 1 and 2A inhibition of a new generation of cantharidin analogues
    • Sakoff JA, Ackland SP, Baldwin ML, et al. Anticancer activity and protein phosphatase 1 and 2A inhibition of a new generation of cantharidin analogues. Invest New Drugs 2002;20:1-11
    • (2002) Invest New Drugs , vol.20 , pp. 1-11
    • Sakoff, J.A.1    Ackland, S.P.2    Baldwin, M.L.3
  • 20
    • 0037305812 scopus 로고    scopus 로고
    • Cantharidin analogues: Synthesis and evaluation of growth inhibition in a panel of selected tumour cell lines
    • McCluskey A, Ackland SP, Bowyer MC, et al. Cantharidin analogues: synthesis and evaluation of growth inhibition in a panel of selected tumour cell lines. Bioorg Chem 2003;31:68-79
    • (2003) Bioorg Chem , vol.31 , pp. 68-79
    • McCluskey, A.1    Ackland, S.P.2    Bowyer, M.C.3
  • 21
    • 68549115381 scopus 로고    scopus 로고
    • Structural basis of serine/threonine phosphatase inhibition by the archetypal small molecules cantharidin and norcantharidin
    • Bertini I, Calderone V, Fragai M, et al. Structural basis of serine/threonine phosphatase inhibition by the archetypal small molecules cantharidin and norcantharidin. J Med Chem 2009;52:4838-43
    • (2009) J Med Chem , vol.52 , pp. 4838-4843
    • Bertini, I.1    Calderone, V.2    Fragai, M.3
  • 22
    • 79953000069 scopus 로고    scopus 로고
    • Protein phosphatase 2a as a potential target for anticancer therapy
    • Kalev P, Sablina AA. Protein phosphatase 2A as a potential target for anticancer therapy. Anticancer Agents Med Chem 2011;11:38-46
    • (2011) Anticancer Agents Med Chem , vol.11 , pp. 38-46
    • Kalev, P.1    Sablina, A.A.2
  • 23
    • 79952739700 scopus 로고    scopus 로고
    • PP2A inhibitors induce apoptosis in pancreatic cancer cell line PANC-1 through persistent phosphorylation of IKKalpha and sustained activation of the NF-kappaB pathway
    • Li W, Chen Z, Zong Y, et al. PP2A inhibitors induce apoptosis in pancreatic cancer cell line PANC-1 through persistent phosphorylation of IKKalpha and sustained activation of the NF-kappaB pathway. Cancer Lett 2011;304:117-27
    • (2011) Cancer Lett , vol.304 , pp. 117-127
    • Li, W.1    Chen, Z.2    Zong, Y.3
  • 24
    • 77953204596 scopus 로고    scopus 로고
    • Cantharidin, a potent and selective PP2A inhibitor, induces an oxidative stress-independent growth inhibition of pancreatic cancer cells through G2/M cell-cycle arrest and apoptosis
    • Li W, Xie L, Chen Z, et al. Cantharidin, a potent and selective PP2A inhibitor, induces an oxidative stress-independent growth inhibition of pancreatic cancer cells through G2/M cell-cycle arrest and apoptosis. Cancer Sci 2010;101:1226-33
    • (2010) Cancer Sci , vol.101 , pp. 1226-1233
    • Li, W.1    Xie, L.2    Chen, Z.3
  • 25
    • 0005289665 scopus 로고    scopus 로고
    • Artemisinin antimalarials: Mechanisms of action and resistance
    • Meshnick SR. Artemisinin antimalarials: mechanisms of action and resistance. Med Trop (Mars) 1998;58:13-17
    • (1998) Med Trop (Mars) , vol.58 , pp. 13-17
    • Meshnick, S.R.1
  • 26
    • 79959959354 scopus 로고    scopus 로고
    • The state of the art in anti-malarial drug discovery and development
    • Burrows JN, Chibale K, Wells TN. The state of the art in anti-malarial drug discovery and development. Curr Top Med Chem 2011;11:1226-54
    • (2011) Curr Top Med Chem , vol.11 , pp. 1226-1254
    • Burrows, J.N.1    Chibale, K.2    Wells, T.N.3
  • 27
    • 84863497510 scopus 로고    scopus 로고
    • Artemisinin: The gifts from traditional chinese medicine not only for malaria control but also for schistosomiasis control
    • Liu R, Dong HF, Jiang MS. Artemisinin: the gifts from traditional Chinese medicine not only for malaria control but also for schistosomiasis control. Parasitol Res 2011;110(5):2071-4
    • (2011) Parasitol Res , vol.110 , Issue.5 , pp. 2071-2074
    • Liu, R.1    Dong, H.F.2    Jiang, M.S.3
  • 28
    • 22244471426 scopus 로고    scopus 로고
    • Synthesis and immunosuppressive activity of new artemisinin derivatives. 1. [12 (beta or alpha)-Dihydroartemisininoxy] phen(ox)yl aliphatic acids and esters
    • Yang ZS, Zhou WL, Sui Y, et al. Synthesis and immunosuppressive activity of new artemisinin derivatives. 1. [12 (beta or alpha)-Dihydroartemisininoxy] phen(ox)yl aliphatic acids and esters. J Med Chem 2005;48:4608-17
    • (2005) J Med Chem , vol.484 , pp. 608-617
    • Yang, Z.S.1    Zhou, W.L.2    Sui, Y.3
  • 29
    • 77952795743 scopus 로고    scopus 로고
    • Anticancer activities of artemisinin and its bioactive derivatives
    • Firestone GL, Sundar SN. Anticancer activities of artemisinin and its bioactive derivatives. Expert Rev Mol Med 2009;11:e32
    • (2009) Expert Rev Mol Med , vol.11
    • Firestone, G.L.1    Sundar, S.N.2
  • 30
    • 79960555687 scopus 로고    scopus 로고
    • Anti-cancer natural products isolated from chinese medicinal herbs
    • Tan W, Lu J, Huang M, et al. Anti-cancer natural products isolated from chinese medicinal herbs. Chin Med 2011;6:27
    • (2011) Chin Med , vol.6 , pp. 27
    • Tan, W.1    Lu, J.2    Huang, M.3
  • 31
    • 75749141972 scopus 로고    scopus 로고
    • Artemisinin and its derivatives: A novel class of anti-malarial and anti-cancer agents
    • Chaturvedi D, Goswami A, Saikia PP, et al. Artemisinin and its derivatives: a novel class of anti-malarial and anti-cancer agents. Chem Soc Rev 2010;39:435-54
    • (2010) Chem Soc Rev , vol.39 , pp. 435-454
    • Chaturvedi, D.1    Goswami, A.2    Saikia, P.P.3
  • 32
    • 34347389476 scopus 로고    scopus 로고
    • Dihydroartemisinin is an inhibitor of ovarian cancer cell growth
    • Jiao Y, Ge CM, Meng QH, et al. Dihydroartemisinin is an inhibitor of ovarian cancer cell growth. Acta Pharmacol Sin 2007;28:1045-56
    • (2007) Acta Pharmacol Sin , vol.28 , pp. 1045-1056
    • Jiao, Y.1    Ge, C.M.2    Meng, Q.H.3
  • 33
    • 53049086416 scopus 로고    scopus 로고
    • Experimental therapy of hepatoma with artemisinin and its derivatives: In vitro and in vivo activity, chemosensitization, and mechanisms of action
    • Hou J, Wang D, Zhang R, et al. Experimental therapy of hepatoma with artemisinin and its derivatives: in vitro and in vivo activity, chemosensitization, and mechanisms of action. Clin Cancer Res 2008;14:5519-30
    • (2008) Clin Cancer Res , vol.14 , pp. 5519-5530
    • Hou, J.1    Wang, D.2    Zhang, R.3
  • 34
    • 56149102126 scopus 로고    scopus 로고
    • Dihydroartemisinin induces apoptosis in HL-60 leukemia cells dependent of iron and p38 mitogen-activated protein kinase activation but independent of reactive oxygen species
    • Lu JJ, Meng LH, Cai YJ, et al. Dihydroartemisinin induces apoptosis in HL-60 leukemia cells dependent of iron and p38 mitogen-activated protein kinase activation but independent of reactive oxygen species. Cancer Biol Ther 2008;7:1017-23
    • (2008) Cancer Biol Ther , vol.7 , pp. 1017-1023
    • Lu, J.J.1    Meng, L.H.2    Cai, Y.J.3
  • 35
    • 59449109184 scopus 로고    scopus 로고
    • Dihydroartemisinin inhibits growth of pancreatic cancer cells in vitro and in vivo
    • Chen H, Sun B, Pan S, et al. Dihydroartemisinin inhibits growth of pancreatic cancer cells in vitro and in vivo. Anticancer Drugs 2009;20:131-40
    • (2009) Anticancer Drugs , vol.20 , pp. 131-140
    • Chen, H.1    Sun, B.2    Pan, S.3
  • 36
    • 77953714651 scopus 로고    scopus 로고
    • Dihydroartemisinin upregulates death receptor 5 expression and cooperates with trail to induce apoptosis in human prostate cancer cells
    • He Q, Shi J, Shen XL, et al. Dihydroartemisinin upregulates death receptor 5 expression and cooperates with TRAIL to induce apoptosis in human prostate cancer cells. Cancer Biol Ther 2010;9:819-24
    • (2010) Cancer Biol Ther , vol.9 , pp. 819-824
    • He, Q.1    Shi, J.2    Shen, X.L.3
  • 37
    • 84856035765 scopus 로고    scopus 로고
    • The anti-cancer activity of dihydroartemisinin is associated with induction of iron-dependent endoplasmic reticulum stress in colorectal carcinoma HCT116 cells
    • Lu JJ, Chen SM, Zhang XW, et al. The anti-cancer activity of dihydroartemisinin is associated with induction of iron-dependent endoplasmic reticulum stress in colorectal carcinoma HCT116 cells. Invest New Drugs 2011;29:1276-83
    • (2011) Invest New Drugs , vol.29 , pp. 1276-1283
    • Lu, J.J.1    Chen, S.M.2    Zhang, X.W.3
  • 38
    • 77952266182 scopus 로고    scopus 로고
    • Dihydroartemisinin accelerates c-MYC oncoprotein degradation and induces apoptosis in c-MYC-overexpressing tumor cells
    • Lu JJ, Meng LH, Shankavaram UT, et al. Dihydroartemisinin accelerates c-MYC oncoprotein degradation and induces apoptosis in c-MYC-overexpressing tumor cells. Biochem Pharmacol 2010;80:22-30
    • (2010) Biochem Pharmacol , vol.80 , pp. 22-30
    • Lu, J.J.1    Meng, L.H.2    Shankavaram, U.T.3
  • 39
    • 77952420656 scopus 로고    scopus 로고
    • Dihydroartemisinin inactivates NF-kappaB and potentiates the anti-tumor effect of gemcitabine on pancreatic cancer both in vitro and in vivo
    • Wang SJ, Gao Y, Chen H, et al. Dihydroartemisinin inactivates NF-kappaB and potentiates the anti-tumor effect of gemcitabine on pancreatic cancer both in vitro and in vivo. Cancer Lett 2010;293:99-108
    • (2010) Cancer Lett , vol.293 , pp. 99-108
    • Wang, S.J.1    Gao, Y.2    Chen, H.3
  • 40
    • 73449131476 scopus 로고    scopus 로고
    • Effect of artesunate on inhibiting proliferation and inducing apoptosis of SP2/0 myeloma cells through affecting NFkappaB p65
    • Li S, Xue F, Cheng Z, et al. Effect of artesunate on inhibiting proliferation and inducing apoptosis of SP2/0 myeloma cells through affecting NFkappaB p65. Int J Hematol 2009;90:513-21
    • (2009) Int J Hematol , vol.90 , pp. 513-521
    • Li, S.1    Xue, F.2    Cheng, Z.3
  • 41
    • 0035315838 scopus 로고    scopus 로고
    • The anti-malarial artesunate is also active against cancer
    • Efferth T, Dunstan H, Sauerbrey A, et al. The anti-malarial artesunate is also active against cancer. Int J Oncol 2001;18:767-73
    • (2001) Int J Oncol , vol.18 , pp. 767-773
    • Efferth, T.1    Dunstan, H.2    Sauerbrey, A.3
  • 42
    • 0042844638 scopus 로고    scopus 로고
    • Molecular modes of action of artesunate in tumor cell lines
    • Efferth T, Sauerbrey A, Olbrich A, et al. Molecular modes of action of artesunate in tumor cell lines. Mol Pharmacol 2003;64:382-94
    • (2003) Mol Pharmacol , vol.64 , pp. 382-394
    • Efferth, T.1    Sauerbrey, A.2    Olbrich, A.3
  • 43
    • 72049100401 scopus 로고    scopus 로고
    • Anti-cancer effects of artesunate in a panel of chemoresistant neuroblastoma cell lines
    • Michaelis M, Kleinschmidt MC, Barth S, et al. Anti-cancer effects of artesunate in a panel of chemoresistant neuroblastoma cell lines. Biochem Pharmacol 2010;79:130-6
    • (2010) Biochem Pharmacol , vol.79 , pp. 130-136
    • Michaelis, M.1    Kleinschmidt, M.C.2    Barth, S.3
  • 44
    • 1842856050 scopus 로고    scopus 로고
    • Modulation of multidrug resistance by artemisinin, artesunate and dihydroartemisinin in K562/adr and GLC4/adr resistant cell lines
    • Reungpatthanaphong P, Mankhetkorn S. Modulation of multidrug resistance by artemisinin, artesunate and dihydroartemisinin in K562/adr and GLC4/adr resistant cell lines. Biol Pharm Bull 2002;25:1555-61
    • (2002) Biol Pharm Bull , vol.25 , pp. 1555-1561
    • Reungpatthanaphong, P.1    Mankhetkorn, S.2
  • 45
    • 83555163925 scopus 로고    scopus 로고
    • Characterization of dihydroartemisinin-resistant colon carcinoma HCT116R cell line
    • Lu JJ, Chen SM, Ding J, et al. Characterization of dihydroartemisinin- resistant colon carcinoma HCT116/R cell line. Mol Cell Biochem 2012;360:329-37
    • (2012) Mol Cell Biochem , vol.360 , pp. 329-337
    • Lu, J.J.1    Chen, S.M.2    Ding, J.3
  • 46
    • 77649189381 scopus 로고    scopus 로고
    • Artesunate induces oncosis-like cell death in vitro and has antitumor activity against pancreatic cancer xenografts in vivo
    • Du JH, Zhang HD, Ma ZJ, et al. Artesunate induces oncosis-like cell death in vitro and has antitumor activity against pancreatic cancer xenografts in vivo. Cancer Chemother Pharmacol 2010;65:895-902
    • (2010) Cancer Chemother Pharmacol , vol.65 , pp. 895-902
    • Du, J.H.1    Zhang, H.D.2    Ma, Z.J.3
  • 47
    • 68149173166 scopus 로고    scopus 로고
    • Dihydroartemisinin induces apoptosis and sensitizes human ovarian cancer cells to carboplatin therapy
    • Chen T, Li M, Zhang R, et al. Dihydroartemisinin induces apoptosis and sensitizes human ovarian cancer cells to carboplatin therapy. J Cell Mol Med 2009;13:1358-70
    • (2009) J Cell Mol Med , vol.13 , pp. 1358-1370
    • Chen, T.1    Li, M.2    Zhang, R.3
  • 48
    • 70449562761 scopus 로고    scopus 로고
    • Heme mediates cytotoxicity from artemisinin and serves as a general anti-proliferation target
    • Zhang S, Gerhard GS. Heme mediates cytotoxicity from artemisinin and serves as a general anti-proliferation target. PLoS One 2009;4:e7472
    • (2009) PLoS One , vol.4
    • Zhang, S.1    Gerhard, G.S.2
  • 49
    • 4444383530 scopus 로고    scopus 로고
    • Enhancement of cytotoxicity of artemisinins toward cancer cells by ferrous iron
    • Efferth T, Benakis A, Romero MR, et al. Enhancement of cytotoxicity of artemisinins toward cancer cells by ferrous iron. Free Radic Biol Med 2004;37:998-1009
    • (2004) Free Radic Biol Med , vol.37 , pp. 998-1009
    • Efferth, T.1    Benakis, A.2    Romero, M.R.3
  • 50
    • 77950958513 scopus 로고    scopus 로고
    • Factors determining sensitivity or resistance of tumor cell lines towards artesunate
    • Sertel S, Eichhorn T, Sieber S, et al. Factors determining sensitivity or resistance of tumor cell lines towards artesunate. Chem Biol Interact 2010;185:42-52
    • (2010) Chem Biol Interact , vol.185 , pp. 42-52
    • Sertel, S.1    Eichhorn, T.2    Sieber, S.3
  • 51
    • 59049096024 scopus 로고    scopus 로고
    • Artemisinin blocks prostate cancer growth and cell cycle progression by disrupting Sp1 interactions with the cyclin-dependent kinase-4 (CDK4) promoter and inhibiting CDK4 gene expression
    • Willoughby JA Sr, Sundar SN, Cheung M, et al. Artemisinin blocks prostate cancer growth and cell cycle progression by disrupting Sp1 interactions with the cyclin-dependent kinase-4 (CDK4) promoter and inhibiting CDK4 gene expression. J Biol Chem 2009;284:2203-13
    • (2009) J Biol Chem , vol.284 , pp. 2203-2213
    • Willoughby, J.A.S.R.1    Sundar, S.N.2    Cheung, M.3
  • 52
    • 77951651771 scopus 로고    scopus 로고
    • Growth inhibitory effects of dihydroartemisinin on pancreatic cancer cells: Involvement of cell cycle arrest and inactivation of nuclear factor-kappaB
    • Chen H, Sun B, Wang S, et al. Growth inhibitory effects of dihydroartemisinin on pancreatic cancer cells: involvement of cell cycle arrest and inactivation of nuclear factor-kappaB. J Cancer Res Clin Oncol 2010;136:897-903
    • (2010) J Cancer Res Clin Oncol , vol.136 , pp. 897-903
    • Chen, H.1    Sun, B.2    Wang, S.3
  • 53
    • 38049022026 scopus 로고    scopus 로고
    • The role of calcium P38 MAPK in dihydroartemisinin-induced apoptosis of lung cancer PC-14 cells
    • Mu D, Zhang W, Chu D, et al. The role of calcium, P38 MAPK in dihydroartemisinin-induced apoptosis of lung cancer PC-14 cells. Cancer Chemother Pharmacol 2008;61:639-45
    • (2008) Cancer Chemother Pharmacol , vol.61 , pp. 639-645
    • Mu, D.1    Zhang, W.2    Chu, D.3
  • 54
    • 77956591096 scopus 로고    scopus 로고
    • Dihydroartemisinin induces apoptosis by a bak-dependent intrinsic pathway
    • Handrick R, Ontikatze T, Bauer KD, et al. Dihydroartemisinin induces apoptosis by a Bak-dependent intrinsic pathway. Mol Cancer Ther 2010;9:2497-510
    • (2010) Mol Cancer Ther , vol.9 , pp. 2497-2510
    • Handrick, R.1    Ontikatze, T.2    Bauer, K.D.3
  • 55
    • 77955641219 scopus 로고    scopus 로고
    • First evidence that the antimalarial drug artesunate inhibits invasion and in vivo metastasis in lung cancer by targeting essential extracellular proteases
    • Rasheed SA, Efferth T, Asangani IA, et al. First evidence that the antimalarial drug artesunate inhibits invasion and in vivo metastasis in lung cancer by targeting essential extracellular proteases. Int J Cancer 2010;127:1475-85
    • (2010) Int J Cancer , vol.127 , pp. 1475-1485
    • Rasheed, S.A.1    Efferth, T.2    Asangani, I.A.3
  • 56
    • 77951298166 scopus 로고    scopus 로고
    • Suppression of PMA-induced tumor cell invasion by dihydroartemisinin via inhibition of PKCalpha/Raf/MAPKs and NF-kappaB/AP-1-dependent mechanisms
    • Hwang YP, Yun HJ, Kim HG, et al. Suppression of PMA-induced tumor cell invasion by dihydroartemisinin via inhibition of PKCalpha/Raf/MAPKs and NF-kappaB/AP-1-dependent mechanisms. Biochem Pharmacol 2010;79:1714-26
    • (2010) Biochem Pharmacol , vol.79 , pp. 1714-1726
    • Hwang, Y.P.1    Yun, H.J.2    Kim, H.G.3
  • 57
    • 2142650659 scopus 로고    scopus 로고
    • Antimalarial dihydroartemisinin also inhibits angiogenesis
    • Chen HH, Zhou HJ, Wang WQ, et al. Antimalarial dihydroartemisinin also inhibits angiogenesis. Cancer Chemother Pharmacol 2004;53:423-32
    • (2004) Cancer Chemother Pharmacol , vol.53 , pp. 423-432
    • Chen, H.H.1    Zhou, H.J.2    Wang, W.Q.3
  • 58
    • 70349260586 scopus 로고    scopus 로고
    • Artemisinin reduces human melanoma cell migration by down-regulating alpha v beta 3 integrin and reducing metalloproteinase 2 production
    • Buommino E, Baroni A, Canozo N, et al. Artemisinin reduces human melanoma cell migration by down-regulating alpha V beta 3 integrin and reducing metalloproteinase 2 production. Invest New Drugs 2009;27:412-18
    • (2009) Invest New Drugs , vol.27 , pp. 412-418
    • Buommino, E.1    Baroni, A.2    Canozo, N.3
  • 59
    • 27844511101 scopus 로고    scopus 로고
    • Danshen: An overview of its chemistry, pharmacology, pharmacokinetics, and clinical use
    • Zhou L, Zuo Z, Chow MS. Danshen: an overview of its chemistry, pharmacology, pharmacokinetics, and clinical use. J Clin Pharmacol 2005;45:1345-59
    • (2005) J Clin Pharmacol , vol.45 , pp. 1345-1359
    • Zhou, L.1    Zuo, Z.2    Chow, M.S.3
  • 60
    • 0033621456 scopus 로고    scopus 로고
    • Tanshinone IIA, an ingredient of Salvia miltiorrhiza BUNGE, induces apoptosis in human leukemia cell lines through the activation of caspase-3
    • Sung HJ, Choi SM, Yoon Y, et al. Tanshinone IIA, an ingredient of Salvia miltiorrhiza BUNGE, induces apoptosis in human leukemia cell lines through the activation of caspase-3. Exp Mol Med 1999;31:174-8
    • (1999) Exp Mol Med , vol.31 , pp. 174-178
    • Sung, H.J.1    Choi, S.M.2    Yoon, Y.3
  • 61
    • 33845418736 scopus 로고    scopus 로고
    • Induction of apoptosis and inhibition of cell adhesive and invasive effects by tanshinone IIA in acute promyelocytic leukemia cells in vitro
    • Liu JJ, Lin DJ, Liu PQ, et al. Induction of apoptosis and inhibition of cell adhesive and invasive effects by tanshinone IIA in acute promyelocytic leukemia cells in vitro. J Biomed Sci 2006;13:813-23
    • (2006) J Biomed Sci , vol.13 , pp. 813-823
    • Liu, J.J.1    Lin, D.J.2    Liu, P.Q.3
  • 62
    • 23244463302 scopus 로고    scopus 로고
    • Potential anticancer activity of tanshinone IIA against human breast cancer
    • Wang X, Wei Y, Yuan S, et al. Potential anticancer activity of tanshinone IIA against human breast cancer. Int J Cancer 2005;116:799-807
    • (2005) Int J Cancer , vol.116 , pp. 799-807
    • Wang, X.1    Wei, Y.2    Yuan, S.3
  • 63
    • 53049099464 scopus 로고    scopus 로고
    • Tanshinone IIA inhibits human breast cancer cells through increased Bax to Bcl-xL ratios
    • Su CC, Lin YH. Tanshinone IIA inhibits human breast cancer cells through increased Bax to Bcl-xL ratios. Int J Mol Med 2008;22:357-61
    • (2008) Int J Mol Med , vol.22 , pp. 357-361
    • Su, C.C.1    Lin, Y.H.2
  • 64
    • 52949101728 scopus 로고    scopus 로고
    • Growth inhibition and apoptosis induction by tanshinone IIA in human colon adenocarcinoma cells
    • Su CC, Chen GW, Kang JC, et al. Growth inhibition and apoptosis induction by tanshinone IIA in human colon adenocarcinoma cells. Planta Med 2008;74:1357-62
    • (2008) Planta Med , vol.74 , pp. 1357-1362
    • Su, C.C.1    Chen, G.W.2    Kang, J.C.3
  • 65
    • 0026309074 scopus 로고
    • Cytotoxic activities of tanshinones against human carcinoma cell lines
    • Wu WL, Chang WL, Chen CF. Cytotoxic activities of tanshinones against human carcinoma cell lines. Am J Chin Med 1991;19:207-16
    • (1991) Am J Chin Med , vol.19 , pp. 207-216
    • Wu, W.L.1    Chang, W.L.2    Chen, C.F.3
  • 66
    • 3142604774 scopus 로고    scopus 로고
    • Growth inhibition and apoptosis induction in human hepatoma cells by tanshinone II A
    • Tang Z, Tang Y, Fu L. Growth inhibition and apoptosis induction in human hepatoma cells by tanshinone II A. J Huazhong Univ Sci Technolog Med Sci 2003;23:166-8; 172
    • (2003) J Huazhong Univ Sci Technolog Med Sci , vol.23 , Issue.166-168 , pp. 172
    • Tang, Z.1    Tang, Y.2    Fu, L.3
  • 67
    • 4043125279 scopus 로고    scopus 로고
    • Growth inhibition and apoptosis induction of tanshinone II-A on human hepatocellular carcinoma cells
    • Yuan SL, Wei YQ, Wang XJ, et al. Growth inhibition and apoptosis induction of tanshinone II-A on human hepatocellular carcinoma cells. World J Gastroenterol 2004;10:2024-8
    • (2004) World J Gastroenterol , vol.10 , pp. 2024-2028
    • Yuan, S.L.1    Wei, Y.Q.2    Wang, X.J.3
  • 68
    • 77952996906 scopus 로고    scopus 로고
    • Cytotoxic effects of tanshinones from Salvia miltiorrhiza on doxorubicin-resistant human liver cancer cells
    • Lee WY, Cheung CC, Liu KW, et al. Cytotoxic effects of tanshinones from Salvia miltiorrhiza on doxorubicin-resistant human liver cancer cells. J Nat Prod 2010;73:854-9
    • (2010) J Nat Prod , vol.73 , pp. 854-859
    • Lee, W.Y.1    Cheung, C.C.2    Liu, K.W.3
  • 69
    • 79551595325 scopus 로고    scopus 로고
    • Tanshinone IIA acts via p38 MAPK to induce apoptosis and the down-regulation of ERCC1 and lung-resistance protein in cisplatin-resistant ovarian cancer cells
    • Jiao JW, Wen F. Tanshinone IIA acts via p38 MAPK to induce apoptosis and the down-regulation of ERCC1 and lung-resistance protein in cisplatin-resistant ovarian cancer cells. Oncol Rep 2011;25:781-8
    • (2011) Oncol Rep , vol.25 , pp. 781-788
    • Jiao, J.W.1    Wen, F.2
  • 70
    • 65449179230 scopus 로고    scopus 로고
    • Tanshinone IIA down-regulates the protein expression of ErbB-2 and up-regulates TNF-alpha in colon cancer cells in vitro and in vivo
    • Su CC, Lin YH. Tanshinone IIA down-regulates the protein expression of ErbB-2 and up-regulates TNF-alpha in colon cancer cells in vitro and in vivo. Int J Mol Med 2008;22:847-51
    • (2008) Int J Mol Med , vol.22 , pp. 847-851
    • Su, C.C.1    Lin, Y.H.2
  • 71
    • 84856219184 scopus 로고    scopus 로고
    • Tanshinone IIA activates calcium-dependent apoptosis signaling pathway in human hepatoma cells
    • Dai ZK, Qin JK, Huang JE, et al. Tanshinone IIA activates calcium-dependent apoptosis signaling pathway in human hepatoma cells. J Nat Med 2011;66(1):192-201
    • (2011) J Nat Med , vol.66 , Issue.1 , pp. 192-201
    • Dai, Z.K.1    Qin, J.K.2    Huang, J.E.3
  • 72
    • 78249235878 scopus 로고    scopus 로고
    • Tanshinone IIA induces mitochondria dependent apoptosis in prostate cancer cells in association with an inhibition of phosphoinositide 3-kinase/AKT pathway
    • Won SH, Lee HJ, Jeong SJ, et al. Tanshinone IIA induces mitochondria dependent apoptosis in prostate cancer cells in association with an inhibition of phosphoinositide 3-kinase/AKT pathway. Biol Pharm Bull 2010;33:1828-34
    • (2010) Biol Pharm Bull , vol.33 , pp. 1828-1834
    • Won, S.H.1    Lee, H.J.2    Jeong, S.J.3
  • 73
    • 73649126883 scopus 로고    scopus 로고
    • Tanshinone IIA induces apoptosis in human lung cancer A549 cells through the induction of reactive oxygen species and decreasing the mitochondrial membrane potential
    • Chiu TL, Su CC. Tanshinone IIA induces apoptosis in human lung cancer A549 cells through the induction of reactive oxygen species and decreasing the mitochondrial membrane potential. Int J Mol Med 2010;25:231-6
    • (2010) Int J Mol Med , vol.25 , pp. 231-236
    • Chiu, T.L.1    Su, C.C.2
  • 74
    • 77956671599 scopus 로고    scopus 로고
    • Tanshinone IIA may inhibit the growth of small cell lung cancer H146 cells by up-regulating the Bax/Bcl-2 ratio and decreasing mitochondrial membrane potential
    • Cheng CY, Su CC. Tanshinone IIA may inhibit the growth of small cell lung cancer H146 cells by up-regulating the Bax/Bcl-2 ratio and decreasing mitochondrial membrane potential. Mol Med Report 2010;3:645-50
    • (2010) Mol Med Report , vol.3 , pp. 645-650
    • Cheng, C.Y.1    Su, C.C.2
  • 75
    • 78549263680 scopus 로고    scopus 로고
    • C/EBPbeta and CHOP participate in tanshinone IIA-induced differentiation and apoptosis of acute promyelocytic leukemia cells in vitro
    • Zhang K, Li J, Meng W, et al. C/EBPbeta and CHOP participate in tanshinone IIA-induced differentiation and apoptosis of acute promyelocytic leukemia cells in vitro. Int J Hematol 2010;92:571-8
    • (2010) Int J Hematol , vol.92 , pp. 571-578
    • Zhang, K.1    Li, J.2    Meng, W.3
  • 76
    • 33846965992 scopus 로고    scopus 로고
    • Growth inhibition and induction of apoptosis and differentiation of tanshinone IIA in human glioma cells
    • Wang J, Wang X, Jiang S, et al. Growth inhibition and induction of apoptosis and differentiation of tanshinone IIA in human glioma cells. J Neurooncol 2007;82:11-21
    • (2007) J Neurooncol , vol.82 , pp. 11-21
    • Wang, J.1    Wang, X.2    Jiang, S.3
  • 77
    • 76349098019 scopus 로고    scopus 로고
    • Tanshinone II-A inhibits invasion and metastasis of human hepatocellular carcinoma cells in vitro and in vivo
    • Yuxian X, Feng T, Ren L, et al. Tanshinone II-A inhibits invasion and metastasis of human hepatocellular carcinoma cells in vitro and in vivo. Tumori 2009;95:789-95
    • (2009) Tumori , vol.95 , pp. 789-795
    • Yuxian, X.1    Feng, T.2    Ren, L.3
  • 78
    • 70449339753 scopus 로고    scopus 로고
    • Inhibitory effects of tanshinone II-A on invasion and metastasis of human colon carcinoma cells
    • Shan YF, Shen X, Xie YK, et al. Inhibitory effects of tanshinone II-A on invasion and metastasis of human colon carcinoma cells. Acta Pharmacol Sin 2009;30:1537-42
    • (2009) Acta Pharmacol Sin , vol.30 , pp. 1537-1542
    • Shan, Y.F.1    Shen, X.2    Xie, Y.K.3
  • 79
    • 80051695345 scopus 로고    scopus 로고
    • Anti-angiogenic effect of Tanshinone IIA involves inhibition of matrix invasion and modification of MMP-2/TIMP-2 secretion in vascular endothelial cells
    • Tsai MY, Yang RC, Wu HT, et al. Anti-angiogenic effect of Tanshinone IIA involves inhibition of matrix invasion and modification of MMP-2/TIMP-2 secretion in vascular endothelial cells. Cancer Lett 2011;310:198-206
    • (2011) Cancer Lett , vol.310 , pp. 198-206
    • Tsai, M.Y.1    Yang, R.C.2    Wu, H.T.3
  • 80
    • 56249087235 scopus 로고    scopus 로고
    • Anticancer effects of tanshinone i in human non-small cell lung cancer
    • Lee CY, Sher HF, Chen HW, et al. Anticancer effects of tanshinone I in human non-small cell lung cancer. Mol Cancer Ther 2008;7:3527-38
    • (2008) Mol Cancer Ther , vol.7 , pp. 3527-3538
    • Lee, C.Y.1    Sher, H.F.2    Chen, H.W.3
  • 81
    • 59149096511 scopus 로고    scopus 로고
    • Growth inhibition and apoptosis induction by tanshinone i in human colon cancer Colo 205 cells
    • Su CC, Chen GW, Lin JG. Growth inhibition and apoptosis induction by tanshinone I in human colon cancer Colo 205 cells. Int J Mol Med 2008;22:613-18
    • (2008) Int J Mol Med , vol.22 , pp. 613-618
    • Su, C.C.1    Chen, G.W.2    Lin, J.G.3
  • 82
    • 54849187400 scopus 로고    scopus 로고
    • Tanshinone i effectively induces apoptosis in estrogen receptor-positive (MCF-7) and estrogen receptor-negative (MDA-MB-231) breast cancer cells
    • Nizamutdinova IT, Lee GW, Son KH, et al. Tanshinone I effectively induces apoptosis in estrogen receptor-positive (MCF-7) and estrogen receptor-negative (MDA-MB-231) breast cancer cells. Int J Oncol 2008;33:485-91
    • (2008) Int J Oncol , vol.33 , pp. 485-491
    • Nizamutdinova, I.T.1    Lee, G.W.2    Son, K.H.3
  • 83
    • 53349127298 scopus 로고    scopus 로고
    • Tanshinone i suppresses growth and invasion of human breast cancer cells, MDA-MB-231, through regulation of adhesion molecules
    • Nizamutdinova IT, Lee GW, Lee JS, et al. Tanshinone I suppresses growth and invasion of human breast cancer cells, MDA-MB-231, through regulation of adhesion molecules. Carcinogenesis 2008;29:1885-92
    • (2008) Carcinogenesis , vol.29 , pp. 1885-1892
    • Nizamutdinova, I.T.1    Lee, G.W.2    Lee, J.S.3
  • 84
    • 77956975737 scopus 로고    scopus 로고
    • Inactivation of PI3k/Akt signaling pathway and activation of caspase-3 are involved in tanshinone I-induced apoptosis in myeloid leukemia cells in vitro
    • Liu JJ, Liu WD, Yang HZ, et al. Inactivation of PI3k/Akt signaling pathway and activation of caspase-3 are involved in tanshinone I-induced apoptosis in myeloid leukemia cells in vitro. Ann Hematol 2010;89:1089-97
    • (2010) Ann Hematol , vol.89 , pp. 1089-1097
    • Liu, J.J.1    Liu, W.D.2    Yang, H.Z.3
  • 85
    • 70349301394 scopus 로고    scopus 로고
    • Reactive oxygen species-mediated kinase activation by dihydrotanshinone in tanshinones-induced apoptosis in HepG2 cells
    • Lee WY, Liu KW, Yeung JH. Reactive oxygen species-mediated kinase activation by dihydrotanshinone in tanshinones-induced apoptosis in HepG2 cells. Cancer Lett 2009;285:46-57
    • (2009) Cancer Lett , vol.285 , pp. 46-57
    • Lee, W.Y.1    Liu, K.W.2    Yeung, J.H.3
  • 86
    • 77956945287 scopus 로고    scopus 로고
    • Cryptotanshinone sensitizes DU145 prostate cancer cells to Fas (APO1/CD95)-mediated apoptosis through Bcl-2 and MAPK regulation
    • Park IJ, Kim MJ, Park OJ, et al. Cryptotanshinone sensitizes DU145 prostate cancer cells to Fas (APO1/CD95)-mediated apoptosis through Bcl-2 and MAPK regulation. Cancer Lett 2010;298:88-98
    • (2010) Cancer Lett , vol.298 , pp. 88-98
    • Park, I.J.1    Kim, M.J.2    Park, O.J.3
  • 87
    • 79952106497 scopus 로고    scopus 로고
    • Triptolide and its expanding multiple pharmacological functions
    • Liu Q. Triptolide and its expanding multiple pharmacological functions. Int Immunopharmacol 2011;11:377-83
    • (2011) Int Immunopharmacol , vol.11 , pp. 377-383
    • Liu, Q.1
  • 88
    • 0033532177 scopus 로고    scopus 로고
    • Immunosuppressant PG490 (triptolide) inhibits T-cell interleukin-2 expression at the level of purine-box/nuclear factor of activated T-cells and NF-kappaB transcriptional activation
    • Qiu D, Zhao G, Aoki Y, et al. Immunosuppressant PG490 (triptolide) inhibits T-cell interleukin-2 expression at the level of purine-box/nuclear factor of activated T-cells and NF-kappaB transcriptional activation. J Biol Chem 1999;274:13443-50
    • (1999) J Biol Chem , vol.274 , pp. 13443-13450
    • Qiu, D.1    Zhao, G.2    Aoki, Y.3
  • 89
    • 33646900838 scopus 로고    scopus 로고
    • Triptolide, an inhibitor of the human heat shock response that enhances stress-induced cell death
    • Westerheide SD, Kawahara TL, Orton K, et al. Triptolide, an inhibitor of the human heat shock response that enhances stress-induced cell death. J Biol Chem 2006;281:9616-22
    • (2006) J Biol Chem , vol.281 , pp. 9616-9622
    • Westerheide, S.D.1    Kawahara, T.L.2    Orton, K.3
  • 90
    • 0035910420 scopus 로고    scopus 로고
    • Triptolide and chemotherapy cooperate in tumor cell apoptosis. A role for the p53 pathway
    • Chang WT, Kang JJ, Lee KY, et al. Triptolide and chemotherapy cooperate in tumor cell apoptosis. A role for the p53 pathway. J Biol Chem 2001;276:2221-7
    • (2001) J Biol Chem , vol.276 , pp. 2221-2227
    • Chang, W.T.1    Kang, J.J.2    Lee, K.Y.3
  • 91
    • 34547203981 scopus 로고    scopus 로고
    • Triptolide binds covalently to a 90 kDa nuclear protein. Role of epoxides in binding and activity
    • McCallum C, Kwon S, Leavitt P, et al. Triptolide binds covalently to a 90 kDa nuclear protein. Role of epoxides in binding and activity. Immunobiology 2007;212:549-56
    • (2007) Immunobiology , vol.212 , pp. 549-556
    • McCallum, C.1    Kwon, S.2    Leavitt, P.3
  • 92
    • 70350241487 scopus 로고    scopus 로고
    • Triptolide is an inhibitor of RNA polymerase i and II-dependent transcription leading predominantly to down-regulation of short-lived mRNA
    • Vispe S, DeVries L, Creancier L, et al. Triptolide is an inhibitor of RNA polymerase I and II-dependent transcription leading predominantly to down-regulation of short-lived mRNA. Mol Cancer Ther 2009;8:2780-90
    • (2009) Mol Cancer Ther , vol.8 , pp. 2780-2790
    • Vispe, S.1    Devries, L.2    Creancier, L.3
  • 93
    • 80052726052 scopus 로고    scopus 로고
    • Triptolide (TPL) Inhibits Global Transcription by Inducing Proteasome-Dependent Degradation of RNA Polymerase II (Pol II)
    • Wang Y, Lu JJ, He L, et al. Triptolide (TPL) Inhibits Global Transcription by Inducing Proteasome-Dependent Degradation of RNA Polymerase II (Pol II). PLoS One 2011;6:e23993
    • (2011) PLoS One , vol.6
    • Wang, Y.1    Lu, J.J.2    He, L.3
  • 94
    • 34248350004 scopus 로고    scopus 로고
    • Triptolide is a traditional Chinese medicine-derived inhibitor of polycystic kidney disease
    • Leuenroth SJ, Okuhara D, Shotwell JD, et al. Triptolide is a traditional Chinese medicine-derived inhibitor of polycystic kidney disease. Proc Natl Acad Sci USA 2007;104:4389-94
    • (2007) Proc Natl Acad Sci USA , vol.104 , pp. 4389-4394
    • Leuenroth, S.J.1    Okuhara, D.2    Shotwell, J.D.3
  • 95
    • 79951851704 scopus 로고    scopus 로고
    • XPB, a subunit of TFIIH, is a target of the natural product triptolide
    • Titov DV, Gilman B, He QL, et al. XPB, a subunit of TFIIH, is a target of the natural product triptolide. Nat Chem Biol 2011;7:182-8
    • (2011) Nat Chem Biol , vol.7 , pp. 182-188
    • Titov, D.V.1    Gilman, B.2    He, Q.L.3
  • 96
    • 77957674310 scopus 로고    scopus 로고
    • Increased accumulation of hypoxia-inducible factor-1alpha with reduced transcriptional activity mediates the antitumor effect of triptolide
    • Zhou ZL, Luo ZG, Yu B, et al. Increased accumulation of hypoxia-inducible factor-1alpha with reduced transcriptional activity mediates the antitumor effect of triptolide. Mol Cancer 2010;9:268
    • (2010) Mol Cancer , vol.9 , pp. 268
    • Zhou, Z.L.1    Luo, Z.G.2    Yu, B.3
  • 97
    • 77954676306 scopus 로고    scopus 로고
    • Pseudolaric acids: Isolation bioactivity and synthetic studies
    • Chiu P, Leung LT, Ko BC. Pseudolaric acids: isolation, bioactivity and synthetic studies. Nat Prod Rep 2010;27:1066-83
    • (2010) Nat Prod Rep , vol.27 , pp. 1066-1083
    • Chiu, P.1    Leung, L.T.2    Ko, B.C.3
  • 98
    • 0025011152 scopus 로고
    • The cytotoxic principles of Pseudolarix kaempferi: Pseudolaric acid-A and-B and related derivatives
    • Pan DJ, Li ZL, Hu CQ, et al. The cytotoxic principles of Pseudolarix kaempferi: pseudolaric acid-A and-B and related derivatives. Planta Med 1990;56:383-5
    • (1990) Planta Med , vol.56 , pp. 383-385
    • Pan, D.J.1    Li, Z.L.2    Hu, C.Q.3
  • 99
    • 23844557173 scopus 로고    scopus 로고
    • Pseudolaric acid B, a novel microtubule-destabilizing agent that circumvents multidrug resistance phenotype and exhibits antitumor activity in vivo
    • Wong VK, Chiu P, Chung SS, et al. Pseudolaric acid B, a novel microtubule-destabilizing agent that circumvents multidrug resistance phenotype and exhibits antitumor activity in vivo. Clin Cancer Res 2005;11:6002-11
    • (2005) Clin Cancer Res , vol.11 , pp. 6002-6011
    • Wong, V.K.1    Chiu, P.2    Chung, S.S.3
  • 100
    • 1942438028 scopus 로고    scopus 로고
    • Microtubules as a target for anticancer drugs
    • Jordan MA, Wilson L. Microtubules as a target for anticancer drugs. Nat Rev Cancer 2004;4:253-65
    • (2004) Nat Rev Cancer , vol.4 , pp. 253-265
    • Jordan, M.A.1    Wilson, L.2
  • 101
    • 82555170343 scopus 로고    scopus 로고
    • Vinflunine: A new vision that may translate into antiangiogenic and antimetastatic activity
    • Anton Aparicio LM, Pulido EG, Gallego GA. Vinflunine: a new vision that may translate into antiangiogenic and antimetastatic activity. Anticancer Drugs 2011;23(1):1-11
    • (2011) Anticancer Drugs , vol.23 , pp. 11-11
    • Anton Aparicio, L.M.1    Pulido, E.G.2    Gallego, G.A.3
  • 102
    • 4544375253 scopus 로고    scopus 로고
    • Pseudolarix acid B inhibits angiogenesis by antagonizing the vascular endothelial growth factor-mediated anti-apoptotic effect
    • Tan WF, Zhang XW, Li MH, et al. Pseudolarix acid B inhibits angiogenesis by antagonizing the vascular endothelial growth factor-mediated anti-apoptotic effect. Eur J Pharmacol 2004;499:219-28
    • (2004) Eur J Pharmacol , vol.499 , pp. 219-228
    • Tan, W.F.1    Zhang, X.W.2    Li, M.H.3
  • 103
    • 11144220863 scopus 로고    scopus 로고
    • Pseudolaric acid B inhibits angiogenesis and reduces hypoxia-inducible factor 1alpha by promoting proteasome-mediated degradation
    • Li MH, Miao ZH, Tan WF, et al. Pseudolaric acid B inhibits angiogenesis and reduces hypoxia-inducible factor 1alpha by promoting proteasome-mediated degradation. Clin Cancer Res 2004;10:8266-74
    • (2004) Clin Cancer Res , vol.10 , pp. 8266-8274
    • Li, M.H.1    Zh, M.2    Tan, W.F.3
  • 104
    • 51349130575 scopus 로고    scopus 로고
    • Protein tyrosine kinase, JNK, and ERK involvement in pseudolaric acid B-induced apoptosis of human breast cancer MCF-7 cells
    • Yu JH, Wang HJ, Li XR, et al. Protein tyrosine kinase, JNK, and ERK involvement in pseudolaric acid B-induced apoptosis of human breast cancer MCF-7 cells. Acta Pharmacol Sin 2008;29:1069-76
    • (2008) Acta Pharmacol Sin , vol.29 , pp. 1069-1076
    • Yu, J.H.1    Wang, H.J.2    Li, X.R.3
  • 105
    • 47749149209 scopus 로고    scopus 로고
    • Bcl-2 family proteins were involved in pseudolaric acid B-induced autophagy in murine fibrosarcoma L929 cells
    • Yu J, Li X, Tashiro S, et al. Bcl-2 family proteins were involved in pseudolaric acid B-induced autophagy in murine fibrosarcoma L929 cells. J Pharmacol Sci 2008;107:295-302
    • (2008) J Pharmacol Sci , vol.107 , pp. 295-302
    • Yu, J.1    Li, X.2    Tashiro, S.3
  • 106
    • 84857381831 scopus 로고    scopus 로고
    • Andrographolide and its analogues: Versatile bioactive molecules for combating inflammation and cancer
    • Lim CW, Chan TK, Ng DS, et al. Andrographolide and its analogues: versatile bioactive molecules for combating inflammation and cancer. Clin Exp Pharmacol Physiol 2011;39(3):300-10
    • (2011) Clin Exp Pharmacol Physiol , vol.39 , Issue.3 , pp. 300-310
    • Lim, C.W.1    Chan, T.K.2    Ng, D.S.3
  • 107
    • 1242329231 scopus 로고    scopus 로고
    • Antihyperglycemic effect of andrographolide in streptozotocin-induced diabetic rats
    • Yu BC, Hung CR, Chen WC, et al. Antihyperglycemic effect of andrographolide in streptozotocin-induced diabetic rats. Planta Med 2003;69:1075-9
    • (2003) Planta Med , vol.69 , pp. 1075-1079
    • Yu, B.C.1    Hung, C.R.2    Chen, W.C.3
  • 108
    • 4644241127 scopus 로고    scopus 로고
    • Andrographolide attenuates inflammation by inhibition of NF-kappa B activation through covalent modification of reduced cysteine 62 of p50
    • Xia YF, Ye BQ, Li YD, et al. Andrographolide attenuates inflammation by inhibition of NF-kappa B activation through covalent modification of reduced cysteine 62 of p50. J Immunol 2004;173:4207-17
    • (2004) J Immunol , vol.173 , pp. 4207-4217
    • Xia, Y.F.1    Ye, B.Q.2    Li, Y.D.3
  • 109
    • 36248995912 scopus 로고    scopus 로고
    • Andrographolide inhibits NF-kappaBeta activation and attenuates neointimal hyperplasia in arterial restenosis
    • Wang YJ, Wang JT, Fan QX, et al. Andrographolide inhibits NF-kappaBeta activation and attenuates neointimal hyperplasia in arterial restenosis. Cell Res 2007;17:933-41
    • (2007) Cell Res , vol.17 , pp. 933-941
    • Wang, Y.J.1    Wang, J.T.2    Fan, Q.X.3
  • 110
    • 80052811947 scopus 로고    scopus 로고
    • Andrographolide inhibits oral squamous cell carcinogenesis through NF-kappaB inactivation
    • Wang LJ, Zhou X, Wang W, et al. Andrographolide inhibits oral squamous cell carcinogenesis through NF-kappaB inactivation. J Dent Res 2011;90:1246-52
    • (2011) J Dent Res , vol.90 , pp. 1246-1252
    • Wang, L.J.1    Zhou, X.2    Wang, W.3
  • 111
    • 80053088605 scopus 로고    scopus 로고
    • Inhibition of tumor progression by naturally occurring terpenoids
    • Kuttan G, Pratheeshkumar P, Manu KA, et al. Inhibition of tumor progression by naturally occurring terpenoids. Pharm Biol 2011;49:995-1007
    • (2011) Pharm Biol , vol.49 , pp. 995-1007
    • Kuttan, G.1    Pratheeshkumar, P.2    Manu, K.A.3
  • 112
    • 18044387257 scopus 로고    scopus 로고
    • Oridonin, a diterpenoid purified from Rabdosia rubescens, inhibits the proliferation of cells from lymphoid malignancies in association with blockade of the NF-kappa B signal pathways
    • Ikezoe T, Yang Y, Bandobashi K, et al. Oridonin, a diterpenoid purified from Rabdosia rubescens, inhibits the proliferation of cells from lymphoid malignancies in association with blockade of the NF-kappa B signal pathways. Mol Cancer Ther 2005;4:578-86
    • (2005) Mol Cancer Ther , vol.4 , pp. 578-586
    • Ikezoe, T.1    Yang, Y.2    Bandobashi, K.3
  • 113
    • 79952822049 scopus 로고    scopus 로고
    • Downregulation of AP-1 gene expression is an initial event in the oridonin-mediated inhibition of colorectal cancer: Studies in vitro and in vivo
    • Jin H, Tan X, Liu X, et al. Downregulation of AP-1 gene expression is an initial event in the oridonin-mediated inhibition of colorectal cancer: studies in vitro and in vivo. J Gastroenterol Hepatol 2011;26:706-15
    • (2011) J Gastroenterol Hepatol , vol.26 , pp. 706-715
    • Jin, H.1    Tan, X.2    Liu, X.3
  • 114
    • 35648937054 scopus 로고    scopus 로고
    • Oridonin induces apoptosis via PI3K/Akt pathway in cervical carcinoma HeLa cell line
    • Hu HZ, Yang YB, Xu XD, et al. Oridonin induces apoptosis via PI3K/Akt pathway in cervical carcinoma HeLa cell line. Acta Pharmacol Sin 2007;28:1819-26
    • (2007) Acta Pharmacol Sin , vol.28 , pp. 1819-1826
    • Hu, H.Z.1    Yang, Y.B.2    Xu, X.D.3
  • 115
    • 69849112209 scopus 로고    scopus 로고
    • NF-kappab facilitates oridonin-induced apoptosis and autophagy in HT1080 cells through a p53-mediated pathway
    • Zhang Y, Wu Y, Wu D, et al. NF-kappab facilitates oridonin-induced apoptosis and autophagy in HT1080 cells through a p53-mediated pathway. Arch Biochem Biophys 2009;489:25-33
    • (2009) Arch Biochem Biophys , vol.489 , pp. 25-33
    • Zhang, Y.1    Wu, Y.2    Wu, D.3
  • 116
    • 65249128643 scopus 로고    scopus 로고
    • Molecular mechanisms of oridonin-induced apoptosis and autophagy in murine fibrosarcoma L929 cells
    • Cheng Y, Qiu F, Ikejima T. Molecular mechanisms of oridonin-induced apoptosis and autophagy in murine fibrosarcoma L929 cells. Autophagy 2009;5:430-1
    • (2009) Autophagy , vol.5 , pp. 430-431
    • Cheng, Y.1    Qiu, F.2    Ikejima, T.3
  • 117
    • 60349085032 scopus 로고    scopus 로고
    • Autophagy inhibits reactive oxygen species-mediated apoptosis via activating p38-nuclear factor-kappa B survival pathways in oridonin-treated murine fibrosarcoma L929 cells
    • Cheng Y, Qiu F, Ye YC, et al. Autophagy inhibits reactive oxygen species-mediated apoptosis via activating p38-nuclear factor-kappa B survival pathways in oridonin-treated murine fibrosarcoma L929 cells. FEBS J 2009;276:1291-306
    • (2009) FEBS J , vol.276 , pp. 1291-1306
    • Cheng, Y.1    Qiu, F.2    Ye, Y.C.3
  • 118
    • 80053923208 scopus 로고    scopus 로고
    • Reactive oxygen species contribute to oridonin-induced apoptosis and autophagy in human cervical carcinoma hela cells
    • Zhang YH, Wu YL, Tashiro S, et al. Reactive oxygen species contribute to oridonin-induced apoptosis and autophagy in human cervical carcinoma HeLa cells. Acta Pharmacol Sin 2011;32:1266-75
    • (2011) Acta Pharmacol Sin , vol.32 , pp. 1266-1275
    • Zhang, Y.H.1    Wu, Y.L.2    Tashiro, S.3
  • 119
    • 1542344495 scopus 로고    scopus 로고
    • Anti-inflammatory compounds of plant origin. Part II. modulation of pro-inflammatory cytokines, chemokines and adhesion molecules
    • Calixto JB, Campos MM, Otuki MF, et al. Anti-inflammatory compounds of plant origin. Part II. modulation of pro-inflammatory cytokines, chemokines and adhesion molecules. Planta Med 2004;70:93-103
    • (2004) Planta Med , vol.70 , pp. 93-103
    • Calixto, J.B.1    Campos, M.M.2    Otuki, M.F.3
  • 120
    • 0034752311 scopus 로고    scopus 로고
    • Celastrol, a potent antioxidant and anti-inflammatory drug, as a possible treatment for Alzheimer's disease
    • Allison AC, Cacabelos R, Lombardi VR, et al. Celastrol, a potent antioxidant and anti-inflammatory drug, as a possible treatment for Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry 2001;25:1341-57
    • (2001) Prog Neuropsychopharmacol Biol Psychiatry , vol.25 , pp. 1341-1357
    • Allison, A.C.1    Cacabelos, R.2    Lombardi, V.R.3
  • 121
    • 17444428634 scopus 로고    scopus 로고
    • Beneficial effect of tripterine on systemic lupus erythematosus induced by active chromatin in BALB/c mice
    • Li H, Zhang YY, Huang XY, et al. Beneficial effect of tripterine on systemic lupus erythematosus induced by active chromatin in BALB/c mice. Eur J Pharmacol 2005;512:231-7
    • (2005) Eur J Pharmacol , vol.512 , pp. 231-237
    • Li, H.1    Zhang, Y.Y.2    Huang, X.Y.3
  • 122
    • 33748854271 scopus 로고    scopus 로고
    • Tripterine inhibits the expression of adhesion molecules in activated endothelial cells
    • Zhang DH, Marconi A, Xu LM, et al. Tripterine inhibits the expression of adhesion molecules in activated endothelial cells. J Leukoc Biol 2006;80:309-19
    • (2006) J Leukoc Biol , vol.80 , pp. 309-319
    • Zhang, D.H.1    Marconi, A.2    Xu, L.M.3
  • 123
    • 4444238155 scopus 로고    scopus 로고
    • Celastrol inhibits pro-inflammatory cytokine secretion in Crohn's disease biopsies
    • Pinna GF, Fiorucci M, Reimund JM, et al. Celastrol inhibits pro-inflammatory cytokine secretion in Crohn's disease biopsies. Biochem Biophys Res Commun 2004;322:778-86
    • (2004) Biochem Biophys Res Commun , vol.322 , pp. 778-786
    • Pinna, G.F.1    Fiorucci, M.2    Reimund, J.M.3
  • 124
    • 77955771876 scopus 로고    scopus 로고
    • Celastrol attenuates hypertension-induced inflammation and oxidative stress in vascular smooth muscle cells via induction of heme oxygenase-1
    • Yu X, Tao W, Jiang F, et al. Celastrol attenuates hypertension-induced inflammation and oxidative stress in vascular smooth muscle cells via induction of heme oxygenase-1. Am J Hypertens 2010;23:895-903
    • (2010) Am J Hypertens , vol.23 , pp. 895-903
    • Yu, X.1    Tao, W.2    Jiang, F.3
  • 125
    • 77954758343 scopus 로고    scopus 로고
    • Celastrol suppresses IFN-gamma-induced ICAM-1 expression and subsequent monocyte adhesiveness via the induction of heme oxygenase-1 in the HaCaT cells
    • Seo WY, Ju SM, Song HY, et al. Celastrol suppresses IFN-gamma-induced ICAM-1 expression and subsequent monocyte adhesiveness via the induction of heme oxygenase-1 in the HaCaT cells. Biochem Biophys Res Commun 2010;398:140-5
    • (2010) Biochem Biophys Res Commun , vol.398 , pp. 140-145
    • Seo, W.Y.1    Ju, S.M.2    Song, H.Y.3
  • 126
    • 77950234271 scopus 로고    scopus 로고
    • Celastrol suppresses angiogenesis-mediated tumor growth through inhibition of AKT/mammalian target of rapamycin pathway
    • Pang X, Yi Z, Zhang J, et al. Celastrol suppresses angiogenesis-mediated tumor growth through inhibition of AKT/mammalian target of rapamycin pathway. Cancer Res 2010;70:1951-9
    • (2010) Cancer Res , vol.70 , pp. 1951-1959
    • Pang, X.1    Yi, Z.2    Zhang, J.3
  • 127
    • 33646406554 scopus 로고    scopus 로고
    • Celastrol a triterpene extracted from the Chinese "thunder of God Vine," is a potent proteasome inhibitor and suppresses human prostate cancer growth in nude mice
    • Yang H, Chen D, Cui QC, et al. Celastrol, a triterpene extracted from the Chinese "Thunder of God Vine," is a potent proteasome inhibitor and suppresses human prostate cancer growth in nude mice. Cancer Res 2006;66:4758-65
    • (2006) Cancer Res , vol.66 , pp. 4758-4765
    • Yang, H.1    Chen, D.2    Cui, Q.C.3
  • 128
    • 33947598693 scopus 로고    scopus 로고
    • Celastrol, a novel triterpene, potentiates TNF-induced apoptosis and suppresses invasion of tumor cells by inhibiting NF-kappaB-regulated gene products and TAK1-mediated NF-kappaB activation
    • Sethi G, Ahn KS, Pandey MK, et al. Celastrol, a novel triterpene, potentiates TNF-induced apoptosis and suppresses invasion of tumor cells by inhibiting NF-kappaB-regulated gene products and TAK1-mediated NF-kappaB activation. Blood 2007;109:2727-35
    • (2007) Blood , vol.109 , pp. 2727-2735
    • Sethi, G.1    Ahn, K.S.2    Pandey, M.K.3
  • 130
    • 0001113203 scopus 로고
    • Cucurbitacins: Specific insect attractants in Cucurbitaceae
    • Chambliss OL, Jones CM. Cucurbitacins: specific insect attractants in Cucurbitaceae. Science 1966;153:1392-3
    • (1966) Science , vol.153 , pp. 1392-1393
    • Chambliss, O.L.1    Jones, C.M.2
  • 131
    • 0028910214 scopus 로고
    • Cucurbitacins and their pharmacological effects
    • Miró M. Cucurbitacins and their pharmacological effects. Phytotherapy Research 1995;9:159-68
    • (1995) Phytotherapy Research , vol.9 , pp. 159-168
    • Miró, M.1
  • 132
    • 77951723076 scopus 로고    scopus 로고
    • Cucurbitacin: Ancient compound shedding new light on cancer treatment
    • Lee DH, Iwanski GB, Thoennissen NH. Cucurbitacin: ancient compound shedding new light on cancer treatment. Sci World J 2010;10:413-18
    • (2010) Sci World J , vol.10 , pp. 413-418
    • Lee, D.H.1    Iwanski, G.B.2    Thoennissen, N.H.3
  • 133
    • 77949356144 scopus 로고    scopus 로고
    • The induction of G2/M cell-cycle arrest and apoptosis by cucurbitacin e is associated with increased phosphorylation of eIF2alpha in leukemia cells
    • Li Y, Wang R, Ma E, et al. The induction of G2/M cell-cycle arrest and apoptosis by cucurbitacin E is associated with increased phosphorylation of eIF2alpha in leukemia cells. Anticancer Drugs 2010;21:389-400
    • (2010) Anticancer Drugs , vol.21 , pp. 389-400
    • Li, Y.1    Wang, R.2    Ma, E.3
  • 134
    • 77956532375 scopus 로고    scopus 로고
    • Cucurbitacin B inhibits growth, arrests the cell cycle, and potentiates antiproliferative efficacy of cisplatin in cutaneous squamous cell carcinoma cell lines
    • Chen W, Leiter A, Yin D, et al. Cucurbitacin B inhibits growth, arrests the cell cycle, and potentiates antiproliferative efficacy of cisplatin in cutaneous squamous cell carcinoma cell lines. Int J Oncol 2010;37:737-43
    • (2010) Int J Oncol , vol.37 , pp. 737-743
    • Chen, W.1    Leiter, A.2    Yin, D.3
  • 135
    • 43049175018 scopus 로고    scopus 로고
    • Cucurbitacin B induces differentiation, cell cycle arrest, and actin cytoskeletal alterations in myeloid leukemia cells
    • Haritunians T, Gueller S, Zhang L, et al. Cucurbitacin B induces differentiation, cell cycle arrest, and actin cytoskeletal alterations in myeloid leukemia cells. Leuk Res 2008;32:1366-73
    • (2008) Leuk Res , vol.32 , pp. 1366-1373
    • Haritunians, T.1    Gueller, S.2    Zhang, L.3
  • 136
    • 77953021503 scopus 로고    scopus 로고
    • Cucurbitacin B induces apoptosis and S phase cell cycle arrest in BEL-7402 human hepatocellular carcinoma cells and is effective via oral administration
    • Chan KT, Meng FY, Li Q, et al. Cucurbitacin B induces apoptosis and S phase cell cycle arrest in BEL-7402 human hepatocellular carcinoma cells and is effective via oral administration. Cancer Lett 2010;294:118-24
    • (2010) Cancer Lett , vol.294 , pp. 118-124
    • Chan, K.T.1    Meng, F.Y.2    Li, Q.3
  • 137
    • 79959622792 scopus 로고    scopus 로고
    • Cucurbitacin i inhibits tumorigenic ability and enhances radiochemosensitivity in nonsmall cell lung cancer-derived cd133-positive cells
    • Hsu HS, Huang PI, Chang YL, et al. Cucurbitacin I inhibits tumorigenic ability and enhances radiochemosensitivity in nonsmall cell lung cancer-derived CD133-positive cells. Cancer 2011;117:2970-85
    • (2011) Cancer , vol.117 , pp. 2970-2985
    • Hsu, H.S.1    Huang, P.I.2    Chang, Y.L.3
  • 138
    • 79958260109 scopus 로고    scopus 로고
    • Histone deacetylase inhibitor valproic acid sensitizes B16F10 melanoma cells to cucurbitacin B treatment
    • Ouyang D, Zhang Y, Xu L, et al. Histone deacetylase inhibitor valproic acid sensitizes B16F10 melanoma cells to cucurbitacin B treatment. Acta Biochim Biophys Sin (Shanghai) 2011;43:487-95
    • (2011) Acta Biochim Biophys Sin (Shanghai) , vol.43 , pp. 487-495
    • Ouyang, D.1    Zhang, Y.2    Xu, L.3
  • 139
    • 78649648154 scopus 로고    scopus 로고
    • Cucurbitacin i suppressed stem-like property and enhanced radiation-induced apoptosis in head and neck squamous carcinoma-derived CD44(+)ALDH1(+) cells
    • Chen YW, Chen KH, Huang PI, et al. Cucurbitacin I suppressed stem-like property and enhanced radiation-induced apoptosis in head and neck squamous carcinoma-derived CD44(+)ALDH1(+) cells. Mol Cancer Ther 2010;9:2879-92
    • (2010) Mol Cancer Ther , vol.9 , pp. 2879-2892
    • Chen, Y.W.1    Chen, K.H.2    Huang, P.I.3
  • 140
    • 73949160835 scopus 로고    scopus 로고
    • Cucurbitacin i elicits anoikis sensitization, inhibits cellular invasion and in vivo tumor formation ability of nasopharyngeal carcinoma cells
    • Lui VW, Yau DM, Wong EY, et al. Cucurbitacin I elicits anoikis sensitization, inhibits cellular invasion and in vivo tumor formation ability of nasopharyngeal carcinoma cells. Carcinogenesis 2009;30:2085-94
    • (2009) Carcinogenesis , vol.30 , pp. 2085-2094
    • Lui, V.W.1    Yau, D.M.2    Wong, E.Y.3
  • 141
    • 11244255857 scopus 로고    scopus 로고
    • Implication of STAT3 signaling in human colonic cancer cells during intestinal trefoil factor 3 (TFF3)-and vascular endothelial growth factor-mediated cellular invasion and tumor growth
    • Rivat C, Rodrigues S, Bruyneel E, et al. Implication of STAT3 signaling in human colonic cancer cells during intestinal trefoil factor 3 (TFF3)-and vascular endothelial growth factor-mediated cellular invasion and tumor growth. Cancer Res 2005;65:195-202
    • (2005) Cancer Res , vol.65 , pp. 195-202
    • Rivat, C.1    Rodrigues, S.2    Bruyneel, E.3
  • 142
    • 77956020768 scopus 로고    scopus 로고
    • STAT3alpha is oncogenic for endometrial carcinoma cells and mediates the oncogenic effects of autocrine human growth hormone
    • Tang JZ, Kong XJ, Banerjee A, et al. STAT3alpha is oncogenic for endometrial carcinoma cells and mediates the oncogenic effects of autocrine human growth hormone. Endocrinology 2010;151:4133-45
    • (2010) Endocrinology , vol.151 , pp. 4133-4145
    • Tang, J.Z.1    Kong, X.J.2    Banerjee, A.3
  • 143
    • 0013813823 scopus 로고
    • The combined effect of cucurbitacins and X-ray treatment on transplanted tumors in mice
    • Shohat B, Gitter S, Levie B, et al. The combined effect of cucurbitacins and X-ray treatment on transplanted tumors in mice. Cancer Res 1965;25:1828-35
    • (1965) Cancer Res , vol.25 , pp. 1828-1835
    • Shohat, B.1    Gitter, S.2    Levie, B.3
  • 144
    • 67650995911 scopus 로고    scopus 로고
    • Cucurbitacin B induces apoptosis by inhibition of the JAK/STAT pathway and potentiates antiproliferative effects of gemcitabine on pancreatic cancer cells
    • Thoennissen NH, Iwanski GB, Doan NB, et al. Cucurbitacin B induces apoptosis by inhibition of the JAK/STAT pathway and potentiates antiproliferative effects of gemcitabine on pancreatic cancer cells. Cancer Res 2009;69:5876-84
    • (2009) Cancer Res , vol.69 , pp. 5876-5884
    • Thoennissen, N.H.1    Iwanski, G.B.2    Doan, N.B.3
  • 145
    • 77952869512 scopus 로고    scopus 로고
    • Cucurbitacin B, a novel in vivo potentiator of gemcitabine with low toxicity in the treatment of pancreatic cancer
    • Iwanski GB, Lee DH, En-Gal S, et al. Cucurbitacin B, a novel in vivo potentiator of gemcitabine with low toxicity in the treatment of pancreatic cancer. Br J Pharmacol 2010;160:998-1007
    • (2010) Br J Pharmacol , vol.160 , pp. 998-1007
    • Iwanski, G.B.1    Lee, D.H.2    En-Gal, S.3
  • 146
    • 79959622792 scopus 로고    scopus 로고
    • Cucurbitacin i inhibits tumorigenic ability and enhances radiochemosensitivity in nonsmall cell lung cancer-derived cd133-positive cells
    • Hsu HS, Huang PI, Chang YL, et al. Cucurbitacin i inhibits tumorigenic ability and enhances radiochemosensitivity in nonsmall cell lung cancer-derived CD133-positive cells. Cancer 2011;117(13):2970-85
    • (2011) Cancer , vol.117 , Issue.13 , pp. 2970-2985
    • Hsu, H.S.1    Huang, P.I.2    Chang, Y.L.3
  • 147
    • 44149087816 scopus 로고    scopus 로고
    • Combined antitumor activity of cucurbitacin B and docetaxel in laryngeal cancer
    • Liu T, Zhang M, Zhang H, et al. Combined antitumor activity of cucurbitacin B and docetaxel in laryngeal cancer. Eur J Pharmacol 2008;587:78-84
    • (2008) Eur J Pharmacol , vol.587 , pp. 78-84
    • Liu, T.1    Zhang, M.2    Zhang, H.3
  • 148
    • 58149166871 scopus 로고    scopus 로고
    • Actin-aggregating cucurbitacins from Physocarpus capitatus
    • Maloney KN, Fujita M, Eggert US, et al. Actin-aggregating cucurbitacins from Physocarpus capitatus. J Nat Prod 2008;71:1927-9
    • (2008) J Nat Prod , vol.71 , pp. 1927-1929
    • Maloney, K.N.1    Fujita, M.2    Eggert, U.S.3
  • 149
    • 37549072685 scopus 로고    scopus 로고
    • Direct interaction of Cucurbitacin e isolated from Alsomitra macrocarpa to actin filament
    • Momma K, Masuzawa Y, Nakai N, et al. Direct interaction of Cucurbitacin E isolated from Alsomitra macrocarpa to actin filament. Cytotechnology 2008;56:33-9
    • (2008) Cytotechnology , vol.56 , pp. 33-39
    • Momma, K.1    Masuzawa, Y.2    Nakai, N.3
  • 150
    • 49749119221 scopus 로고    scopus 로고
    • Cucurbitacin B markedly inhibits growth and rapidly affects the cytoskeleton in glioblastoma multiforme
    • Yin D, Wakimoto N, Xing H, et al. Cucurbitacin B markedly inhibits growth and rapidly affects the cytoskeleton in glioblastoma multiforme. Int J Cancer 2008;123:1364-75
    • (2008) Int J Cancer , vol.123 , pp. 1364-1375
    • Yin, D.1    Wakimoto, N.2    Xing, H.3
  • 151
    • 78649674867 scopus 로고    scopus 로고
    • Cucurbitacin i inhibits cell motility by indirectly interfering with actin dynamics
    • Knecht DA, LaFleur RA, Kahsai AW, et al. Cucurbitacin I inhibits cell motility by indirectly interfering with actin dynamics. PLoS One 2010;5:e14039
    • (2010) PLoS One , vol.5
    • Knecht, D.A.1    Lafleur, R.A.2    Kahsai, A.W.3
  • 152
    • 79952281492 scopus 로고    scopus 로고
    • Cucurbitacin IIa: a novel class of anti-cancer drug inducing non-reversible actin aggregation and inhibiting survivin independent of JAK2/STAT3 phosphorylation
    • Boykin C, Zhang G, Chen YH, et al. Cucurbitacin IIa: a novel class of anti-cancer drug inducing non-reversible actin aggregation and inhibiting survivin independent of JAK2/STAT3 phosphorylation. Br J Cancer 2011;104:781-9
    • (2011) Br J Cancer , vol.104 , pp. 781-789
    • Boykin, C.1    Zhang, G.2    Chen, Y.H.3
  • 153
    • 0037444373 scopus 로고    scopus 로고
    • Discovery of JSI-124 (cucurbitacin I), a selective Janus kinase/signal transducer and activator of transcription 3 signaling pathway inhibitor with potent antitumor activity against human and murine cancer cells in mice
    • Blaskovich MA, Sun J, Cantor A, et al. Discovery of JSI-124 (cucurbitacin I), a selective Janus kinase/signal transducer and activator of transcription 3 signaling pathway inhibitor with potent antitumor activity against human and murine cancer cells in mice. Cancer Res 2003;63:1270-9
    • (2003) Cancer Res , vol.63 , pp. 1270-1279
    • Blaskovich, M.A.1    Sun, J.2    Cantor, A.3
  • 154
    • 18844370367 scopus 로고    scopus 로고
    • Cucurbitacin Q: A selective STAT3 activation inhibitor with potent antitumor activity
    • Sun J, Blaskovich MA, Jove R, et al. Cucurbitacin Q: a selective STAT3 activation inhibitor with potent antitumor activity. Oncogene 2005;24:3236-45
    • (2005) Oncogene , vol.24 , pp. 3236-3245
    • Sun, J.1    Blaskovich, M.A.2    Jove, R.3
  • 155
    • 33748341084 scopus 로고    scopus 로고
    • JSI-124 (cucurbitacin I) inhibits Janus kinase-3/signal transducer and activator of transcription-3 signalling, downregulatesnucleophosmin-anaplastic lymphoma kinase (ALK), and induces apoptosis in ALK-positive anaplastic large cell lymphoma cells
    • Shi X, Franko B, Frantz C, et al. JSI-124 (cucurbitacin I) inhibits Janus kinase-3/signal transducer and activator of transcription-3 signalling, downregulatesnucleophosmin-anaplastic lymphoma kinase (ALK), and induces apoptosis in ALK-positive anaplastic large cell lymphoma cells. Br J Haematol 2006;135:26-32
    • (2006) Br J Haematol , vol.135 , pp. 26-32
    • Shi, X.1    Franko, B.2    Frantz, C.3
  • 156
    • 78650488834 scopus 로고    scopus 로고
    • Inhibition of constitutive activation of STAT3 by curcurbitacin-I (JSI-124) sensitized human B-leukemia cells to apoptosis
    • Ishdorj G, Johnston JB, Gibson SB. Inhibition of constitutive activation of STAT3 by curcurbitacin-I (JSI-124) sensitized human B-leukemia cells to apoptosis. Mol Cancer Ther 2010;9:3302-14
    • (2010) Mol Cancer Ther , vol.9 , pp. 3302-3314
    • Ishdorj, G.1    Johnston, J.B.2    Gibson, S.B.3
  • 157
    • 77955199094 scopus 로고    scopus 로고
    • Inhibition of pancreatic cancer cell growth by cucurbitacin b through modulation of signal transducer and activator of transcription 3 signaling
    • Zhang M, Sun C, Shan X, et al. Inhibition of pancreatic cancer cell growth by cucurbitacin B through modulation of signal transducer and activator of transcription 3 signaling. Pancreas 2010;39:923-9
    • (2010) Pancreas , vol.39 , pp. 923-929
    • Zhang, M.1    Sun, C.2    Shan, X.3
  • 158
    • 0037427563 scopus 로고    scopus 로고
    • Anticancer and antiinflammatory activities of cucurbitacins from Cucurbita andreana
    • Jayaprakasam B, Seeram NP, Nair MG. Anticancer and antiinflammatory activities of cucurbitacins from Cucurbita andreana. Cancer Lett 2003;189:11-16
    • (2003) Cancer Lett , vol.189 , pp. 11-16
    • Jayaprakasam, B.1    Seeram, N.P.2    Nair, M.G.3
  • 159
    • 33847619674 scopus 로고    scopus 로고
    • Anti-inflammatory effects of a triterpenoid isolated from Wilbrandia ebracteata Cogn
    • Siqueira JM Jr, Peters RR, Gazola AC, et al. Anti-inflammatory effects of a triterpenoid isolated from Wilbrandia ebracteata Cogn. Life Sci 2007;80:1382-7
    • (2007) Life Sci , vol.80 , pp. 1382-1387
    • Siqueira Jr., J.M.1    Peters, R.R.2    Gazola, A.C.3
  • 160
    • 80054889550 scopus 로고    scopus 로고
    • Anti-inflammatory activities of cucurbitacin e isolated from citrullus lanatus var. citroides: Role of reactive nitrogen species and cyclooxygenase enzyme inhibition
    • Abdelwahab SI, Hassan LE, Sirat HM, et al. Anti-inflammatory activities of cucurbitacin E isolated from Citrullus lanatus var. citroides: role of reactive nitrogen species and cyclooxygenase enzyme inhibition. Fitoterapia 2011;82:1190-7
    • (2011) Fitoterapia , vol.82 , pp. 1190-1197
    • Abdelwahab, S.I.1    Hassan, L.E.2    Sirat, H.M.3
  • 161
    • 0036747181 scopus 로고    scopus 로고
    • Cucurbitacins from Trichosanthes kirilowii as the inhibitory components on tyrosinase activity and melanin synthesis of B16/F10 melanoma cells
    • Oh H, Mun YJ, Im SJ, et al. Cucurbitacins from Trichosanthes kirilowii as the inhibitory components on tyrosinase activity and melanin synthesis of B16/F10 melanoma cells. Planta Med 2002;68:832-3
    • (2002) Planta Med , vol.68 , pp. 832-833
    • Oh, H.1    Mun, Y.J.2    Im, S.J.3
  • 163
    • 0035751393 scopus 로고    scopus 로고
    • Cytotoxic triterpenoides from Alismatis Rhizoma
    • Lee S, Kho Y, Min B, et al. Cytotoxic triterpenoides from Alismatis Rhizoma. Arch Pharm Res 2001;24:524-6
    • (2001) Arch Pharm Res , vol.24 , pp. 524-526
    • Lee, S.1    Kho, Y.2    Min, B.3
  • 164
    • 77949736802 scopus 로고    scopus 로고
    • Alisol B, a novel inhibitor of the sarcoplasmic/endoplasmic reticulum Ca(2+) ATPase pump, induces autophagy, endoplasmic reticulum stress, and apoptosis
    • Law BY, Wang M, Ma DL, et al. Alisol B, a novel inhibitor of the sarcoplasmic/endoplasmic reticulum Ca(2+) ATPase pump, induces autophagy, endoplasmic reticulum stress, and apoptosis. Mol Cancer Ther 2010;9:718-30
    • (2010) Mol Cancer Ther , vol.9 , pp. 718-730
    • Law, B.Y.1    Wang, M.2    Ma, D.L.3
  • 165
    • 0041589549 scopus 로고    scopus 로고
    • Pharmacological evaluation of several major ingredients of Chinese herbal medicines in human hepatoma Hep3B cells
    • Chou CC, Pan SL, Teng CM, et al. Pharmacological evaluation of several major ingredients of Chinese herbal medicines in human hepatoma Hep3B cells. Eur J Pharm Sci 2003;19:403-12
    • (2003) Eur J Pharm Sci , vol.19 , pp. 403-412
    • Chou, C.C.1    Pan, S.L.2    Teng, C.M.3
  • 166
    • 29844445293 scopus 로고    scopus 로고
    • Alisol b acetate a triterpene from alismatis rhizoma induces bax nuclear translocation and apoptosis in human hormone-resistant prostate cancer PC-3 cells
    • Huang YT, Huang DM, Chueh SC, et al. Alisol B acetate, a triterpene from Alismatis rhizoma, induces Bax nuclear translocation and apoptosis in human hormone-resistant prostate cancer PC-3 cells. Cancer Lett 2006;231:270-8
    • (2006) Cancer Lett , vol.231 , pp. 270-278
    • Huang, Y.T.1    Huang, D.M.2    Chueh, S.C.3
  • 167
    • 3843091577 scopus 로고    scopus 로고
    • Reversal of P-glycoprotein-mediated multidrug resistance by Alisol B 23-acetate
    • Wang C, Zhang JX, Shen XL, et al. Reversal of P-glycoprotein-mediated multidrug resistance by Alisol B 23-acetate. Biochem Pharmacol 2004;68:843-55
    • (2004) Biochem Pharmacol , vol.68 , pp. 843-855
    • Wang, C.1    Zhang, J.X.2    Shen, X.L.3
  • 168
    • 0141794300 scopus 로고    scopus 로고
    • Influence of traditional Chinese anti-inflammatory medicinal plants on leukocyte and platelet functions
    • Prieto JM, Recio MC, Giner RM, et al. Influence of traditional Chinese anti-inflammatory medicinal plants on leukocyte and platelet functions. J Pharm Pharmacol 2003;55:1275-82
    • (2003) J Pharm Pharmacol , vol.55 , pp. 1275-1282
    • Prieto, J.M.1    Recio, M.C.2    Giner, R.M.3
  • 169
    • 0034063793 scopus 로고    scopus 로고
    • In vivo studies on the anti-inflammatory activity of pachymic and dehydrotumulosic acids
    • Giner EM, Manez S, Recio MC, et al. In vivo studies on the anti-inflammatory activity of pachymic and dehydrotumulosic acids. Planta Med 2000;66:221-7
    • (2000) Planta Med , vol.66 , pp. 221-227
    • Giner, E.M.1    Manez, S.2    Recio, M.C.3
  • 170
    • 79958723466 scopus 로고    scopus 로고
    • Pachymic acid impairs breast cancer cell invasion by suppressing nuclear factor-kappab-dependent matrix metalloproteinase-9 expression
    • Ling H, Zhang Y, Ng KY, et al. Pachymic acid impairs breast cancer cell invasion by suppressing nuclear factor-kappaB-dependent matrix metalloproteinase-9 expression. Breast Cancer Res Treat 2011;126:609-20
    • (2011) Breast Cancer Res Treat , vol.126 , pp. 609-620
    • Ling, H.1    Zhang, Y.2    Ng, K.Y.3
  • 171
    • 77949883363 scopus 로고    scopus 로고
    • Pachymic acid inhibits cell growth and modulates arachidonic acid metabolism in nonsmall cell lung cancer a549 cells
    • Ling H, Jia X, Zhang Y, et al. Pachymic acid inhibits cell growth and modulates arachidonic acid metabolism in nonsmall cell lung cancer A549 cells. Mol Carcinog 2010;49:271-82
    • (2010) Mol Carcinog , vol.49 , pp. 271-282
    • Ling, H.1    Jia, X.2    Zhang, Y.3
  • 172
    • 54349114907 scopus 로고    scopus 로고
    • Cytotoxic and anti-oxidant activities of lanostane-type triterpenes isolated from Poria cocos
    • Zhou L, Zhang Y, Gapter LA, et al. Cytotoxic and anti-oxidant activities of lanostane-type triterpenes isolated from Poria cocos. Chem Pharm Bull (Tokyo) 2008;56:1459-62
    • (2008) Chem Pharm Bull (Tokyo) , vol.56 , pp. 1459-1462
    • Zhou, L.1    Zhang, Y.2    Gapter, L.A.3
  • 173
    • 20444429394 scopus 로고    scopus 로고
    • Induction of apoptosis in prostate cancer cells by pachymic acid from Poria cocos
    • Gapter L, Wang Z, Glinski J, et al. Induction of apoptosis in prostate cancer cells by pachymic acid from Poria cocos. Biochem Biophys Res Commun 2005;332:1153-61
    • (2005) Biochem Biophys Res Commun , vol.332 , pp. 1153-1161
    • Gapter, L.1    Wang, Z.2    Glinski, J.3
  • 174
    • 16644389757 scopus 로고    scopus 로고
    • Cytotoxicity and DNA topoisomerases inhibitory activity of constituents from the sclerotium of Poria cocos
    • Li G, Xu ML, Lee CS, et al. Cytotoxicity and DNA topoisomerases inhibitory activity of constituents from the sclerotium of Poria cocos. Arch Pharm Res 2004;27:829-33
    • (2004) Arch Pharm Res , vol.27 , pp. 829-833
    • Li, G.1    Xu, M.L.2    Lee, C.S.3
  • 175
    • 1342306726 scopus 로고    scopus 로고
    • The role of carotenoids in the prevention of human pathologies
    • Tapiero H, Townsend DM, Tew KD. The role of carotenoids in the prevention of human pathologies. Biomed Pharmacother 2004;58:100-10
    • (2004) Biomed Pharmacother , vol.58 , pp. 100-110
    • Tapiero, H.1    Townsend, D.M.2    Tew, K.D.3
  • 176
    • 84858965591 scopus 로고    scopus 로고
    • Cancer chemoprevention by carotenoids
    • Tanaka T, Shnimizu M, Moriwaki H. Cancer chemoprevention by carotenoids. Molecules 2012;17:3202-42
    • (2012) Molecules , vol.17 , pp. 3202-3242
    • Tanaka, T.1    Shnimizu, M.2    Moriwaki, H.3
  • 177
    • 35148875476 scopus 로고    scopus 로고
    • Lycopene and soy isoflavones in the treatment of prostate cancer
    • Vaishampayan U, Hussain M, Banerjee M, et al. Lycopene and soy isoflavones in the treatment of prostate cancer. Nutr Cancer 2007;59:1-7
    • (2007) Nutr Cancer , vol.59 , pp. 1-7
    • Vaishampayan, U.1    Hussain, M.2    Banerjee, M.3
  • 178
    • 0034906966 scopus 로고    scopus 로고
    • Phase II randomized clinical trial of lycopene supplementation before radical prostatectomy
    • Kucuk O, Sarkar FH, Sakr W, et al. Phase II randomized clinical trial of lycopene supplementation before radical prostatectomy. Cancer Epidemiol Biomarkers Prev 2001;10:861-8
    • (2001) Cancer Epidemiol Biomarkers Prev , vol.10 , pp. 861-868
    • Kucuk, O.1    Sarkar, F.H.2    Sakr, W.3
  • 179
    • 33846675306 scopus 로고    scopus 로고
    • Combinations of tomato and broccoli enhance antitumor activity in dunning r3327-h prostate adenocarcinomas
    • Canene-Adams K, Lindshield BL, Wang S, et al. Combinations of tomato and broccoli enhance antitumor activity in dunning r3327-h prostate adenocarcinomas. Cancer Res 2007;67:836-43
    • (2007) Cancer Res , vol.67 , pp. 836-843
    • Canene-Adams, K.1    Lindshield, B.L.2    Wang, S.3
  • 180
    • 79960108284 scopus 로고    scopus 로고
    • Antioxidant and anti-proliferative properties of lycopene
    • Kelkel M, Schumacher M, Dicato M, et al. Antioxidant and anti-proliferative properties of lycopene. Free Radic Res 2011;45:925-40
    • (2011) Free Radic Res , vol.45 , pp. 925-940
    • Kelkel, M.1    Schumacher, M.2    Dicato, M.3
  • 181
    • 84865219359 scopus 로고    scopus 로고
    • Lycopene inhibits angiogenesis in human umbilical vein endothelial cells and rat aortic rings
    • Elgass S, Cooper A, Chopra M. Lycopene inhibits angiogenesis in human umbilical vein endothelial cells and rat aortic rings. Br J Nutr 2012;108:431-9
    • (2012) Br J Nutr , vol.108 , pp. 431-439
    • Elgass, S.1    Cooper, A.2    Chopra, M.3
  • 182
    • 84872761474 scopus 로고    scopus 로고
    • Anti-angiogenic effects of lycopene through immunomodualtion of cytokine secretion in human peripheral blood mononuclear cells
    • [Epub ahead of print]
    • Huang CS, Chuang CH, Lo TF, et al. Anti-angiogenic effects of lycopene through immunomodualtion of cytokine secretion in human peripheral blood mononuclear cells. J Nutr Biochem 2012; [Epub ahead of print]
    • (2012) J Nutr Biochem
    • Huang, C.S.1    Chuang, C.H.2    Lo, T.F.3
  • 183
    • 84862560347 scopus 로고    scopus 로고
    • Lycopene inhibits angiogenesis both in vitro and in vivo by inhibiting MMP-2/uPA system through VEGFR2-mediated PI3K-Akt and ERK/p38 signaling pathways
    • Chen ML, Lin YH, Yang CM, et al. Lycopene inhibits angiogenesis both in vitro and in vivo by inhibiting MMP-2/uPA system through VEGFR2-mediated PI3K-Akt and ERK/p38 signaling pathways. Mol Nutr Food Res 2012;56:889-99
    • (2012) Mol Nutr Food Res , vol.56 , pp. 889-899
    • Chen, M.L.1    Lin, Y.H.2    Yang, C.M.3
  • 184
    • 84876292530 scopus 로고    scopus 로고
    • Selective Inhibition of Cell Proliferation by Lycopene in MCF-7 Breast Cancer Cells in vitro: A Proteomic Analysis
    • Epub ahead of print
    • Uppala PT, Dissmore T, Lau BH, et al. Selective Inhibition of Cell Proliferation by Lycopene in MCF-7 Breast Cancer Cells In vitro: a Proteomic Analysis. Phytother Res 2012; Epub ahead of print
    • (2012) Phytother Res
    • Uppala, P.T.1    Dissmore, T.2    Lau, B.H.3
  • 185
    • 36049032810 scopus 로고    scopus 로고
    • Beta-Carotene induces apoptosis and up-regulates peroxisome proliferator-activated receptor gamma expression and reactive oxygen species production in MCF-7 cancer cells
    • Cui Y, Lu Z, Bai L, et al. beta-Carotene induces apoptosis and up-regulates peroxisome proliferator-activated receptor gamma expression and reactive oxygen species production in MCF-7 cancer cells. Eur J Cancer 2007;43:2590-601
    • (2007) Eur J Cancer , vol.43 , pp. 2590-2601
    • Cui, Y.1    Lu, Z.2    Bai, L.3
  • 186
    • 80052726769 scopus 로고    scopus 로고
    • Beta-Carotene and lutein inhibit hydrogen peroxide-induced activation of NF-kappaB and IL-8 expression in gastric epithelial AGS cells
    • Kim Y, Seo JH, Kim H. beta-Carotene and lutein inhibit hydrogen peroxide-induced activation of NF-kappaB and IL-8 expression in gastric epithelial AGS cells. J Nutr Sci Vitaminol (Tokyo) 2011;57:216-23
    • (2011) J Nutr Sci Vitaminol (Tokyo) , vol.57 , pp. 216-223
    • Kim, Y.1    Seo, J.H.2    Kim, H.3
  • 187
    • 84865222831 scopus 로고    scopus 로고
    • Carotenoids reverse multidrug resistance in cancer cells by interfering with ABC-transporters
    • Eid SY, El-Readi MZ, Wink M. Carotenoids reverse multidrug resistance in cancer cells by interfering with ABC-transporters. Phytomedicine 2012;19:977-87
    • (2012) Phytomedicine , vol.19 , pp. 977-987
    • Eid, S.Y.1    El-Readi, M.Z.2    Wink, M.3
  • 188
    • 32144459525 scopus 로고    scopus 로고
    • Reversal of multidrug resistance of cancer cells in vitro: Modification of drug resistance by selected carotenoids
    • Gyemant N, Tanaka M, Molnar P, et al. Reversal of multidrug resistance of cancer cells in vitro: modification of drug resistance by selected carotenoids. Anticancer Res 2006;26:367-74
    • (2006) Anticancer Res , vol.26 , pp. 367-374
    • Gyemant, N.1    Tanaka, M.2    Molnar, P.3
  • 189
    • 33750008443 scopus 로고    scopus 로고
    • Inhibition of NF-kappa B activation through targeting i kappa B kinase by celastrol, a quinone methide triterpenoid
    • Lee JH, Koo TH, Yoon H, et al. Inhibition of NF-kappa B activation through targeting I kappa B kinase by celastrol, a quinone methide triterpenoid. Biochem Pharmacol 2006;72:1311-21
    • (2006) Biochem Pharmacol , vol.72 , pp. 1311-1321
    • Lee, J.H.1    Koo, T.H.2    Yoon, H.3
  • 190
    • 33646406554 scopus 로고    scopus 로고
    • Celastrol a triterpene extracted from the Chinese "thunder of God Vine," is a potent proteasome inhibitor and suppresses human prostate cancer growth in nude mice
    • Yang H, Chen D, Cui QC, et al. Celastrol, a triterpene extracted from the Chinese "Thunder of God Vine," is a potent proteasome inhibitor and suppresses human prostate cancer growth in nude mice. Cancer Res 2006;66:4758-65
    • (2006) Cancer Res , vol.66 , pp. 4758-4765
    • Yang, H.1    Chen, D.2    Cui, Q.C.3
  • 191
    • 2842616065 scopus 로고    scopus 로고
    • Apoptosis induction in HL-60 cells and inhibition of topoisomerase II by triterpene celastrol
    • Nagase M, Oto J, Sugiyama S, et al. Apoptosis induction in HL-60 cells and inhibition of topoisomerase II by triterpene celastrol. Biosci Biotechnol Biochem 2003;67:1883-7
    • (2003) Biosci Biotechnol Biochem , vol.67 , pp. 1883-1887
    • Nagase, M.1    Oto, J.2    Sugiyama, S.3
  • 192
    • 33749433916 scopus 로고    scopus 로고
    • Gene expression signature-based chemical genomic prediction identifies a novel class of HSP90 pathway modulators
    • Hieronymus H, Lamb J, Ross KN, et al. Gene expression signature-based chemical genomic prediction identifies a novel class of HSP90 pathway modulators. Cancer Cell 2006;10:321-30
    • (2006) Cancer Cell , vol.10 , pp. 321-330
    • Hieronymus, H.1    Lamb, J.2    Ross, K.N.3
  • 193
    • 38349153572 scopus 로고    scopus 로고
    • A novel Hsp90 inhibitor to disrupt Hsp90/Cdc37 complex against pancreatic cancer cells
    • Zhang T, Hamza A, Cao X, et al. A novel Hsp90 inhibitor to disrupt Hsp90/Cdc37 complex against pancreatic cancer cells. Mol Cancer Ther 2008;7:162-70
    • (2008) Mol Cancer Ther , vol.7 , pp. 162-170
    • Zhang, T.1    Hamza, A.2    Cao, X.3
  • 194
    • 41649104650 scopus 로고    scopus 로고
    • Activation of heat shock and antioxidant responses by the natural product celastrol: Transcriptional signatures of a thiol-targeted molecule
    • Trott A, West JD, Klaic L, et al. Activation of heat shock and antioxidant responses by the natural product celastrol: transcriptional signatures of a thiol-targeted molecule. Mol Biol Cell 2008;19:1104-12
    • (2008) Mol Biol Cell , vol.19 , pp. 1104-1112
    • Trott, A.1    West, J.D.2    Klaic, L.3
  • 195
    • 77950818403 scopus 로고    scopus 로고
    • HSP90 inhibitor, celastrol, arrests human monocytic leukemia cell U937 at G0/G1 in thiol-containing agents reversible way
    • Peng B, Xu L, Cao F, et al. HSP90 inhibitor, celastrol, arrests human monocytic leukemia cell U937 at G0/G1 in thiol-containing agents reversible way. Mol Cancer 2010;9:79
    • (2010) Mol Cancer , vol.9 , pp. 79
    • Peng, B.1    Xu, L.2    Cao, F.3
  • 196
    • 77954917015 scopus 로고    scopus 로고
    • Celastrol acts as a potent antimetastatic agent targeting beta1 integrin and inhibiting cell-extracellular matrix adhesion, in part via the p38 mitogen-activated protein kinase pathway
    • Zhu H, Liu XW, Cai TY, et al. Celastrol acts as a potent antimetastatic agent targeting beta1 integrin and inhibiting cell-extracellular matrix adhesion, in part via the p38 mitogen-activated protein kinase pathway. J Pharmacol Exp Ther 2010;334:489-99
    • (2010) J Pharmacol Exp Ther , vol.334 , pp. 489-499
    • Zhu, H.1    Liu, X.W.2    Cai, T.Y.3
  • 197
    • 42649088411 scopus 로고    scopus 로고
    • Celastrol inhibits the growth of human glioma xenografts in nude mice through suppressing VEGFR expression
    • Huang Y, Zhou Y, Fan Y, et al. Celastrol inhibits the growth of human glioma xenografts in nude mice through suppressing VEGFR expression. Cancer Lett 2008;264:101-6
    • (2008) Cancer Lett , vol.264 , pp. 101-106
    • Huang, Y.1    Zhou, Y.2    Fan, Y.3
  • 198
    • 84455163076 scopus 로고    scopus 로고
    • Synergistic antitumor activity of oridonin and arsenic trioxide on hepatocellular carcinoma cells
    • Chen G, Wang K, Yang BY, et al. Synergistic antitumor activity of oridonin and arsenic trioxide on hepatocellular carcinoma cells. Int J Oncol 2012;40:139-47
    • (2012) Int J Oncol , vol.40 , pp. 139-147
    • Chen, G.1    Wang, K.2    Yang, B.Y.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.