1,272
Views
214
CrossRef citations to date
0
Altmetric
Reviews

Terpenoids: natural products for cancer therapy

, , , , &
Pages 1801-1818 | Published online: 23 Oct 2012

Bibliography

  • 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
  • Gershenzon J, Dudareva N. The function of terpene natural products in the natural world. Nat Chem Biol 2007;3:408-14
  • Lai HC, Singh NP, Sasaki T. Development of artemisinin compounds for cancer treatment. Invest New Drugs 2012. [Epub ahead of print]
  • Petronelli A, Pannitteri G, Testa U. Triterpenoids as new promising anticancer drugs. Anticancer Drugs 2009;20:880-92
  • Sun J. D-Limonene: safety and clinical applications. Altern Med Rev 2007;12:259-64
  • 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
  • Vigushin DM, Poon GK, Boddy A, 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
  • Clegg RJ, Middleton B, Bell GD, 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
  • Kawata S, Nagase T, Yamasaki E, 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
  • Kaji I, Tatsuta M, Iishi H, 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
  • Ji J, Zhang L, Wu YY, 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
  • Liu D, Chen Z. The effects of cantharidin and cantharidin derivates on tumour cells. Anticancer Agents Med Chem 2009;9:392-6
  • McCluskey A, Bowyer MC, Collins E, Anhydride modified cantharidin analogues: synthesis, inhibition of protein phosphatases 1 and 2A and anticancer activity. Bioorg Med Chem Lett 2000;10:1687-90
  • Hennings H, Boutwell RK. Studies on the mechanism of skin tumor promotion. Cancer Res 1970;30:312-20
  • Chen YN, Chen JC, Yin SC, Effector mechanisms of norcantharidin-induced mitotic arrest and apoptosis in human hepatoma cells. Int J Cancer 2002;100:158-65
  • Huan SK, Lee HH, Liu DZ, Cantharidin-induced cytotoxicity and cyclooxygenase 2 expression in human bladder carcinoma cell line. Toxicology 2006;223:136-43
  • Huh JE, Kang KS, Chae C, Roles of p38 and JNK mitogen-activated protein kinase pathways during cantharidin-induced apoptosis in U937 cells. Biochem Pharmacol 2004;67:1811-18
  • 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
  • Sakoff JA, Ackland SP, Baldwin ML, Anticancer activity and protein phosphatase 1 and 2A inhibition of a new generation of cantharidin analogues. Invest New Drugs 2002;20:1-11
  • McCluskey A, Ackland SP, Bowyer MC, Cantharidin analogues: synthesis and evaluation of growth inhibition in a panel of selected tumour cell lines. Bioorg Chem 2003;31:68-79
  • Bertini I, Calderone V, Fragai M, Structural basis of serine/threonine phosphatase inhibition by the archetypal small molecules cantharidin and norcantharidin. J Med Chem 2009;52:4838-43
  • Kalev P, Sablina AA. Protein phosphatase 2A as a potential target for anticancer therapy. Anticancer Agents Med Chem 2011;11:38-46
  • Li W, Chen Z, Zong Y, 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
  • Li W, Xie L, Chen Z, 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
  • Meshnick SR. Artemisinin antimalarials: mechanisms of action and resistance. Med Trop (Mars) 1998;58:13-17
  • 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
  • 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
  • Yang ZS, Zhou WL, Sui Y, 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
  • Firestone GL, Sundar SN. Anticancer activities of artemisinin and its bioactive derivatives. Expert Rev Mol Med 2009;11:e32
  • Tan W, Lu J, Huang M, Anti-cancer natural products isolated from chinese medicinal herbs. Chin Med 2011;6:27
  • Chaturvedi D, Goswami A, Saikia PP, Artemisinin and its derivatives: a novel class of anti-malarial and anti-cancer agents. Chem Soc Rev 2010;39:435-54
  • Jiao Y, Ge CM, Meng QH, Dihydroartemisinin is an inhibitor of ovarian cancer cell growth. Acta Pharmacol Sin 2007;28:1045-56
  • Hou J, Wang D, Zhang R, 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
  • Lu JJ, Meng LH, Cai YJ, 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
  • Chen H, Sun B, Pan S, Dihydroartemisinin inhibits growth of pancreatic cancer cells in vitro and in vivo. Anticancer Drugs 2009;20:131-40
  • He Q, Shi J, Shen XL, 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
  • Lu JJ, Chen SM, Zhang XW, 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
  • Lu JJ, Meng LH, Shankavaram UT, Dihydroartemisinin accelerates c-MYC oncoprotein degradation and induces apoptosis in c-MYC-overexpressing tumor cells. Biochem Pharmacol 2010;80:22-30
  • Wang SJ, Gao Y, Chen H, 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
  • Li S, Xue F, Cheng Z, 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
  • Efferth T, Dunstan H, Sauerbrey A, The anti-malarial artesunate is also active against cancer. Int J Oncol 2001;18:767-73
  • Efferth T, Sauerbrey A, Olbrich A, Molecular modes of action of artesunate in tumor cell lines. Mol Pharmacol 2003;64:382-94
  • Michaelis M, Kleinschmidt MC, Barth S, Anti-cancer effects of artesunate in a panel of chemoresistant neuroblastoma cell lines. Biochem Pharmacol 2010;79:130-6
  • 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
  • Lu JJ, Chen SM, Ding J, Characterization of dihydroartemisinin-resistant colon carcinoma HCT116/R cell line. Mol Cell Biochem 2012;360:329-37
  • Du JH, Zhang HD, Ma ZJ, 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
  • Chen T, Li M, Zhang R, Dihydroartemisinin induces apoptosis and sensitizes human ovarian cancer cells to carboplatin therapy. J Cell Mol Med 2009;13:1358-70
  • Zhang S, Gerhard GS. Heme mediates cytotoxicity from artemisinin and serves as a general anti-proliferation target. PLoS One 2009;4:e7472
  • Efferth T, Benakis A, Romero MR, Enhancement of cytotoxicity of artemisinins toward cancer cells by ferrous iron. Free Radic Biol Med 2004;37:998-1009
  • Sertel S, Eichhorn T, Sieber S, Factors determining sensitivity or resistance of tumor cell lines towards artesunate. Chem Biol Interact 2010;185:42-52
  • Willoughby JA Sr, Sundar SN, Cheung M, 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
  • Chen H, Sun B, Wang S, 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
  • Mu D, Zhang W, Chu D, The role of calcium, P38 MAPK in dihydroartemisinin-induced apoptosis of lung cancer PC-14 cells. Cancer Chemother Pharmacol 2008;61:639-45
  • Handrick R, Ontikatze T, Bauer KD, Dihydroartemisinin induces apoptosis by a Bak-dependent intrinsic pathway. Mol Cancer Ther 2010;9:2497-510
  • Rasheed SA, Efferth T, Asangani IA, 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
  • Hwang YP, Yun HJ, Kim HG, 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
  • Chen HH, Zhou HJ, Wang WQ, Antimalarial dihydroartemisinin also inhibits angiogenesis. Cancer Chemother Pharmacol 2004;53:423-32
  • Buommino E, Baroni A, Canozo N, 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
  • Zhou L, Zuo Z, Chow MS. Danshen: an overview of its chemistry, pharmacology, pharmacokinetics, and clinical use. J Clin Pharmacol 2005;45:1345-59
  • Sung HJ, Choi SM, Yoon Y, 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
  • Liu JJ, Lin DJ, Liu PQ, 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
  • Wang X, Wei Y, Yuan S, Potential anticancer activity of tanshinone IIA against human breast cancer. Int J Cancer 2005;116:799-807
  • 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
  • Su CC, Chen GW, Kang JC, Growth inhibition and apoptosis induction by tanshinone IIA in human colon adenocarcinoma cells. Planta Med 2008;74:1357-62
  • Wu WL, Chang WL, Chen CF. Cytotoxic activities of tanshinones against human carcinoma cell lines. Am J Chin Med 1991;19:207-16
  • 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
  • Yuan SL, Wei YQ, Wang XJ, Growth inhibition and apoptosis induction of tanshinone II-A on human hepatocellular carcinoma cells. World J Gastroenterol 2004;10:2024-8
  • Lee WY, Cheung CC, Liu KW, Cytotoxic effects of tanshinones from Salvia miltiorrhiza on doxorubicin-resistant human liver cancer cells. J Nat Prod 2010;73:854-9
  • 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
  • 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
  • Dai ZK, Qin JK, Huang JE, Tanshinone IIA activates calcium-dependent apoptosis signaling pathway in human hepatoma cells. J Nat Med 2011;66(1):192-201
  • Won SH, Lee HJ, Jeong SJ, 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
  • 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
  • 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
  • Zhang K, Li J, Meng W, 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
  • Wang J, Wang X, Jiang S, Growth inhibition and induction of apoptosis and differentiation of tanshinone IIA in human glioma cells. J Neurooncol 2007;82:11-21
  • Yuxian X, Feng T, Ren L, Tanshinone II-A inhibits invasion and metastasis of human hepatocellular carcinoma cells in vitro and in vivo. Tumori 2009;95:789-95
  • Shan YF, Shen X, Xie YK, Inhibitory effects of tanshinone II-A on invasion and metastasis of human colon carcinoma cells. Acta Pharmacol Sin 2009;30:1537-42
  • Tsai MY, Yang RC, Wu HT, 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
  • Lee CY, Sher HF, Chen HW, Anticancer effects of tanshinone I in human non-small cell lung cancer. Mol Cancer Ther 2008;7:3527-38
  • 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
  • Nizamutdinova IT, Lee GW, Son KH, 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
  • Nizamutdinova IT, Lee GW, Lee JS, Tanshinone I suppresses growth and invasion of human breast cancer cells, MDA-MB-231, through regulation of adhesion molecules. Carcinogenesis 2008;29:1885-92
  • Liu JJ, Liu WD, Yang HZ, 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
  • 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
  • Park IJ, Kim MJ, Park OJ, Cryptotanshinone sensitizes DU145 prostate cancer cells to Fas(APO1/CD95)-mediated apoptosis through Bcl-2 and MAPK regulation. Cancer Lett 2010;298:88-98
  • Liu Q. Triptolide and its expanding multiple pharmacological functions. Int Immunopharmacol 2011;11:377-83
  • Qiu D, Zhao G, Aoki Y, 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
  • Westerheide SD, Kawahara TL, Orton K, Triptolide, an inhibitor of the human heat shock response that enhances stress-induced cell death. J Biol Chem 2006;281:9616-22
  • Chang WT, Kang JJ, Lee KY, Triptolide and chemotherapy cooperate in tumor cell apoptosis. A role for the p53 pathway. J Biol Chem 2001;276:2221-7
  • McCallum C, Kwon S, Leavitt P, Triptolide binds covalently to a 90 kDa nuclear protein. Role of epoxides in binding and activity. Immunobiology 2007;212:549-56
  • Vispe S, DeVries L, Creancier L, 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
  • Wang Y, Lu JJ, He L, Triptolide (TPL) Inhibits Global Transcription by Inducing Proteasome-Dependent Degradation of RNA Polymerase II (Pol II). PLoS One 2011;6:e23993
  • Leuenroth SJ, Okuhara D, Shotwell JD, Triptolide is a traditional Chinese medicine-derived inhibitor of polycystic kidney disease. Proc Natl Acad Sci USA 2007;104:4389-94
  • Titov DV, Gilman B, He QL, XPB, a subunit of TFIIH, is a target of the natural product triptolide. Nat Chem Biol 2011;7:182-8
  • Zhou ZL, Luo ZG, Yu B, Increased accumulation of hypoxia-inducible factor-1alpha with reduced transcriptional activity mediates the antitumor effect of triptolide. Mol Cancer 2010;9:268
  • Chiu P, Leung LT, Ko BC. Pseudolaric acids: isolation, bioactivity and synthetic studies. Nat Prod Rep 2010;27:1066-83
  • Pan DJ, Li ZL, Hu CQ, The cytotoxic principles of Pseudolarix kaempferi: pseudolaric acid-A and -B and related derivatives. Planta Med 1990;56:383-5
  • Wong VK, Chiu P, Chung SS, 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
  • Jordan MA, Wilson L. Microtubules as a target for anticancer drugs. Nat Rev Cancer 2004;4:253-65
  • 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
  • Tan WF, Zhang XW, Li MH, Pseudolarix acid B inhibits angiogenesis by antagonizing the vascular endothelial growth factor-mediated anti-apoptotic effect. Eur J Pharmacol 2004;499:219-28
  • Li MH, Miao ZH, Tan WF, Pseudolaric acid B inhibits angiogenesis and reduces hypoxia-inducible factor 1alpha by promoting proteasome-mediated degradation. Clin Cancer Res 2004;10:8266-74
  • Yu JH, Wang HJ, Li XR, 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
  • Yu J, Li X, Tashiro S, Bcl-2 family proteins were involved in pseudolaric acid B-induced autophagy in murine fibrosarcoma L929 cells. J Pharmacol Sci 2008;107:295-302
  • Lim CW, Chan TK, Ng DS, Andrographolide and its analogues: versatile bioactive molecules for combating inflammation and cancer. Clin Exp Pharmacol Physiol 2011;39(3):300-10
  • Yu BC, Hung CR, Chen WC, Antihyperglycemic effect of andrographolide in streptozotocin-induced diabetic rats. Planta Med 2003;69:1075-9
  • Xia YF, Ye BQ, Li YD, 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
  • Wang YJ, Wang JT, Fan QX, Andrographolide inhibits NF-kappaBeta activation and attenuates neointimal hyperplasia in arterial restenosis. Cell Res 2007;17:933-41
  • Wang LJ, Zhou X, Wang W, Andrographolide inhibits oral squamous cell carcinogenesis through NF-kappaB inactivation. J Dent Res 2011;90:1246-52
  • Kuttan G, Pratheeshkumar P, Manu KA, Inhibition of tumor progression by naturally occurring terpenoids. Pharm Biol 2011;49:995-1007
  • Ikezoe T, Yang Y, Bandobashi K, 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
  • Jin H, Tan X, Liu X, 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
  • Hu HZ, Yang YB, Xu XD, Oridonin induces apoptosis via PI3K/Akt pathway in cervical carcinoma HeLa cell line. Acta Pharmacol Sin 2007;28:1819-26
  • Zhang Y, Wu Y, Wu D, NF-kappab facilitates oridonin-induced apoptosis and autophagy in HT1080 cells through a p53-mediated pathway. Arch Biochem Biophys 2009;489:25-33
  • Cheng Y, Qiu F, Ikejima T. Molecular mechanisms of oridonin-induced apoptosis and autophagy in murine fibrosarcoma L929 cells. Autophagy 2009;5:430-1
  • Cheng Y, Qiu F, Ye YC, 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
  • Zhang YH, Wu YL, Tashiro S, Reactive oxygen species contribute to oridonin-induced apoptosis and autophagy in human cervical carcinoma HeLa cells. Acta Pharmacol Sin 2011;32:1266-75
  • Calixto JB, Campos MM, Otuki MF, Anti-inflammatory compounds of plant origin. Part II. modulation of pro-inflammatory cytokines, chemokines and adhesion molecules. Planta Med 2004;70:93-103
  • Allison AC, Cacabelos R, Lombardi VR, Celastrol, a potent antioxidant and anti-inflammatory drug, as a possible treatment for Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry 2001;25:1341-57
  • Li H, Zhang YY, Huang XY, Beneficial effect of tripterine on systemic lupus erythematosus induced by active chromatin in BALB/c mice. Eur J Pharmacol 2005;512:231-7
  • Zhang DH, Marconi A, Xu LM, Tripterine inhibits the expression of adhesion molecules in activated endothelial cells. J Leukoc Biol 2006;80:309-19
  • Pinna GF, Fiorucci M, Reimund JM, Celastrol inhibits pro-inflammatory cytokine secretion in Crohn's disease biopsies. Biochem Biophys Res Commun 2004;322:778-86
  • Yu X, Tao W, Jiang F, 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
  • Seo WY, Ju SM, Song HY, 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
  • Pang X, Yi Z, Zhang J, Celastrol suppresses angiogenesis-mediated tumor growth through inhibition of AKT/mammalian target of rapamycin pathway. Cancer Res 2010;70:1951-9
  • Yang H, Chen D, Cui QC, 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
  • Sethi G, Ahn KS, Pandey MK, 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
  • Salminen A, Lehtonen M, Paimela T, Celastrol: molecular targets of Thunder God Vine. Biochem Biophys Res Commun 2010;394:439-42
  • Chambliss OL, Jones CM. Cucurbitacins: specific insect attractants in Cucurbitaceae. Science 1966;153:1392-3
  • Miró M. Cucurbitacins and their pharmacological effects. Phytotherapy Research 1995;9:159-68
  • Lee DH, Iwanski GB, Thoennissen NH. Cucurbitacin: ancient compound shedding new light on cancer treatment. Sci World J 2010;10:413-18
  • Li Y, Wang R, Ma E, 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
  • Chen W, Leiter A, Yin D, 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
  • Haritunians T, Gueller S, Zhang L, Cucurbitacin B induces differentiation, cell cycle arrest, and actin cytoskeletal alterations in myeloid leukemia cells. Leuk Res 2008;32:1366-73
  • Chan KT, Meng FY, Li Q, 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
  • Hsu HS, Huang PI, Chang YL, Cucurbitacin I inhibits tumorigenic ability and enhances radiochemosensitivity in nonsmall cell lung cancer-derived CD133-positive cells. Cancer 2011;117:2970-85
  • Ouyang D, Zhang Y, Xu L, Histone deacetylase inhibitor valproic acid sensitizes B16F10 melanoma cells to cucurbitacin B treatment. Acta Biochim Biophys Sin (Shanghai) 2011;43:487-95
  • Chen YW, Chen KH, Huang PI, 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
  • Lui VW, Yau DM, Wong EY, Cucurbitacin I elicits anoikis sensitization, inhibits cellular invasion and in vivo tumor formation ability of nasopharyngeal carcinoma cells. Carcinogenesis 2009;30:2085-94
  • Rivat C, Rodrigues S, Bruyneel E, 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
  • Tang JZ, Kong XJ, Banerjee A, STAT3alpha is oncogenic for endometrial carcinoma cells and mediates the oncogenic effects of autocrine human growth hormone. Endocrinology 2010;151:4133-45
  • Shohat B, Gitter S, Levie B, The combined effect of cucurbitacins and X-ray treatment on transplanted tumors in mice. Cancer Res 1965;25:1828-35
  • Thoennissen NH, Iwanski GB, Doan NB, 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
  • Iwanski GB, Lee DH, En-Gal S, 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
  • Hsu HS, Huang PI, Chang YL, Cucurbitacin i inhibits tumorigenic ability and enhances radiochemosensitivity in nonsmall cell lung cancer-derived CD133-positive cells. Cancer 2011;117(13):2970-85
  • Liu T, Zhang M, Zhang H, Combined antitumor activity of cucurbitacin B and docetaxel in laryngeal cancer. Eur J Pharmacol 2008;587:78-84
  • Maloney KN, Fujita M, Eggert US, Actin-aggregating cucurbitacins from Physocarpus capitatus. J Nat Prod 2008;71:1927-9
  • Momma K, Masuzawa Y, Nakai N, Direct interaction of Cucurbitacin E isolated from Alsomitra macrocarpa to actin filament. Cytotechnology 2008;56:33-9
  • Yin D, Wakimoto N, Xing H, Cucurbitacin B markedly inhibits growth and rapidly affects the cytoskeleton in glioblastoma multiforme. Int J Cancer 2008;123:1364-75
  • Knecht DA, LaFleur RA, Kahsai AW, Cucurbitacin I inhibits cell motility by indirectly interfering with actin dynamics. PLoS One 2010;5:e14039
  • Boykin C, Zhang G, Chen YH, 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
  • Blaskovich MA, Sun J, Cantor A, 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
  • Sun J, Blaskovich MA, Jove R, Cucurbitacin Q: a selective STAT3 activation inhibitor with potent antitumor activity. Oncogene 2005;24:3236-45
  • Shi X, Franko B, Frantz C, 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
  • 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
  • Zhang M, Sun C, Shan X, 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
  • Jayaprakasam B, Seeram NP, Nair MG. Anticancer and antiinflammatory activities of cucurbitacins from Cucurbita andreana. Cancer Lett 2003;189:11-16
  • Siqueira JM Jr, Peters RR, Gazola AC, Anti-inflammatory effects of a triterpenoid isolated from Wilbrandia ebracteata Cogn. Life Sci 2007;80:1382-7
  • Abdelwahab SI, Hassan LE, Sirat HM, 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
  • Oh H, Mun YJ, Im SJ, 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
  • The State Pharmacopoeia Commission of P.R.China. Pharmacopoeia of the People's Republic of China, 2005
  • Lee S, Kho Y, Min B, Cytotoxic triterpenoides from Alismatis Rhizoma. Arch Pharm Res 2001;24:524-6
  • Law BY, Wang M, Ma DL, 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
  • Chou CC, Pan SL, Teng CM, Pharmacological evaluation of several major ingredients of Chinese herbal medicines in human hepatoma Hep3B cells. Eur J Pharm Sci 2003;19:403-12
  • Huang YT, Huang DM, Chueh SC, 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
  • Wang C, Zhang JX, Shen XL, Reversal of P-glycoprotein-mediated multidrug resistance by Alisol B 23-acetate. Biochem Pharmacol 2004;68:843-55
  • Prieto JM, Recio MC, Giner RM, Influence of traditional Chinese anti-inflammatory medicinal plants on leukocyte and platelet functions. J Pharm Pharmacol 2003;55:1275-82
  • Giner EM, Manez S, Recio MC, In vivo studies on the anti-inflammatory activity of pachymic and dehydrotumulosic acids. Planta Med 2000;66:221-7
  • Ling H, Zhang Y, Ng KY, 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
  • Ling H, Jia X, Zhang Y, Pachymic acid inhibits cell growth and modulates arachidonic acid metabolism in nonsmall cell lung cancer A549 cells. Mol Carcinog 2010;49:271-82
  • Zhou L, Zhang Y, Gapter LA, Cytotoxic and anti-oxidant activities of lanostane-type triterpenes isolated from Poria cocos. Chem Pharm Bull (Tokyo) 2008;56:1459-62
  • Gapter L, Wang Z, Glinski J, Induction of apoptosis in prostate cancer cells by pachymic acid from Poria cocos. Biochem Biophys Res Commun 2005;332:1153-61
  • Li G, Xu ML, Lee CS, Cytotoxicity and DNA topoisomerases inhibitory activity of constituents from the sclerotium of Poria cocos. Arch Pharm Res 2004;27:829-33
  • Tapiero H, Townsend DM, Tew KD. The role of carotenoids in the prevention of human pathologies. Biomed Pharmacother 2004;58:100-10
  • Tanaka T, Shnimizu M, Moriwaki H. Cancer chemoprevention by carotenoids. Molecules 2012;17:3202-42
  • Vaishampayan U, Hussain M, Banerjee M, Lycopene and soy isoflavones in the treatment of prostate cancer. Nutr Cancer 2007;59:1-7
  • Kucuk O, Sarkar FH, Sakr W, Phase II randomized clinical trial of lycopene supplementation before radical prostatectomy. Cancer Epidemiol Biomarkers Prev 2001;10:861-8
  • Canene-Adams K, Lindshield BL, Wang S, Combinations of tomato and broccoli enhance antitumor activity in dunning r3327-h prostate adenocarcinomas. Cancer Res 2007;67:836-43
  • Kelkel M, Schumacher M, Dicato M, Antioxidant and anti-proliferative properties of lycopene. Free Radic Res 2011;45:925-40
  • 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
  • Huang CS, Chuang CH, Lo TF, Anti-angiogenic effects of lycopene through immunomodualtion of cytokine secretion in human peripheral blood mononuclear cells. J Nutr Biochem 2012; [Epub ahead of print]
  • Chen ML, Lin YH, Yang CM, 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
  • Uppala PT, Dissmore T, Lau BH, 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
  • Cui Y, Lu Z, Bai L, 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
  • 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
  • Eid SY, El-Readi MZ, Wink M. Carotenoids reverse multidrug resistance in cancer cells by interfering with ABC-transporters. Phytomedicine 2012;19:977-87
  • Gyemant N, Tanaka M, Molnar P, Reversal of multidrug resistance of cancer cells in vitro: modification of drug resistance by selected carotenoids. Anticancer Res 2006;26:367-74
  • Lee JH, Koo TH, Yoon H, Inhibition of NF-kappa B activation through targeting I kappa B kinase by celastrol, a quinone methide triterpenoid. Biochem Pharmacol 2006;72:1311-21
  • Yang H, Chen D, Cui QC, 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
  • Nagase M, Oto J, Sugiyama S, Apoptosis induction in HL-60 cells and inhibition of topoisomerase II by triterpene celastrol. Biosci Biotechnol Biochem 2003;67:1883-7
  • Hieronymus H, Lamb J, Ross KN, Gene expression signature-based chemical genomic prediction identifies a novel class of HSP90 pathway modulators. Cancer Cell 2006;10:321-30
  • Zhang T, Hamza A, Cao X, A novel Hsp90 inhibitor to disrupt Hsp90/Cdc37 complex against pancreatic cancer cells. Mol Cancer Ther 2008;7:162-70
  • Trott A, West JD, Klaic L, 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
  • Peng B, Xu L, Cao F, HSP90 inhibitor, celastrol, arrests human monocytic leukemia cell U937 at G0/G1 in thiol-containing agents reversible way. Mol Cancer 2010;9:79
  • Zhu H, Liu XW, Cai TY, 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
  • Huang Y, Zhou Y, Fan Y, Celastrol inhibits the growth of human glioma xenografts in nude mice through suppressing VEGFR expression. Cancer Lett 2008;264:101-6
  • Chen G, Wang K, Yang BY, Synergistic antitumor activity of oridonin and arsenic trioxide on hepatocellular carcinoma cells. Int J Oncol 2012;40:139-47

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.