Anticancer Effect of AntiMalarial Artemisinin Compounds

A K Das, A K Das

Abstract

The anti-malarial drug artemisinin has shown anticancer activity in vitro and animal experiments, but experience in human cancer is scarce. However, the ability of artemisinins to kill cancer cells through a variety of molecular mechanisms has been explored. A PubMed search of about 127 papers on anti-cancer effects of antimalarials has revealed that this class of drug, including other antimalarials, have several biological characteristics that include anticancer properties. Experimental evidences suggest that artemisinin compounds may be a therapeutic alternative in highly aggressive cancers with rapid dissemination, without developing drug resistance. They also exhibit synergism with other anticancer drugs with no increased toxicity toward normal cells. It has been found that semisynthetic artemisinin derivatives have much higher antitumor activity than their monomeric counterparts via mechanisms like apoptosis, arrest of cell cycle at G0/G1, and oxidative stress. The exact mechanism of activation and molecular basis of these anticancer effects are not fully elucidated. Artemisinins seem to regulate key factors such as nuclear factor-kappa B, survivin, NOXA, hypoxia-inducible factor-1α, and BMI-1, involving multiple pathways that may affect drug response, drug interactions, drug resistance, and associated parameters upon normal cells. Newer synthetic artemisinins have been developed showing substantial antineoplastic activity, but there is still limited information regarding the mode of action of these synthetic compounds. In view of the emerging data, specific interactions with established chemotherapy need to be further investigated in different cancer cells and their phenotypes and validated further using different semisynthetic and synthetic artemisinin derivatives.

Keywords: Anticancer agents; Antimalarials; Antitumor activity; Artemisinins; Novel chemotherapy.

Conflict of interest statement

Conflict of Interest: None declared.

References

    1. Hsiao WL, Liu L. The role of traditional Chinese herbal medicines in cancer therapy – From TCM theory to mechanistic insights. Planta Med. 2010;76:1118–31.
    1. Hurria A, Togawa K, Mohile SG, Owusu C, Klepin HD, Gross CP, et al. Predicting chemotherapy toxicity in older adults with cancer: A prospective multicenter study. J Clin Oncol. 2011;29:3457–65.
    1. Lai HC, Singh NP, Sasaki T. Development of artemisinin compounds for cancer treatment. Invest New Drugs. 2013;31:230–46.
    1. Efferth T, Dunstan H, Sauerbrey A, Miyachi H, Chitambar CR. The anti-malarial artesunate is also active against cancer. Int J Oncol. 2001;18:767–73.
    1. Gordi T, Lepist EI. Artemisinin derivatives: Toxic for laboratory animals, safe for humans? Toxicol Lett. 2004;147:99–107.
    1. Dondorp AM, Nosten F, Yi P, Das D, Phyo AP, Tarning J, et al. Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2009;361:455–67.
    1. Woerdenbag HJ, Moskal TA, Pras N, Malingré TM, el-Feraly FS, Kampinga HH, et al. Cytotoxicity of artemisinin-related endoperoxides to Ehrlich ascites tumor cells. J Nat Prod. 1993;56:849–56.
    1. 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.
    1. Ashton M, Nguyen DS, Nguyen VH, Gordi T, Trinh NH, Dinh XH, et al. Artemisinin kinetics and dynamics during oral and rectal treatment of uncomplicated malaria. Clin Pharmacol Ther. 1998;63:482–93.
    1. Li Q, Weina PJ, Milhous WK. Pharmacokinetic and pharmacodynamic profiles of rapid-acting artemisinins in the antimalarial therapy. Curr Drug Ther. 2007;2:210–23.
    1. Vlahopoulos S, Critselis E, Voutsas IF, Perez SA, Moschovi M, Baxevanis CN, et al. New use for old drugs? Prospective targets of chloroquines in cancer therapy. Curr Drug Targets. 2014;15:843–51.
    1. Abdel-Aziz AK, Shouman S, El-Demerdash E, Elgendy M, Abdel-Naim AB. Chloroquine synergizes sunitinib cytotoxicity via modulating autophagic, apoptotic and angiogenic machineries. Chem Biol Interact. 2014;217:28–40.
    1. Meshnick SR, Thomas A, Ranz A, Xu CM, Pan HZ. Artemisinin (qinghaosu): The role of intracellular hemin in its mechanism of antimalarial action. Mol Biochem Parasitol. 1991;49:181–9.
    1. Olliaro PL, Haynes RK, Meunier B, Yuthavong Y. Possible modes of action of the artemisinin-type compounds. Trends Parasitol. 2001;17:122–6.
    1. Beekman AC, Wierenga PK, Woerdenbag HJ, Van Uden W, Pras N, Konings AW, et al. Artemisinin-derived sesquiterpene lactones as potential antitumour compounds: Cytotoxic action against bone marrow and tumour cells. Planta Med. 1998;64:615–9.
    1. Zhang S, Gerhard GS. Heme mediates cytotoxicity from artemisinin and serves as a general anti-proliferation target. PLoS One. 2009;4:e7472.
    1. Berger TG, Dieckmann D, Efferth T, Schultz ES, Funk JO, Baur A, et al. Artesunate in the treatment of metastatic uveal melanoma – First experiences. Oncol Rep. 2005;14:1599–603.
    1. Hamacher-Brady A, Stein HA, Turschner S, Toegel I, Mora R, Jennewein N, et al. Artesunate activates mitochondrial apoptosis in breast cancer cells via iron-catalyzed lysosomal reactive oxygen species production. J Biol Chem. 2011;286:6587–601.
    1. Lai H, Singh NP. Selective cancer cell cytotoxicity from exposure to dihydroartemisinin and holotransferrin. Cancer Lett. 1995;91:41–6.
    1. Lu JJ, Meng LH, Shankavaram UT, Zhu CH, Tong LJ, Chen G, et al. Dihydroartemisinin accelerates c-MYC oncoprotein degradation and induces apoptosis in c-MYC-overexpressing tumor cells. Biochem Pharmacol. 2010;80:22–30.
    1. Lu JJ, Chen SM, Zhang XW, Ding J, Meng LH. 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.
    1. Mercer AE, Copple IM, Maggs JL, O’Neill PM, Park BK. The role of heme and the mitochondrion in the chemical and molecular mechanisms of mammalian cell death induced by the artemisinin antimalarials. J Biol Chem. 2011;286:987–96.
    1. Lai H, Nakase I, Lacoste E, Singh NP, Sasaki T. Artemisinin-transferrin conjugate retards growth of breast tumors in the rat. Anticancer Res. 2009;29:3807–10.
    1. Nakase I, Lai H, Singh NP, Sasaki T. Anticancer properties of artemisinin derivatives and their targeted delivery by transferrin conjugation. Int J Pharm. 2008;354:28–33.
    1. Lai H, Sasaki T, Singh NP. Targeted treatment of cancer with artemisinin and artemisinin-tagged iron-carrying compounds. Expert Opin Ther Targets. 2005;9:995–1007.
    1. Stockwin LH, Han B, Yu SX, Hollingshead MG, ElSohly MA, Gul W, et al. Artemisinin dimer anticancer activity correlates with heme-catalyzed reactive oxygen species generation and endoplasmic reticulum stress induction. Int J Cancer. 2009;125:1266–75.
    1. Huang XJ, Ma ZQ, Zhang WP, Lu YB, Wei EQ. Dihydroartemisinin exerts cytotoxic effects and inhibits hypoxia inducible factor-1alpha activation in C6 glioma cells. J Pharm Pharmacol. 2007;59:849–56.
    1. Kwok JC, Richardson DR. The iron metabolism of neoplastic cells: Alterations that facilitate proliferation? Crit Rev Oncol Hematol. 2002;42:65–78.
    1. Efferth T, Benakis A, Romero MR, Tomicic M, Rauh R, Steinbach D, et al. Enhancement of cytotoxicity of artemisinins toward cancer cells by ferrous iron. Free Radic Biol Med. 2004;37:998–1009.
    1. May WS, Jr, Cuatrecasas P. Transferrin receptor: Its biological significance. J Membr Biol. 1985;88:205–15.
    1. Bostwick DG, Alexander EE, Singh R, Shan A, Qian J, Santella RM, et al. Antioxidant enzyme expression and reactive oxygen species damage in prostatic intraepithelial neoplasia and cancer. Cancer. 2000;89:123–34.
    1. Mercer AE, Maggs JL, Sun XM, Cohen GM, Chadwick J, O’Neill PM, et al. Evidence for the involvement of carbon-centered radicals in the induction of apoptotic cell death by artemisinin compounds. J Biol Chem. 2007;282:9372–82.
    1. Zhou C, Pan W, Wang XP, Chen TS. Artesunate induces apoptosis via a Bak-mediated caspase-independent intrinsic pathway in human lung adenocarcinoma cells. J Cell Physiol. 2012;227:3778–86.
    1. Hansen JB, Fisker N, Westergaard M, Kjaerulff LS, Hansen HF, Thrue CA, et al. SPC3042: A proapoptotic survivin inhibitor. Mol Cancer Ther. 2008;7:2736–45.
    1. Efferth T. Willmar Schwabe Award 2006: Antiplasmodial and antitumor activity of artemisinin – From bench to bedside. Planta Med. 2007;73:299–309.
    1. Riganti C, Doublier S, Viarisio D, Miraglia E, Pescarmona G, Ghigo D, et al. Artemisinin induces doxorubicin resistance in human colon cancer cells via calcium-dependent activation of HIF-1alpha and P-glycoprotein overexpression. Br J Pharmacol. 2009;156:1054–66.
    1. Posner GH, Paik IH, Sur S, McRiner AJ, Borstnik K, Xie S, et al. Orally active, antimalarial, anticancer, artemisinin-derived trioxane dimers with high stability and efficacy. J Med Chem. 2003;46:1060–5.
    1. Lombard MC, N’Da DD, Breytenbach JC, Kolesnikova NI, Tran Van Ba C, Wein S, et al. Antimalarial and anticancer activities of artemisinin-quinoline hybrid-dimers and pharmacokinetic properties in mice. Eur J Pharm Sci. 2012;47:834–41.
    1. Anfosso L, Efferth T, Albini A, Pfeffer U. Microarray expression profiles of angiogenesis-related genes predict tumor cell response to artemisinins. Pharmacogenomics J. 2006;6:269–78.
    1. Efferth T, Sauerbrey A, Olbrich A, Gebhart E, Rauch P, Weber HO, et al. Molecular modes of action of artesunate in tumor cell lines. Mol Pharmacol. 2003;64:382–94.
    1. Lu YY, Chen TS, Qu JL, Pan WL, Sun L, Wei XB. Dihydroartemisinin (DHA) induces caspase-3-dependent apoptosis in human lung adenocarcinoma ASTC-a-1 cells. J Biomed Sci. 2009;16:16.
    1. Gravett AM, Liu WM, Krishna S, Chan WC, Haynes RK, Wilson NL, et al. In vitro study of the anti-cancer effects of artemisone alone or in combination with other chemotherapeutic agents. Cancer Chemother Pharmacol. 2011;67:569–77.
    1. Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med. 2004;10:789–99.
    1. Yao L, Xie H, Jin QY, Hu WL, Chen LJ. Analyzing anti-cancer action mechanisms of dihydroartemisinin using gene chip. Zhongguo Zhong Yao Za Zhi. 2008;33:1583–6.
    1. Jiang Z, Chai J, Chuang HH, Li S, Wang T, Cheng Y, et al. Artesunate induces G0/G1 cell cycle arrest and iron-mediated mitochondrial apoptosis in A431 human epidermoid carcinoma cells. Anticancer Drugs. 2012;23:606–13.
    1. Firestone GL, Sundar SN. Anticancer activities of artemisinin and its bioactive derivatives. Expert Rev Mol Med. 2009;11:e32.
    1. Youns M, Efferth T, Reichling J, Fellenberg K, Bauer A, Hoheisel JD. Gene expression profiling identifies novel key players involved in the cytotoxic effect of Artesunate on pancreatic cancer cells. Biochem Pharmacol. 2009;78:273–83.
    1. Elmore S. Apoptosis: A review of programmed cell death. Toxicol Pathol. 2007;35:495–516.
    1. Karnak D, Xu L. Chemosensitization of prostate cancer by modulating Bcl-2 family proteins. Curr Drug Targets. 2010;11:699–707.
    1. Lu YY, Chen TS, Wang XP, Li L. Single-cell analysis of dihydroartemisinin-induced apoptosis through reactive oxygen species-mediated caspase-8 activation and mitochondrial pathway in ASTC-a-1 cells using fluorescence imaging techniques. J Biomed Opt. 2010;15:046028.
    1. Aung W, Sogawa C, Furukawa T, Saga T. Anticancer effect of dihydroartemisinin (DHA) in a pancreatic tumor model evaluated by conventional methods and optical imaging. Anticancer Res. 2011;31:1549–58.
    1. Lijuan W. Effect of artesunate on human endometrial carcinoma. J Med Coll PLA. 2010;25:143–51.
    1. Efferth T, Giaisi M, Merling A, Krammer PH, Li-Weber M. Artesunate induces ROS-mediated apoptosis in doxorubicin-resistant T leukemia cells. PLoS One. 2007;1(2):e693.
    1. Hou J, Wang D, Zhang R, Wang H. 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.
    1. He Q, Shi J, Shen XL, An J, Sun H, Wang L, 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.
    1. Buommino E, Baroni A, Canozo N, Petrazzuolo M, Nicoletti R, Vozza A, 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;27:412–8.
    1. Hwang YP, Yun HJ, Kim HG, Han EH, Lee GW, Jeong 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.
    1. Wang J, Zhang B, Guo Y, Li G, Xie Q, Zhu B, et al. Artemisinin inhibits tumor lymphangiogenesis by suppression of vascular endothelial growth factor C. Pharmacology. 2008;82:148–55.
    1. Steeg PS. Tumor metastasis: Mechanistic insights and clinical challenges. Nat Med. 2006;12:895–904.
    1. Hur E, Kim HH, Choi SM, Kim JH, Yim S, Kwon HJ, et al. Reduction of hypoxia-induced transcription through the repression of hypoxia-inducible factor-1alpha/aryl hydrocarbon receptor nuclear translocator DNA binding by the 90-kDa heat-shock protein inhibitor radicicol. Mol Pharmacol. 2002;62:975–82.
    1. Zhou HJ, Wang WQ, Wu GD, Lee J, Li A. Artesunate inhibits angiogenesis and downregulates vascular endothelial growth factor expression in chronic myeloid leukemia K562 cells. Vascul Pharmacol. 2007;47:131–8.
    1. Baldwin AS. Regulation of cell death and autophagy by IKK and NF-κB: Critical mechanisms in immune function and cancer. Immunol Rev. 2012;246:327–45.
    1. Fan S, Meng Q, Laterra JJ, Rosen EM. Ras effector pathways modulate scatter factor-stimulated NF-kappaB signaling and protection against DNA damage. Oncogene. 2007;26:4774–96.
    1. Li S, Xue F, Cheng Z, Yang X, Wang S, Geng F, 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.
    1. Wang SJ, Gao Y, Chen H, Kong R, Jiang HC, Pan SH, 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.
    1. Zhou HJ, Zhang JL, Li A, Wang Z, Lou XE. Dihydroartemisinin improves the efficiency of chemotherapeutics in lung carcinomas in vivo and inhibits murine Lewis lung carcinoma cell line growth in vitro. Cancer Chemother Pharmacol. 2010;66:21–9.
    1. O’Connor R. The pharmacology of cancer resistance. Anticancer Res. 2007;27:1267–72.
    1. Michaelis M, Kleinschmidt MC, Barth S, Rothweiler F, Geiler J, Breitling R, et al. Anti-cancer effects of artesunate in a panel of chemoresistant neuroblastoma cell lines. Biochem Pharmacol. 2010;79:130–6.
    1. Chaijaroenkul W, Viyanant V, Mahavorasirikul W, Na-Bangchang K. Cytotoxic activity of artemisinin derivatives against cholangiocarcinoma (CL-6) and hepatocarcinoma (Hep-G2) cell lines. Asian Pac J Cancer Prev. 2011;12:55–9.
    1. Crespo-Ortiz MP, Wei MQ. Antitumor activity of artemisinin and its derivatives: From a well-known antimalarial agent to a potential anticancer drug. J Biomed Biotechnol 2012. 2012 247597.
    1. Lambrou GI, Papadimitriou L, Chrousos GP, Vlahopoulos SA. Glucocorticoid and proteasome inhibitor impact on the leukemic lymphoblast: Multiple, diverse signals converging on a few key downstream regulators. Mol Cell Endocrinol. 2012;351:142–51.
    1. Singh NP, Lai HC. Synergistic cytotoxicity of artemisinin and sodium butyrate on human cancer cells. Anticancer Res. 2005;25:4325–31.
    1. Huang XJ, Li CT, Zhang WP, Lu YB, Fang SH, Wei EQ. Dihydroartemisinin potentiates the cytotoxic effect of temozolomide in rat C6 glioma cells. Pharmacology. 2008;82:1–9.
    1. Kim SJ, Kim MS, Lee JW, Lee CH, Yoo H, Shin SH, et al. Dihydroartemisinin enhances radiosensitivity of human glioma cells in vitro. J Cancer Res Clin Oncol. 2006;132:129–35.
    1. Ohgami Y, Elstad CA, Chung E, Shirachi DY, Quock RM, Lai HC. Effect of hyperbaric oxygen on the anticancer effect of artemisinin on molt-4 human leukemia cells. Anticancer Res. 2010;30:4467–70.
    1. Smith Gueye C, Newby G, Hwang J, Phillips AA, Whittaker M, MacArthur JR, et al. The challenge of artemisinin resistance can only be met by eliminating Plasmodium falciparum malaria across the Greater Mekong subregion. Malar J. 2014;13:286.
    1. Witkowski B, Lelièvre J, Barragán MJ, Laurent V, Su XZ, Berry A, et al. Increased tolerance to artemisinin in Plasmodium falciparum is mediated by a quiescence mechanism. Antimicrob Agents Chemother. 2010;54:1872–7.
    1. Hai-Ying Dong, Zhi-Fei Wang. Antitumor effects of artesunate on human breast carcinoma MCF-7 cells and IGF-IR expression in nude mice xenografts. Chin J Cancer Res. 2014 Apr;26:200–207.
    1. Bachmeier B, Fichtner I, Killian PH, Kronski E, Pfeffer U, Efferth T. Development of resistance towards artesunate in MDA-MB-231 human breast cancer cells. PLoS One. 2011;6:e20550.
    1. Si Y, Park J, Lai HC, Singh M, Sasaki T, Singh NP. Development of a dihydroartemisinin-resistant Molt-4 leukemia cell line. Anticancer Res. 2014;34:2807–10.
    1. Li Q, Xie L, Bennett K, Weina PJ, Mog S, et al. Neurotoxicity and toxicokinetics of artelinic acid following repeated oral administration in rats. Int J Toxicol. 2007;26:401–10.
    1. Schmuck G, Roehrdanz E, Haynes RK, Kahl R. Neurotoxic mode of action of artemisinin. Antimicrob Agents Chemother. 2002;46:821–7.
    1. Singh NP, Verma KB. Case report of a laryngeal squamous cell carcinoma treated with artesunate. Arch Oncol. 2002;10:279–80.
    1. Singh NP, Panwar VK. Case report of a pituitary macroadenoma treated with artemether. Integr Cancer Ther. 2006;5:391–4.
    1. Zhang ZY, Yu SQ, Miao LY, Huang XY, Zhang XP, Zhu YP, et al. Artesunate combined with vinorelbine plus cisplatin in treatment of advanced non-small cell lung cancer: A randomized controlled trial. Zhong Xi Yi Jie He Xue Bao. 2008;6:134–8.
    1. Charman SA, Arbe-Barnes S, Bathurst IC, Brun R, Campbell M, Charman WN, et al. Synthetic ozonide drug candidate OZ439 offers new hope for a single-dose cure of uncomplicated malaria. Proc Natl Acad Sci U S A. 2011;108:4400–5.
    1. Jeyadevan JP, Bray PG, Chadwick J, Mercer AE, Byrne A, Ward SA, et al. Antimalarial and antitumor evaluation of novel C-10 non-acetal dimers of 10beta-(2-hydroxyethyl) deoxoartemisinin. J Med Chem. 2004;47:1290–8.
    1. Opsenica I, Opsenica D, Smith KS, Milhous WK, Solaja BA. Chemical stability of the peroxide bond enables diversified synthesis of potent tetraoxane antimalarials. J Med Chem. 2008;51:2261–6.
    1. Terzic N, Opsenica D, Milic D, Tinant B, Smith KS, Milhous WK, et al. Deoxycholic acid-derived tetraoxane antimalarials and antiproliferatives(1) J Med Chem. 2007;50:5118–27.
    1. Efferth T, Olbrich A, Bauer R. mRNA expression profiles for the response of human tumor cell lines to the antimalarial drugs artesunate, arteether, and artemether. Biochem Pharmacol. 2002;64:617–23.
    1. Cavallo F, De Giovanni C, Nanni P, Forni G, Lollini PL. 2011: The immune hallmarks of cancer. Cancer Immunol Immunother. 2011;60:319–26.
    1. Ho WE, Peh HY, Chan TK, Wong WS. Artemisinins: Pharmacological actions beyond anti-malarial. Pharmacol Ther. 2014;142:126–39.
    1. Zhao F, Wang H, Kunda P, Chen X, Liu QL, Liu T. Artesunate exerts specific cytotoxicity in retinoblastoma cells via CD71. Oncol Rep. 2013;30:1473–82.
    1. Rutteman GR, Erich SA, Mol JA, Spee B, Grinwis GC, Fleckenstein L, et al. Safety and efficacy field study of artesunate for dogs with non-resectable tumours. Anticancer Res. 2013;33:1819–27.

Source: PubMed

3
Iratkozz fel