Browse > Article
http://dx.doi.org/10.7314/APJCP.2012.13.10.4855

Snake Venom: A Potent Anticancer Agent  

Jain, Deepika (IGNOU-I2IT Centre of Excellence for Advanced Education and Research)
Kumar, Sudhir (IGNOU-I2IT Centre of Excellence for Advanced Education and Research)
Publication Information
Asian Pacific Journal of Cancer Prevention / v.13, no.10, 2012 , pp. 4855-4860 More about this Journal
Abstract
Since cancer is one of the leading causes of death worldwide, and there is an urgent need to find better treatment. In recent years remarkable progress has been made towards the understanding of proposed hallmarks of cancer development and treatment. Treatment modalities comprise radiation therapy, surgery, chemotherapy, immunotherapy and hormonal therapy. Currently, the use of chemotherapeutics remains the predominant option for clinical control. However, one of the major problems with successful cancer therapy using chemotherapeutics is that patients often do not respond or eventually develop resistance after initial treatment. This has led to the increased use of anticancer drugs developed from natural resources. The biodiversity of venoms and toxins makes them a unique source from which novel therapeutics may be developed. In this review, the anticancer potential of snake venom is discussed. Some of the included molecules are under clinical trial and may find application for anticancer drug development in the near future.
Keywords
Radiation therapy; chemotherapy; immunotherapy; hormonal therapy; venoms; toxins;
Citations & Related Records
Times Cited By KSCI : 2  (Citation Analysis)
연도 인용수 순위
1 AAl-Sadoon MK, Abdel-Maksoud MA, Rabah DM, Badr G (2012). Induction of apoptosis and growth arrest in human breast carcinoma cells by a snake (walterinnesia aegyptia ) venom combined with silica nanoparticles: crosstalk between Bcl2 and Caspase 3. Cell Physiol Biochem, 30, 653-65.   DOI
2 Arimura T, Niwa K, Mitani N, et al (1989). A resected case of triple cancer in the uterus, lung and thyroid. Zasshi J: Nihon Kyobu Geka Gakkai, 37, 1233-7.
3 Armugam A, Cher CD, Lim K, et al (2009). A secretory phospholipase A2-mediated neuroprotection and antiapoptosis. BMC Neuroscience, 10, 120.   DOI
4 Barnett GC, West CM, Dunning AM, et al (2009). Normal tissue reactions to radiotherapy: towards tailoring treatment dose by genotype. Nature Rev Cancer, 9, 134-42.   DOI
5 Baskar R, Lee KA, Yeo R, Yeoh KW (2012). Cancer and radiation therapy: current advances and future directions. Int J Med Sci, 9, 193-9.   DOI
6 Bazaa A, Pasquier E, Defilles C, et al (2010). MVL-PLA2, a snake venom phospholipase A2, inhibits angiogenesis through an increase in microtubule dynamics and disorganization of focal adhesions. PloS One, 5, e10124.   DOI
7 Berzofsky JA, Terabe M, Wood LV (2012). Strategies to use immune modulators in therapeutic vaccines against cancer. Seminars Oncol, 39, 348-57.   DOI   ScienceOn
8 Bradbury MW, Deane R (1993). Permeability of the blood-brain barrier to lead. Neurotoxicology, 14, 131-6.
9 Chien CM, Yang SH, Chang LS, Lin SR (2008). Involvement of both endoplasmic reticulum- and mitochondria-dependent pathways in cardiotoxin III-induced apoptosis in HL-60 cells. Clin and Experimental Pharmacology and Physiology, 35, 1059-64.   DOI
10 Chien CM, Yang SH, Yang CC, et al (2008). Cardiotoxin III induces c-jun N-terminal kinase-dependent apoptosis in HL-60 human leukaemia cells. Cell Biochemistry and Function, 26, 111-8.   DOI
11 Chiu CC, Lin KL, Chien CM, et al (2009). Effects of cardiotoxin III on NF-kappaB function, proliferation, and apoptosis in human breast MCF-7 cancer cells. Oncol Res, 17, 311-21.   DOI
12 Cohen O, Kronman C, Chitlaru T, et al (2001). Effect of chemical modification of recombinant human acetylcholinesterase by polyethylene glycol on its circulatory longevity. The Biochem J, 357, 795-802.   DOI
13 Escalante T, Ortiz N, Rucavado A, et al (2011). Role of collagens and perlecan in microvascular stability: exploring the mechanism of capillary vessel damage by snake venom metalloproteinases. PloS one, 6, 28017.   DOI
14 Costa LA, Miles HA, Diez RA, et al (1997). Phase I study of VRCTC-310, a purified phospholipase A2 purified from snake venom, in patients with refractory cancer: safety and pharmacokinetic data. Anti-Cancer Drugs, 8, 829-34.   DOI
15 Cura JE, Blanzaco DP, Brisson C, et al (2002). Phase I and pharmacokinetics study of crotoxin (Cytotoxic PLA2, NSC-624244) in Patients with advanced cancer. Clin Cancer Res, 8, 1033-41.
16 Debnath A, Chatterjee U, Das M, et al (2007). Venom of Indian monocellate cobra and Russell's viper show anticancer activity in experimental models. J Ethnopharmacology, 111, 681-4.   DOI
17 Faure G, Harvey AL, Thomson E, et al (1993). Comparison of crotoxin isoforms reveals that stability of the complex plays a major role in its pharmacological action. Eur J Biochem, 214, 491-6.   DOI
18 Ferrer (2001). Snake venom: The pain and potential of the venom. The cold blooded news, 28, PAGE?.
19 Gao W, Starkov VG, Tsetlin VI, et al (2005). Isolation and preliminary crystallographic studies of two new phospholipases A2 from vipera nikolskii venom. Acta crystallographica Ssection F, Structural Biol and Crystallization Commun, 61, 189-92.   DOI
20 Geissler M, Weth R (2002). (Immunotherapy: new insights). Praxis, 91, 2236-46.   DOI
21 Gomes A, Bhattacharjee P, Mishra R, et al (2010). Anticancer potential of animal venoms and toxins. Indian J Exp Biol, 48, 93-103.
22 Kang TS, Georgieva D, Genov N, et al (2011). Enzymatic toxins from snake venom: structural characterization and mechanism of catalysis. FEBS J, 278, 4544-76.   DOI
23 Gomes A, Choudhury SR, Saha A, et al (2007). A heat stable protein toxin (drCT-I) from the Indian Viper (Daboia russelli russelli) venom having antiproliferative, cytotoxic and apoptotic activities. Toxicon: Official J Int Society on Toxinology, 49, 46-56.   DOI   ScienceOn
24 Hammerstrom AE, Cauley DH, Atkinson BJ, Sharma P (2011). Cancer immunotherapy: sipuleucel-T and beyond. Pharmacotherapy, 31, 813-28.   DOI
25 Kang IC, Lee YD, Kim DS (1999). A novel disintegrin salmosin inhibits tumor angiogenesis. Cancer Res, 59, 3754-60.
26 Kemparaju K, Girish KS (2006). Snake venom hyaluronidase: a therapeutic target. Cell Biochem Function, 24, 7-12.   DOI
27 Kruger C, Greten TF, Korangy F (2007). Immune based therapies in cancer. Histology Histopathology, 22, 687-96.
28 Lai D, Visser-Grieve S, Yang X (2012). Tumour suppressor genes in chemotherapeutic drug response. Biosci Reports, 32, 361-74.   DOI   ScienceOn
29 Lin KL, Su JC, Chien CM, et al (2010). Down-regulation of the JAK2/PI3K-mediated signaling activation is involved in Taiwan cobra cardiotoxin III-induced apoptosis of human breast MDA-MB-231 cancer cells. Toxicon : Official J Int Society on Toxinology, 55, 1263-73.   DOI
30 Lokeshwar VB, Selzer MG (2008). Hyalurondiase: both a tumor promoter and suppressor. Seminars in Cancer Biol, 18, 281-7.   DOI   ScienceOn
31 Markland FS, Shieh K, Zhou Q, et al (2001). A novel snake venom disintegrin that inhibits human ovarian cancer dissemination and angiogenesis in an orthotopic nude mouse model. Haemostasis, 31, 183-91.
32 Pawelek PD, Cheah J, Coulombe R, et al (2000). The structure of L-amino acid oxidase reveals the substrate trajectory into an enantiomerically conserved active site. The EMBO J, 19, 4204-15.   DOI
33 Newman RA, Vidal JC, Viskatis LJ, et al (1993). VRCTC-310-a novel compound of purified animal toxins separates antitumor efficacy from neurotoxicity. Invest New Drugs, 11, 151-9.   DOI
34 Orentas RJ, Lee DW, Mackall C (2012). Immunotherapy targets in pediatric cancer. Frontiers in Oncol, 2, 3.
35 Panfoli I, Calzia D, Ravera S, Morelli A (2010). Inhibition of hemorragic snake venom components: old and new approaches. Toxins, 2, 417-27.   DOI
36 Rodrigues RS, Izidoro LF, de Oliveira RJ, et al (2009). Snake venom phospholipases A2: a new class of antitumor agents. Protein and Peptide Letters, 16, 894-8.   DOI
37 Rudd CJ, Viskatis LJ, Vidal JC, Etcheverry MA (1994). In vitro comparison of cytotoxic effects of crotoxin against three human tumors and a normal human epidermal keratinocyte cell line. Invest New Drugs, 12, 183-4.   DOI
38 Siegel R, Naishadham D, Jemal A (2012). Cancer statistics, 2012. CA: A Cancer J for Clin, 62, 10-29.   DOI   ScienceOn
39 Song JK, Jo MR, Park MH, et al (2012). Cell growth inhibition and induction of apoptosis by snake venom toxin in ovarian cancer cell via inactivation of nuclear factor kappaB and signal transducer and activator of transcription 3. Archives of Pharmacal Res, 35, 867-76.   DOI
40 Su JC, Lin KL, Chien CM, et al (2010). Concomitant inactivation of the epidermal growth factor receptor, phosphatidylinositol 3-kinase/Akt and Janus tyrosine kinase 2/signal transducer and activator of transcription 3 signalling pathways in cardiotoxin III-treated A549 cells. Clin and Experimental Pharmacology and Physiology, 37, 833-40.
41 Yang SH, Chien CM, Lu MC, et al (2006). Up-regulation of Bax and endonuclease G, and down-modulation of Bcl-XL involved in cardiotoxin III-induced apoptosis in K562 cells. Experimental & Molecular Med, 38, 435-44.   DOI
42 Tang N, Xie Q, Wang X, et al (2011). Inhibition of invasion and metastasis of MHCC97H cells by expression of snake venom cystatin through reduction of proteinases activity and epithelial-mesenchymal transition. Archives of Pharmacal Res, 34, 781-9.   과학기술학회마을   DOI
43 Wei JF, Wei XL, Mo YZ, He SH (2009). Induction of mast cell accumulation, histamine release and skin edema by N49 phospholipase A2. BMC Immunol 10, 21.   DOI
44 Yang SH, Chien CM, Chang LS, Lin SR (2008). Cardiotoxin III-induced apoptosis is mediated by Ca2+-dependent caspase-12 activation in K562 cells. J Biochem And Molecular Toxicology, 22, 209-18.   DOI
45 Yang SH, Chien CM, Lu MC, et al (2005). Cardiotoxin III induces apoptosis in K562 cells through a mitochondrial-mediated pathway. Clin and Experimental Pharmacology & Physiology, 32, 515-20.   DOI