Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 6/2016

01-12-2016 | Review Article

Application of bee venom and its main constituent melittin for cancer treatment

Authors: Cui-cui Liu, Ding-jun Hao, Qian Zhang, Jing An, Jing-jing Zhao, Bo Chen, Ling-ling Zhang, Hao Yang

Published in: Cancer Chemotherapy and Pharmacology | Issue 6/2016

Login to get access

Abstract

Bee venom and its main constituent melittin (MEL) have been extensively studied in the treatment of tumors. However, the non-specific cytotoxicity and hemolytic activity have hampered the clinical application. Currently, a number of research groups have reported a series of optimization strategies, including gene therapy, recombinant immunotoxin incorporating MEL or MEL nanoparticles, targeting tumor cells to attenuate the cytotoxicity and improve its antitumor efficiency and therapeutic capabilities, which have shown very promising in overcoming some of these obstacles. In this review, we summarize the current knowledge regarding anticancer effects of bee venom and its main compound MEL on different kinds of tumor cells as well as elucidate their possible anticancer mechanisms. It could be concluded that MEL exerts multiple effects on cellular functions of cancerous cells such as proliferation, apoptosis, metastasis, angiogenesis as well as cell cycle, and the anticancer processes involve diverse signal molecules and regulatory pathways. We also highlight the recent research progress for efficient delivery of MEL peptide, thus providing new ideas and hopeful strategies for the in vivo application of MEL.
Literature
1.
go back to reference Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F (2015) Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136:E359–E386CrossRefPubMed Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F (2015) Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 136:E359–E386CrossRefPubMed
2.
go back to reference Zugazagoitia J, Guedes C, Ponce S, Ferrer I, Molina-Pinelo S, Paz-Ares L (2016) Current challenges in cancer treatment. Clin Ther Zugazagoitia J, Guedes C, Ponce S, Ferrer I, Molina-Pinelo S, Paz-Ares L (2016) Current challenges in cancer treatment. Clin Ther
4.
go back to reference Al-Sadoon MK, Rabah DM, Badr G (2013) Enhanced anticancer efficacy of snake venom combined with silica nanoparticles in a murine model of human multiple myeloma: molecular targets for cell cycle arrest and apoptosis induction. Cell Immunol 284:129–138CrossRefPubMed Al-Sadoon MK, Rabah DM, Badr G (2013) Enhanced anticancer efficacy of snake venom combined with silica nanoparticles in a murine model of human multiple myeloma: molecular targets for cell cycle arrest and apoptosis induction. Cell Immunol 284:129–138CrossRefPubMed
6.
go back to reference Diaz-Garcia A, Morier-Diaz L, Frion-Herrera Y, Rodriguez-Sanchez H, Caballero-Lorenzo Y, Mendoza-Llanes D, Riquenes-Garlobo Y, Fraga-Castro JA (2013) In vitro anticancer effect of venom from Cuban scorpion Rhopalurus junceus against a panel of human cancer cell lines. J Venom Res 4:5–12PubMedPubMedCentral Diaz-Garcia A, Morier-Diaz L, Frion-Herrera Y, Rodriguez-Sanchez H, Caballero-Lorenzo Y, Mendoza-Llanes D, Riquenes-Garlobo Y, Fraga-Castro JA (2013) In vitro anticancer effect of venom from Cuban scorpion Rhopalurus junceus against a panel of human cancer cell lines. J Venom Res 4:5–12PubMedPubMedCentral
7.
go back to reference Soletti RC, de Faria GP, Vernal J, Terenzi H, Anderluh G, Borges HL, Moura-Neto V, Gabilan NH (2008) Potentiation of anticancer-drug cytotoxicity by sea anemone pore-forming proteins in human glioblastoma cells. Anticancer Drugs 19:517–525CrossRefPubMed Soletti RC, de Faria GP, Vernal J, Terenzi H, Anderluh G, Borges HL, Moura-Neto V, Gabilan NH (2008) Potentiation of anticancer-drug cytotoxicity by sea anemone pore-forming proteins in human glioblastoma cells. Anticancer Drugs 19:517–525CrossRefPubMed
8.
go back to reference Huang T, Gong WH, Li XC, Zou CP, Jiang GJ, Li XH, Qian H (2012) Efficient killing effect of osteosarcoma cells by cinobufacini and cisplatin in combination. Asian Pac J Cancer Prev 13:2847–2851CrossRefPubMed Huang T, Gong WH, Li XC, Zou CP, Jiang GJ, Li XH, Qian H (2012) Efficient killing effect of osteosarcoma cells by cinobufacini and cisplatin in combination. Asian Pac J Cancer Prev 13:2847–2851CrossRefPubMed
9.
go back to reference Liu CC, Yang H, Zhang LL, Zhang Q, Chen B, Wang Y (2014) Biotoxins for cancer therapy. Asian Pac J Cancer Prev 15:4753–4758CrossRefPubMed Liu CC, Yang H, Zhang LL, Zhang Q, Chen B, Wang Y (2014) Biotoxins for cancer therapy. Asian Pac J Cancer Prev 15:4753–4758CrossRefPubMed
10.
go back to reference Gomes A, Bhattacharjee P, Mishra R, Biswas AK, Dasgupta SC, Giri B (2010) Anticancer potential of animal venoms and toxins. Indian J Exp Biol 48:93–103PubMed Gomes A, Bhattacharjee P, Mishra R, Biswas AK, Dasgupta SC, Giri B (2010) Anticancer potential of animal venoms and toxins. Indian J Exp Biol 48:93–103PubMed
11.
go back to reference Orsolic N (2013) Possible molecular targets of bee venom in the treatment of cancer: application and perspectives. Forum Immunopathol Dis Ther 4:275–315CrossRef Orsolic N (2013) Possible molecular targets of bee venom in the treatment of cancer: application and perspectives. Forum Immunopathol Dis Ther 4:275–315CrossRef
12.
go back to reference Moreno M, Giralt E (2015) Three valuable peptides from bee and wasp venoms for therapeutic and biotechnological use: melittin, apamin and mastoparan. Toxins (Basel) 7:1126–1150CrossRef Moreno M, Giralt E (2015) Three valuable peptides from bee and wasp venoms for therapeutic and biotechnological use: melittin, apamin and mastoparan. Toxins (Basel) 7:1126–1150CrossRef
13.
go back to reference Jeong YJ, Shin JM, Bae YS, Cho HJ, Park KK, Choe JY, Han SM, Moon SK, Kim WJ, Choi YH et al (2015) Melittin has a chondroprotective effect by inhibiting MMP-1 and MMP-8 expressions via blocking NF-kappaB and AP-1 signaling pathway in chondrocytes. Int Immunopharmacol 25:400–405CrossRefPubMed Jeong YJ, Shin JM, Bae YS, Cho HJ, Park KK, Choe JY, Han SM, Moon SK, Kim WJ, Choi YH et al (2015) Melittin has a chondroprotective effect by inhibiting MMP-1 and MMP-8 expressions via blocking NF-kappaB and AP-1 signaling pathway in chondrocytes. Int Immunopharmacol 25:400–405CrossRefPubMed
14.
go back to reference Lee JA, Son MJ, Choi J, Jun JH, Kim JI, Lee MS (2014) Bee venom acupuncture for rheumatoid arthritis: a systematic review of randomised clinical trials. BMJ Open 4:e006140CrossRefPubMedPubMedCentral Lee JA, Son MJ, Choi J, Jun JH, Kim JI, Lee MS (2014) Bee venom acupuncture for rheumatoid arthritis: a systematic review of randomised clinical trials. BMJ Open 4:e006140CrossRefPubMedPubMedCentral
15.
go back to reference Son DJ, Lee JW, Lee YH, Song HS, Lee CK, Hong JT (2007) Therapeutic application of anti-arthritis, pain-releasing, and anti-cancer effects of bee venom and its constituent compounds. Pharmacol Ther 115:246–270CrossRefPubMed Son DJ, Lee JW, Lee YH, Song HS, Lee CK, Hong JT (2007) Therapeutic application of anti-arthritis, pain-releasing, and anti-cancer effects of bee venom and its constituent compounds. Pharmacol Ther 115:246–270CrossRefPubMed
16.
go back to reference Hu H, Chen D, Li Y, Zhang X (2006) Effect of polypeptides in bee venom on growth inhibition and apoptosis induction of the human hepatoma cell line SMMC-7721 in vitro and Balb/c nude mice in-vivo. J Pharm Pharmacol 58:83–89CrossRefPubMed Hu H, Chen D, Li Y, Zhang X (2006) Effect of polypeptides in bee venom on growth inhibition and apoptosis induction of the human hepatoma cell line SMMC-7721 in vitro and Balb/c nude mice in-vivo. J Pharm Pharmacol 58:83–89CrossRefPubMed
17.
go back to reference Park MH, Choi MS, Kwak DH, Oh KW, do Yoon Y, Han SB, Song HS, Song MJ, Hong JT (2011) Anti-cancer effect of bee venom in prostate cancer cells through activation of caspase pathway via inactivation of NF-kappaB. Prostate 71:801–812CrossRefPubMed Park MH, Choi MS, Kwak DH, Oh KW, do Yoon Y, Han SB, Song HS, Song MJ, Hong JT (2011) Anti-cancer effect of bee venom in prostate cancer cells through activation of caspase pathway via inactivation of NF-kappaB. Prostate 71:801–812CrossRefPubMed
18.
go back to reference Tu WC, Wu CC, Hsieh HL, Chen CY, Hsu SL (2008) Honeybee venom induces calcium-dependent but caspase-independent apoptotic cell death in human melanoma A2058 cells. Toxicon 52:318–329CrossRefPubMed Tu WC, Wu CC, Hsieh HL, Chen CY, Hsu SL (2008) Honeybee venom induces calcium-dependent but caspase-independent apoptotic cell death in human melanoma A2058 cells. Toxicon 52:318–329CrossRefPubMed
19.
go back to reference Jang MH, Shin MC, Lim S, Han SM, Park HJ, Shin I, Lee JS, Kim KA, Kim EH, Kim CJ et al (2003) Bee venom induces apoptosis and inhibits expression of cyclooxygenase-2 mRNA in human lung cancer cell line NCI-H1299. J Pharmacol Sci 91:95–104CrossRefPubMed Jang MH, Shin MC, Lim S, Han SM, Park HJ, Shin I, Lee JS, Kim KA, Kim EH, Kim CJ et al (2003) Bee venom induces apoptosis and inhibits expression of cyclooxygenase-2 mRNA in human lung cancer cell line NCI-H1299. J Pharmacol Sci 91:95–104CrossRefPubMed
20.
go back to reference Ip SW, Liao SS, Lin SY, Lin JP, Yang JS, Lin ML, Chen GW, Lu HF, Lin MW, Han SM et al (2008) The role of mitochondria in bee venom-induced apoptosis in human breast cancer MCF7 cells. In Vivo 22:237–245PubMed Ip SW, Liao SS, Lin SY, Lin JP, Yang JS, Lin ML, Chen GW, Lu HF, Lin MW, Han SM et al (2008) The role of mitochondria in bee venom-induced apoptosis in human breast cancer MCF7 cells. In Vivo 22:237–245PubMed
21.
go back to reference Jo M, Park MH, Kollipara PS, An BJ, Song HS, Han SB, Kim JH, Song MJ, Hong JT (2012) Anti-cancer effect of bee venom toxin and melittin in ovarian cancer cells through induction of death receptors and inhibition of JAK2/STAT3 pathway. Toxicol Appl Pharmacol 258:72–81CrossRefPubMed Jo M, Park MH, Kollipara PS, An BJ, Song HS, Han SB, Kim JH, Song MJ, Hong JT (2012) Anti-cancer effect of bee venom toxin and melittin in ovarian cancer cells through induction of death receptors and inhibition of JAK2/STAT3 pathway. Toxicol Appl Pharmacol 258:72–81CrossRefPubMed
22.
go back to reference Ip SW, Chu YL, Yu CS, Chen PY, Ho HC, Yang JS, Huang HY, Chueh FS, Lai TY, Chung JG et al (2012) Bee venom induces apoptosis through intracellular Ca2+-modulated intrinsic death pathway in human bladder cancer cells. Int J Urol 19:61–70CrossRefPubMed Ip SW, Chu YL, Yu CS, Chen PY, Ho HC, Yang JS, Huang HY, Chueh FS, Lai TY, Chung JG et al (2012) Bee venom induces apoptosis through intracellular Ca2+-modulated intrinsic death pathway in human bladder cancer cells. Int J Urol 19:61–70CrossRefPubMed
23.
go back to reference Moon DO, Park SY, Heo MS, Kim KC, Park C, Ko WS, Choi YH, Kim GY (2006) Key regulators in bee venom-induced apoptosis are Bcl-2 and caspase-3 in human leukemic U937 cells through downregulation of ERK and Akt. Int Immunopharmacol 6:1796–1807CrossRefPubMed Moon DO, Park SY, Heo MS, Kim KC, Park C, Ko WS, Choi YH, Kim GY (2006) Key regulators in bee venom-induced apoptosis are Bcl-2 and caspase-3 in human leukemic U937 cells through downregulation of ERK and Akt. Int Immunopharmacol 6:1796–1807CrossRefPubMed
24.
go back to reference Choi KE, Hwang CJ, Gu SM, Park MH, Kim JH, Park JH, Ahn YJ, Kim JY, Song MJ, Song HS et al (2014) Cancer cell growth inhibitory effect of bee venom via increase of death receptor 3 expression and inactivation of NF-kappa B in NSCLC cells. Toxins (Basel) 6:2210–2228CrossRef Choi KE, Hwang CJ, Gu SM, Park MH, Kim JH, Park JH, Ahn YJ, Kim JY, Song MJ, Song HS et al (2014) Cancer cell growth inhibitory effect of bee venom via increase of death receptor 3 expression and inactivation of NF-kappa B in NSCLC cells. Toxins (Basel) 6:2210–2228CrossRef
25.
go back to reference Orsolic N, Sver L, Verstovsek S, Terzic S, Basic I (2003) Inhibition of mammary carcinoma cell proliferation in vitro and tumor growth in vivo by bee venom. Toxicon 41:861–870CrossRefPubMed Orsolic N, Sver L, Verstovsek S, Terzic S, Basic I (2003) Inhibition of mammary carcinoma cell proliferation in vitro and tumor growth in vivo by bee venom. Toxicon 41:861–870CrossRefPubMed
26.
go back to reference Liu X, Chen D, Xie L, Zhang R (2002) Effect of honey bee venom on proliferation of K1735M2 mouse melanoma cells in vitro and growth of murine B16 melanomas in-vivo. J Pharm Pharmacol 54:1083–1089CrossRefPubMed Liu X, Chen D, Xie L, Zhang R (2002) Effect of honey bee venom on proliferation of K1735M2 mouse melanoma cells in vitro and growth of murine B16 melanomas in-vivo. J Pharm Pharmacol 54:1083–1089CrossRefPubMed
27.
go back to reference Alizadehnohi M, Nabiuni M, Nazari Z, Safaeinejad Z, Irian S (2012) The synergistic cytotoxic effect of cisplatin and honey bee venom on human ovarian cancer cell line A2780cp. J Venom Res 3:22–27PubMedPubMedCentral Alizadehnohi M, Nabiuni M, Nazari Z, Safaeinejad Z, Irian S (2012) The synergistic cytotoxic effect of cisplatin and honey bee venom on human ovarian cancer cell line A2780cp. J Venom Res 3:22–27PubMedPubMedCentral
28.
go back to reference Kollipara PS, Kim JH, Won D, Lee SM, Sung HC, Chang HS, Lee KT, Lee KS, Park MH, Song MJ et al (2014) Co-culture with NK-92MI cells enhanced the anti-cancer effect of bee venom on NSCLC cells by inactivation of NF-kappaB. Arch Pharm Res 37:379–389CrossRefPubMed Kollipara PS, Kim JH, Won D, Lee SM, Sung HC, Chang HS, Lee KT, Lee KS, Park MH, Song MJ et al (2014) Co-culture with NK-92MI cells enhanced the anti-cancer effect of bee venom on NSCLC cells by inactivation of NF-kappaB. Arch Pharm Res 37:379–389CrossRefPubMed
30.
go back to reference Shi W, Li C, Li M, Zong X, Han D, Chen Y (2016) Antimicrobial peptide melittin against Xanthomonas oryzae pv. oryzae, the bacterial leaf blight pathogen in rice. Appl Microbiol Biotechnol 100:5059–5067CrossRefPubMedPubMedCentral Shi W, Li C, Li M, Zong X, Han D, Chen Y (2016) Antimicrobial peptide melittin against Xanthomonas oryzae pv. oryzae, the bacterial leaf blight pathogen in rice. Appl Microbiol Biotechnol 100:5059–5067CrossRefPubMedPubMedCentral
31.
go back to reference Do N, Weindl G, Grohmann L, Salwiczek M, Koksch B, Korting HC, Schafer-Korting M (2014) Cationic membrane-active peptides—anticancer and antifungal activity as well as penetration into human skin. Exp Dermatol 23:326–331CrossRefPubMed Do N, Weindl G, Grohmann L, Salwiczek M, Koksch B, Korting HC, Schafer-Korting M (2014) Cationic membrane-active peptides—anticancer and antifungal activity as well as penetration into human skin. Exp Dermatol 23:326–331CrossRefPubMed
32.
go back to reference Skalickova S, Heger Z, Krejcova L, Pekarik V, Bastl K, Janda J, Kostolansky F, Vareckova E, Zitka O, Adam V et al (2015) Perspective of use of antiviral peptides against influenza virus. Viruses 7:5428–5442CrossRefPubMedPubMedCentral Skalickova S, Heger Z, Krejcova L, Pekarik V, Bastl K, Janda J, Kostolansky F, Vareckova E, Zitka O, Adam V et al (2015) Perspective of use of antiviral peptides against influenza virus. Viruses 7:5428–5442CrossRefPubMedPubMedCentral
33.
go back to reference Pereira AV, de Barros G, Pinto EG, Tempone AG, Orsi Rde O, Dos Santos LD, Calvi S, Ferreira RS Jr, Pimenta DC, Barraviera B et al (2016) Melittin induces in vitro death of Leishmania (Leishmania) infantum by triggering the cellular innate immune response. J Venom Anim Toxins Incl Trop Dis 22:1CrossRefPubMedPubMedCentral Pereira AV, de Barros G, Pinto EG, Tempone AG, Orsi Rde O, Dos Santos LD, Calvi S, Ferreira RS Jr, Pimenta DC, Barraviera B et al (2016) Melittin induces in vitro death of Leishmania (Leishmania) infantum by triggering the cellular innate immune response. J Venom Anim Toxins Incl Trop Dis 22:1CrossRefPubMedPubMedCentral
34.
go back to reference Adade CM, Oliveira IR, Pais JA, Souto-Padron T (2013) Melittin peptide kills Trypanosoma cruzi parasites by inducing different cell death pathways. Toxicon 69:227–239CrossRefPubMed Adade CM, Oliveira IR, Pais JA, Souto-Padron T (2013) Melittin peptide kills Trypanosoma cruzi parasites by inducing different cell death pathways. Toxicon 69:227–239CrossRefPubMed
35.
go back to reference Gajski G, Garaj-Vrhovac V (2013) Melittin: a lytic peptide with anticancer properties. Environ Toxicol Pharmacol 36:697–705CrossRefPubMed Gajski G, Garaj-Vrhovac V (2013) Melittin: a lytic peptide with anticancer properties. Environ Toxicol Pharmacol 36:697–705CrossRefPubMed
36.
go back to reference Hait WN, Grais L, Benz C, Cadman EC (1985) Inhibition of growth of leukemic cells by inhibitors of calmodulin: phenothiazines and melittin. Cancer Chemother Pharmacol 14:202–205CrossRefPubMed Hait WN, Grais L, Benz C, Cadman EC (1985) Inhibition of growth of leukemic cells by inhibitors of calmodulin: phenothiazines and melittin. Cancer Chemother Pharmacol 14:202–205CrossRefPubMed
37.
go back to reference Jamasbi E, Ciccotosto GD, Tailhades J, Robins-Browne RM, Ugalde CL, Sharples RA, Patil N, Wade JD, Hossain MA, Separovic F et al (2015) Site of fluorescent label modifies interaction of melittin with live cells and model membranes. Biochim Biophys Acta 1848:2031–2039CrossRefPubMed Jamasbi E, Ciccotosto GD, Tailhades J, Robins-Browne RM, Ugalde CL, Sharples RA, Patil N, Wade JD, Hossain MA, Separovic F et al (2015) Site of fluorescent label modifies interaction of melittin with live cells and model membranes. Biochim Biophys Acta 1848:2031–2039CrossRefPubMed
38.
go back to reference Katsu T, Kuroko M, Morikawa T, Sanchika K, Fujita Y, Yamamura H, Uda M (1989) Mechanism of membrane damage induced by the amphipathic peptides gramicidin S and melittin. Biochim Biophys Acta 983:135–141CrossRefPubMed Katsu T, Kuroko M, Morikawa T, Sanchika K, Fujita Y, Yamamura H, Uda M (1989) Mechanism of membrane damage induced by the amphipathic peptides gramicidin S and melittin. Biochim Biophys Acta 983:135–141CrossRefPubMed
39.
go back to reference Lee SY, Park HS, Lee SJ, Choi MU (2001) Melittin exerts multiple effects on the release of free fatty acids from L1210 cells: lack of selective activation of phospholipase A2 by melittin. Arch Biochem Biophys 389:57–67CrossRefPubMed Lee SY, Park HS, Lee SJ, Choi MU (2001) Melittin exerts multiple effects on the release of free fatty acids from L1210 cells: lack of selective activation of phospholipase A2 by melittin. Arch Biochem Biophys 389:57–67CrossRefPubMed
40.
go back to reference Juhaniewicz Joanna, Sek S (2016) Interaction of melittin with negatively charged lipid bilayers supported on gold electrodes. Electrochim Acta 197:336–343CrossRef Juhaniewicz Joanna, Sek S (2016) Interaction of melittin with negatively charged lipid bilayers supported on gold electrodes. Electrochim Acta 197:336–343CrossRef
41.
go back to reference Lee Juneyoung, Lee DG (2014) Melittin triggers apoptosis in Candida albicans through the reactive oxygen species-mediated mitochondria/caspasedependent pathway. FEMS Microbiol Lett 355:36–42CrossRefPubMed Lee Juneyoung, Lee DG (2014) Melittin triggers apoptosis in Candida albicans through the reactive oxygen species-mediated mitochondria/caspasedependent pathway. FEMS Microbiol Lett 355:36–42CrossRefPubMed
42.
go back to reference Jamasbi E, Batinovic S, Sharples RA, Sani MA, Robins-Browne RM, Wade JD, Separovic F, Hossain MA (2014) Melittin peptides exhibit different activity on different cells and model membranes. Amino Acids 46:2759–2766CrossRefPubMed Jamasbi E, Batinovic S, Sharples RA, Sani MA, Robins-Browne RM, Wade JD, Separovic F, Hossain MA (2014) Melittin peptides exhibit different activity on different cells and model membranes. Amino Acids 46:2759–2766CrossRefPubMed
43.
go back to reference Schweizer F (2009) Cationic amphiphilic peptides with cancer-selective toxicity. Eur J Pharmacol 625:190–194CrossRefPubMed Schweizer F (2009) Cationic amphiphilic peptides with cancer-selective toxicity. Eur J Pharmacol 625:190–194CrossRefPubMed
44.
go back to reference Kohno M, Horibe T, Ohara K, Ito S, Kawakami K (2014) The membrane-lytic peptides K8L9 and melittin enter cancer cells via receptor endocytosis following subcytotoxic exposure. Chem Biol 21:1522–1532CrossRefPubMed Kohno M, Horibe T, Ohara K, Ito S, Kawakami K (2014) The membrane-lytic peptides K8L9 and melittin enter cancer cells via receptor endocytosis following subcytotoxic exposure. Chem Biol 21:1522–1532CrossRefPubMed
46.
go back to reference Pistritto G, Trisciuoglio D, Ceci C, Garufi A, D’Orazi G (2016) Apoptosis as anticancer mechanism: function and dysfunction of its modulators and targeted therapeutic strategies. Aging (Albany NY) 8:603–619CrossRef Pistritto G, Trisciuoglio D, Ceci C, Garufi A, D’Orazi G (2016) Apoptosis as anticancer mechanism: function and dysfunction of its modulators and targeted therapeutic strategies. Aging (Albany NY) 8:603–619CrossRef
48.
go back to reference Kondratskyi A, Kondratska K, Skryma R, Prevarskaya N (2015) Ion channels in the regulation of apoptosis. Biochim Biophys Acta 1848:2532–2546CrossRefPubMed Kondratskyi A, Kondratska K, Skryma R, Prevarskaya N (2015) Ion channels in the regulation of apoptosis. Biochim Biophys Acta 1848:2532–2546CrossRefPubMed
49.
go back to reference Hennessy EJ (2016) Selective inhibitors of Bcl-2 and Bcl-xL: balancing antitumor activity with on-target toxicity. Bioorg Med Chem Lett 26:2105–2114CrossRefPubMed Hennessy EJ (2016) Selective inhibitors of Bcl-2 and Bcl-xL: balancing antitumor activity with on-target toxicity. Bioorg Med Chem Lett 26:2105–2114CrossRefPubMed
50.
go back to reference Schultz DR, Harrington WJ Jr (2003) Apoptosis: programmed cell death at a molecular level. Semin Arthritis Rheum 32:345–369CrossRefPubMed Schultz DR, Harrington WJ Jr (2003) Apoptosis: programmed cell death at a molecular level. Semin Arthritis Rheum 32:345–369CrossRefPubMed
51.
go back to reference Kroemer G (2003) Mitochondrial control of apoptosis: an introduction. Biochem Biophys Res Commun 304:433–435CrossRefPubMed Kroemer G (2003) Mitochondrial control of apoptosis: an introduction. Biochem Biophys Res Commun 304:433–435CrossRefPubMed
52.
go back to reference Rossi D, Gaidano G (2003) Messengers of cell death: apoptotic signaling in health and disease. Haematologica 88:212–218PubMed Rossi D, Gaidano G (2003) Messengers of cell death: apoptotic signaling in health and disease. Haematologica 88:212–218PubMed
53.
go back to reference Kiraz Y, Adan A, Kartal Yandim M, Baran Y (2016) Major apoptotic mechanisms and genes involved in apoptosis. Tumour Biol Kiraz Y, Adan A, Kartal Yandim M, Baran Y (2016) Major apoptotic mechanisms and genes involved in apoptosis. Tumour Biol
54.
go back to reference Wang HG, Pathan N, Ethell IM, Krajewski S, Yamaguchi Y, Shibasaki F, McKeon F, Bobo T, Franke TF, Reed JC et al (1999) Ca2+-induced apoptosis through calcineurin dephosphorylation of BAD. Science 284:339–343CrossRefPubMed Wang HG, Pathan N, Ethell IM, Krajewski S, Yamaguchi Y, Shibasaki F, McKeon F, Bobo T, Franke TF, Reed JC et al (1999) Ca2+-induced apoptosis through calcineurin dephosphorylation of BAD. Science 284:339–343CrossRefPubMed
55.
go back to reference Hajnoczky G, Csordas G, Das S, Garcia-Perez C, Saotome M, Sinha Roy S, Yi M (2006) Mitochondrial calcium signalling and cell death: approaches for assessing the role of mitochondrial Ca2+ uptake in apoptosis. Cell Calcium 40:553–560CrossRefPubMedPubMedCentral Hajnoczky G, Csordas G, Das S, Garcia-Perez C, Saotome M, Sinha Roy S, Yi M (2006) Mitochondrial calcium signalling and cell death: approaches for assessing the role of mitochondrial Ca2+ uptake in apoptosis. Cell Calcium 40:553–560CrossRefPubMedPubMedCentral
56.
go back to reference Sharma SV (1993) Melittin-induced hyperactivation of phospholipase A2 activity and calcium influx in ras-transformed cells. Oncogene 8:939–947PubMed Sharma SV (1993) Melittin-induced hyperactivation of phospholipase A2 activity and calcium influx in ras-transformed cells. Oncogene 8:939–947PubMed
57.
go back to reference Saris NE, Carafoli E (2005) A historical review of cellular calcium handling, with emphasis on mitochondria. Biochemistry (Mosc) 70:187–194CrossRef Saris NE, Carafoli E (2005) A historical review of cellular calcium handling, with emphasis on mitochondria. Biochemistry (Mosc) 70:187–194CrossRef
58.
go back to reference Chu ST, Cheng HH, Huang CJ, Chang HC, Chi CC, Su HH, Hsu SS, Wang JL, Chen IS, Liu SI et al (2007) Phospholipase A2-independent Ca2+ entry and subsequent apoptosis induced by melittin in human MG63 osteosarcoma cells. Life Sci 80:364–369CrossRefPubMed Chu ST, Cheng HH, Huang CJ, Chang HC, Chi CC, Su HH, Hsu SS, Wang JL, Chen IS, Liu SI et al (2007) Phospholipase A2-independent Ca2+ entry and subsequent apoptosis induced by melittin in human MG63 osteosarcoma cells. Life Sci 80:364–369CrossRefPubMed
59.
go back to reference Lazo JS, Hait WN, Kennedy KA, Braun ID, Meandzija B (1985) Enhanced bleomycin-induced DNA damage and cytotoxicity with calmodulin antagonists. Mol Pharmacol 27:387–393PubMed Lazo JS, Hait WN, Kennedy KA, Braun ID, Meandzija B (1985) Enhanced bleomycin-induced DNA damage and cytotoxicity with calmodulin antagonists. Mol Pharmacol 27:387–393PubMed
60.
go back to reference Lee GL, Hait WN (1985) Inhibition of growth of C6 astrocytoma cells by inhibitors of calmodulin. Life Sci 36:347–354CrossRefPubMed Lee GL, Hait WN (1985) Inhibition of growth of C6 astrocytoma cells by inhibitors of calmodulin. Life Sci 36:347–354CrossRefPubMed
61.
go back to reference Kong GM, Tao WH, Diao YL, Fang PH, Wang JJ, Bo P, Qian F (2016) Melittin induces human gastric cancer cell apoptosis via activation of mitochondrial pathway. World J Gastroenterol 22:3186–3195CrossRefPubMedPubMedCentral Kong GM, Tao WH, Diao YL, Fang PH, Wang JJ, Bo P, Qian F (2016) Melittin induces human gastric cancer cell apoptosis via activation of mitochondrial pathway. World J Gastroenterol 22:3186–3195CrossRefPubMedPubMedCentral
62.
go back to reference Elrod HA, Sun SY (2008) Modulation of death receptors by cancer therapeutic agents. Cancer Biol Ther 7:163–173CrossRefPubMed Elrod HA, Sun SY (2008) Modulation of death receptors by cancer therapeutic agents. Cancer Biol Ther 7:163–173CrossRefPubMed
63.
go back to reference Wang C, Chen T, Zhang N, Yang M, Li B, Lu X, Cao X, Ling C (2009) Melittin, a major component of bee venom, sensitizes human hepatocellular carcinoma cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis by activating CaMKII-TAK1-JNK/p38 and inhibiting IkappaBalpha kinase-NFkappaB. J Biol Chem 284:3804–3813CrossRefPubMed Wang C, Chen T, Zhang N, Yang M, Li B, Lu X, Cao X, Ling C (2009) Melittin, a major component of bee venom, sensitizes human hepatocellular carcinoma cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis by activating CaMKII-TAK1-JNK/p38 and inhibiting IkappaBalpha kinase-NFkappaB. J Biol Chem 284:3804–3813CrossRefPubMed
64.
go back to reference Fan Q, Hu Y, Pang H, Sun J, Wang Z, Li J (2014) Melittin protein inhibits the proliferation of MG63 cells by activating inositol-requiring protein-1alpha and X-box binding protein 1-mediated apoptosis. Mol Med Rep 9:1365–1370PubMed Fan Q, Hu Y, Pang H, Sun J, Wang Z, Li J (2014) Melittin protein inhibits the proliferation of MG63 cells by activating inositol-requiring protein-1alpha and X-box binding protein 1-mediated apoptosis. Mol Med Rep 9:1365–1370PubMed
65.
go back to reference Moon DO, Park SY, Choi YH, Kim ND, Lee C, Kim GY (2008) Melittin induces Bcl-2 and caspase-3-dependent apoptosis through downregulation of Akt phosphorylation in human leukemic U937 cells. Toxicon 51:112–120CrossRefPubMed Moon DO, Park SY, Choi YH, Kim ND, Lee C, Kim GY (2008) Melittin induces Bcl-2 and caspase-3-dependent apoptosis through downregulation of Akt phosphorylation in human leukemic U937 cells. Toxicon 51:112–120CrossRefPubMed
66.
go back to reference Woodman N, Pinder SE, Tajadura V, Le Bourhis X, Gillett C, Delannoy P, Burchell JM, Julien S (2016) Two E-selectin ligands, BST-2 and LGALS3BP, predict metastasis and poor survival of ER-negative breast cancer. Int J Oncol Woodman N, Pinder SE, Tajadura V, Le Bourhis X, Gillett C, Delannoy P, Burchell JM, Julien S (2016) Two E-selectin ligands, BST-2 and LGALS3BP, predict metastasis and poor survival of ER-negative breast cancer. Int J Oncol
67.
go back to reference Liotta LA (1988) Gene products which play a role in cancer invasion and metastasis. Breast Cancer Res Treat 11:113–124CrossRefPubMed Liotta LA (1988) Gene products which play a role in cancer invasion and metastasis. Breast Cancer Res Treat 11:113–124CrossRefPubMed
68.
go back to reference Liu S, Yu M, He Y, Xiao L, Wang F, Song C, Sun S, Ling C, Xu Z (2008) Melittin prevents liver cancer cell metastasis through inhibition of the Rac1-dependent pathway. Hepatology 47:1964–1973CrossRefPubMed Liu S, Yu M, He Y, Xiao L, Wang F, Song C, Sun S, Ling C, Xu Z (2008) Melittin prevents liver cancer cell metastasis through inhibition of the Rac1-dependent pathway. Hepatology 47:1964–1973CrossRefPubMed
69.
go back to reference Jeong YJ, Cho HJ, Whang K, Lee IS, Park KK, Choe JY, Han SM, Kim CH, Chang HW, Moon SK et al (2012) Melittin has an inhibitory effect on TNF-alpha-induced migration of human aortic smooth muscle cells by blocking the MMP-9 expression. Food Chem Toxicol 50:3996–4002CrossRefPubMed Jeong YJ, Cho HJ, Whang K, Lee IS, Park KK, Choe JY, Han SM, Kim CH, Chang HW, Moon SK et al (2012) Melittin has an inhibitory effect on TNF-alpha-induced migration of human aortic smooth muscle cells by blocking the MMP-9 expression. Food Chem Toxicol 50:3996–4002CrossRefPubMed
70.
go back to reference Jeong YJ, Choi Y, Shin JM, Cho HJ, Kang JH, Park KK, Choe JY, Bae YS, Han SM, Kim CH et al (2014) Melittin suppresses EGF-induced cell motility and invasion by inhibiting PI3 K/Akt/mTOR signaling pathway in breast cancer cells. Food Chem Toxicol 68:218–225CrossRefPubMed Jeong YJ, Choi Y, Shin JM, Cho HJ, Kang JH, Park KK, Choe JY, Bae YS, Han SM, Kim CH et al (2014) Melittin suppresses EGF-induced cell motility and invasion by inhibiting PI3 K/Akt/mTOR signaling pathway in breast cancer cells. Food Chem Toxicol 68:218–225CrossRefPubMed
71.
go back to reference Park JH, Jeong YJ, Park KK, Cho HJ, Chung IK, Min KS, Kim M, Lee KG, Yeo JH, Chang YC et al (2010) Melittin suppresses PMA-induced tumor cell invasion by inhibiting NF-kappaB and AP-1-dependent MMP-9 expression. Mol Cells 29:209–215CrossRefPubMed Park JH, Jeong YJ, Park KK, Cho HJ, Chung IK, Min KS, Kim M, Lee KG, Yeo JH, Chang YC et al (2010) Melittin suppresses PMA-induced tumor cell invasion by inhibiting NF-kappaB and AP-1-dependent MMP-9 expression. Mol Cells 29:209–215CrossRefPubMed
72.
go back to reference Paduch R (2016) The role of lymphangiogenesis and angiogenesis in tumor metastasis. Cell Oncol (Dordr) Paduch R (2016) The role of lymphangiogenesis and angiogenesis in tumor metastasis. Cell Oncol (Dordr)
73.
go back to reference Huh JE, Kang JW, Nam D, Baek YH, Choi DY, Park DS, Lee JD (2012) Melittin suppresses VEGF-A-induced tumor growth by blocking VEGFR-2 and the COX-2-mediated MAPK signaling pathway. J Nat Prod 75:1922–1929CrossRefPubMed Huh JE, Kang JW, Nam D, Baek YH, Choi DY, Park DS, Lee JD (2012) Melittin suppresses VEGF-A-induced tumor growth by blocking VEGFR-2 and the COX-2-mediated MAPK signaling pathway. J Nat Prod 75:1922–1929CrossRefPubMed
74.
go back to reference Shin JM, Jeong YJ, Cho HJ, Park KK, Chung IK, Lee IK, Kwak JY, Chang HW, Kim CH, Moon SK et al (2013) Melittin suppresses HIF-1alpha/VEGF expression through inhibition of ERK and mTOR/p70S6K pathway in human cervical carcinoma cells. PLoS One 8:e69380CrossRefPubMedPubMedCentral Shin JM, Jeong YJ, Cho HJ, Park KK, Chung IK, Lee IK, Kwak JY, Chang HW, Kim CH, Moon SK et al (2013) Melittin suppresses HIF-1alpha/VEGF expression through inhibition of ERK and mTOR/p70S6K pathway in human cervical carcinoma cells. PLoS One 8:e69380CrossRefPubMedPubMedCentral
75.
go back to reference Song CC, Lu X, Cheng BB, Du J, Li B, Ling CQ (2007) Effects of melittin on growth and angiogenesis of human hepatocellular carcinoma BEL-7402 cell xenografts in nude mice. Ai Zheng 26:1315–1322PubMed Song CC, Lu X, Cheng BB, Du J, Li B, Ling CQ (2007) Effects of melittin on growth and angiogenesis of human hepatocellular carcinoma BEL-7402 cell xenografts in nude mice. Ai Zheng 26:1315–1322PubMed
76.
go back to reference Zhang Z, Zhang H, Peng T, Li D, Xu J (2016) Melittin suppresses cathepsin S-induced invasion and angiogenesis via blocking of the VEGF-A/VEGFR-2/MEK1/ERK1/2 pathway in human hepatocellular carcinoma. Oncol Lett 11:610–618PubMed Zhang Z, Zhang H, Peng T, Li D, Xu J (2016) Melittin suppresses cathepsin S-induced invasion and angiogenesis via blocking of the VEGF-A/VEGFR-2/MEK1/ERK1/2 pathway in human hepatocellular carcinoma. Oncol Lett 11:610–618PubMed
78.
go back to reference Wu X, Zhao B, Cheng Y, Yang Y, Huang C, Meng X, Wu B, Zhang L, Lv X, Li J et al (2015) Melittin induces PTCH1 expression by down-regulating MeCP2 in human hepatocellular carcinoma SMMC-7721 cells. Toxicol Appl Pharmacol 288:74–83CrossRefPubMed Wu X, Zhao B, Cheng Y, Yang Y, Huang C, Meng X, Wu B, Zhang L, Lv X, Li J et al (2015) Melittin induces PTCH1 expression by down-regulating MeCP2 in human hepatocellular carcinoma SMMC-7721 cells. Toxicol Appl Pharmacol 288:74–83CrossRefPubMed
79.
go back to reference Zhang H, Zhao B, Huang C, Meng XM, Bian EB, Li J (2014) Melittin restores PTEN expression by down-regulating HDAC2 in human hepatocelluar carcinoma HepG2 cells. PLoS One 9:e95520CrossRefPubMedPubMedCentral Zhang H, Zhao B, Huang C, Meng XM, Bian EB, Li J (2014) Melittin restores PTEN expression by down-regulating HDAC2 in human hepatocelluar carcinoma HepG2 cells. PLoS One 9:e95520CrossRefPubMedPubMedCentral
80.
go back to reference Orsolic N (2009) Potentiation of bleomycin lethality in HeLa and V79 cells by bee venom. Arh Hig Rada Toksikol 60:317–326CrossRefPubMed Orsolic N (2009) Potentiation of bleomycin lethality in HeLa and V79 cells by bee venom. Arh Hig Rada Toksikol 60:317–326CrossRefPubMed
81.
go back to reference Wang RP, Huang SR, Zhou JY, Zou X (2014) Synergistic interaction between melittin and chemotherapeutic agents and their possible mechanisms: an experimental research. Zhongguo Zhong Xi Yi Jie He Za Zhi 34:224–229PubMed Wang RP, Huang SR, Zhou JY, Zou X (2014) Synergistic interaction between melittin and chemotherapeutic agents and their possible mechanisms: an experimental research. Zhongguo Zhong Xi Yi Jie He Za Zhi 34:224–229PubMed
82.
go back to reference Zhu H, Yang X, Liu J, Ge Y, Qin Q, Lu J, Zhan L, Liu Z, Zhang H, Chen X et al (2014) Melittin radiosensitizes esophageal squamous cell carcinoma with induction of apoptosis in vitro and in vivo. Tumour Biol 35:8699–8705CrossRefPubMed Zhu H, Yang X, Liu J, Ge Y, Qin Q, Lu J, Zhan L, Liu Z, Zhang H, Chen X et al (2014) Melittin radiosensitizes esophageal squamous cell carcinoma with induction of apoptosis in vitro and in vivo. Tumour Biol 35:8699–8705CrossRefPubMed
83.
go back to reference Asthana N, Yadav SP, Ghosh JK (2004) Dissection of antibacterial and toxic activity of melittin: a leucine zipper motif plays a crucial role in determining its hemolytic activity but not antibacterial activity. J Biol Chem 279:55042–55050CrossRefPubMed Asthana N, Yadav SP, Ghosh JK (2004) Dissection of antibacterial and toxic activity of melittin: a leucine zipper motif plays a crucial role in determining its hemolytic activity but not antibacterial activity. J Biol Chem 279:55042–55050CrossRefPubMed
84.
go back to reference Dunn RD, Weston KM, Longhurst TJ, Lilley GG, Rivett DE, Hudson PJ, Raison RL (1996) Antigen binding and cytotoxic properties of a recombinant immunotoxin incorporating the lytic peptide, melittin. Immunotechnology 2:229–240CrossRefPubMed Dunn RD, Weston KM, Longhurst TJ, Lilley GG, Rivett DE, Hudson PJ, Raison RL (1996) Antigen binding and cytotoxic properties of a recombinant immunotoxin incorporating the lytic peptide, melittin. Immunotechnology 2:229–240CrossRefPubMed
85.
go back to reference Huang C, Jin H, Qian Y, Qi S, Luo H, Luo Q, Zhang Z (2013) Hybrid melittin cytolytic peptide-driven ultrasmall lipid nanoparticles block melanoma growth in vivo. ACS Nano 7:5791–5800CrossRefPubMed Huang C, Jin H, Qian Y, Qi S, Luo H, Luo Q, Zhang Z (2013) Hybrid melittin cytolytic peptide-driven ultrasmall lipid nanoparticles block melanoma growth in vivo. ACS Nano 7:5791–5800CrossRefPubMed
86.
go back to reference Glinka EM (2012) Eukaryotic expression vectors bearing genes encoding cytotoxic proteins for cancer gene therapy. Plasmid 68:69–85CrossRefPubMed Glinka EM (2012) Eukaryotic expression vectors bearing genes encoding cytotoxic proteins for cancer gene therapy. Plasmid 68:69–85CrossRefPubMed
87.
go back to reference Tuppurainen L, Sallinen H, Kokki E, Koponen J, Anttila M, Pulkkinen K, Heikura T, Toivanen P, Hamalainen K, Kosma VM et al (2013) Preclinical safety, toxicology, and biodistribution study of adenoviral gene therapy with sVEGFR-2 and sVEGFR-3 combined with chemotherapy for ovarian cancer. Hum Gene Ther Clin Dev 24:29–37CrossRefPubMed Tuppurainen L, Sallinen H, Kokki E, Koponen J, Anttila M, Pulkkinen K, Heikura T, Toivanen P, Hamalainen K, Kosma VM et al (2013) Preclinical safety, toxicology, and biodistribution study of adenoviral gene therapy with sVEGFR-2 and sVEGFR-3 combined with chemotherapy for ovarian cancer. Hum Gene Ther Clin Dev 24:29–37CrossRefPubMed
88.
go back to reference Buscail L, Bournet B, Vernejoul F, Cambois G, Lulka H, Hanoun N, Dufresne M, Meulle A, Vignolle-Vidoni A, Ligat L et al (2015) First-in-man phase 1 clinical trial of gene therapy for advanced pancreatic cancer: safety, biodistribution, and preliminary clinical findings. Mol Ther 23:779–789CrossRefPubMedPubMedCentral Buscail L, Bournet B, Vernejoul F, Cambois G, Lulka H, Hanoun N, Dufresne M, Meulle A, Vignolle-Vidoni A, Ligat L et al (2015) First-in-man phase 1 clinical trial of gene therapy for advanced pancreatic cancer: safety, biodistribution, and preliminary clinical findings. Mol Ther 23:779–789CrossRefPubMedPubMedCentral
89.
go back to reference Su M, Chang W, Cui M, Lin Y, Wu S, Xu T (2015) Expression and anticancer activity analysis of recombinant human uPA143-melittin. Int J Oncol 46:619–626PubMed Su M, Chang W, Cui M, Lin Y, Wu S, Xu T (2015) Expression and anticancer activity analysis of recombinant human uPA143-melittin. Int J Oncol 46:619–626PubMed
90.
go back to reference Su M, Chang W, Zhang K, Cui M, Wu S, Xu T (2016) Expression and purification of recombinant ATF-mellitin, a new type fusion protein targeting ovarian cancer cells, in P. pastoris. Oncol Rep 35:1179–1185PubMed Su M, Chang W, Zhang K, Cui M, Wu S, Xu T (2016) Expression and purification of recombinant ATF-mellitin, a new type fusion protein targeting ovarian cancer cells, in P. pastoris. Oncol Rep 35:1179–1185PubMed
91.
go back to reference Wang D, Hu L, Su M, Wang J, Xu T (2015) Preparation and functional characterization of human vascular endothelial growth factor-melittin fusion protein with analysis of the antitumor activity in vitro and in vivo. Int J Oncol 47:1160–1168PubMed Wang D, Hu L, Su M, Wang J, Xu T (2015) Preparation and functional characterization of human vascular endothelial growth factor-melittin fusion protein with analysis of the antitumor activity in vitro and in vivo. Int J Oncol 47:1160–1168PubMed
92.
go back to reference Holle L, Song W, Holle E, Wei Y, Li J, Wagner TE, Yu X (2009) In vitro- and in vivo-targeted tumor lysis by an MMP2 cleavable melittin-LAP fusion protein. Int J Oncol 35:829–835PubMed Holle L, Song W, Holle E, Wei Y, Li J, Wagner TE, Yu X (2009) In vitro- and in vivo-targeted tumor lysis by an MMP2 cleavable melittin-LAP fusion protein. Int J Oncol 35:829–835PubMed
93.
go back to reference Li B, Gu W, Zhang C, Huang XQ, Han KQ, Ling CQ (2006) Growth arrest and apoptosis of the human hepatocellular carcinoma cell line BEL-7402 induced by melittin. Onkologie 29:367–371PubMed Li B, Gu W, Zhang C, Huang XQ, Han KQ, Ling CQ (2006) Growth arrest and apoptosis of the human hepatocellular carcinoma cell line BEL-7402 induced by melittin. Onkologie 29:367–371PubMed
94.
go back to reference Ling CQ, Li B, Zhang C, Zhu DZ, Huang XQ, Gu W, Li SX (2005) Inhibitory effect of recombinant adenovirus carrying melittin gene on hepatocellular carcinoma. Ann Oncol 16:109–115CrossRefPubMed Ling CQ, Li B, Zhang C, Zhu DZ, Huang XQ, Gu W, Li SX (2005) Inhibitory effect of recombinant adenovirus carrying melittin gene on hepatocellular carcinoma. Ann Oncol 16:109–115CrossRefPubMed
95.
go back to reference Qian CY, Wang KL, Fang FF, Gu W, Huang F, Wang FZ, Li B, Wang LN (2015) Triple-controlled oncolytic adenovirus expressing melittin to exert inhibitory efficacy on hepatocellular carcinoma. Int J Clin Exp Pathol 8:10403–10411PubMedPubMedCentral Qian CY, Wang KL, Fang FF, Gu W, Huang F, Wang FZ, Li B, Wang LN (2015) Triple-controlled oncolytic adenovirus expressing melittin to exert inhibitory efficacy on hepatocellular carcinoma. Int J Clin Exp Pathol 8:10403–10411PubMedPubMedCentral
96.
go back to reference Russell PJ, Hewish D, Carter T, Sterling-Levis K, Ow K, Hattarki M, Doughty L, Guthrie R, Shapira D, Molloy PL et al (2004) Cytotoxic properties of immunoconjugates containing melittin-like peptide 101 against prostate cancer: in vitro and in vivo studies. Cancer Immunol Immunother 53:411–421CrossRefPubMed Russell PJ, Hewish D, Carter T, Sterling-Levis K, Ow K, Hattarki M, Doughty L, Guthrie R, Shapira D, Molloy PL et al (2004) Cytotoxic properties of immunoconjugates containing melittin-like peptide 101 against prostate cancer: in vitro and in vivo studies. Cancer Immunol Immunother 53:411–421CrossRefPubMed
97.
go back to reference Zhao X, Yu Z, Dai W, Yao Z, Zhou W, Zhou J, Yang Y, Zhu Y, Chen S, Cao L et al (2011) Construction and characterization of an anti-asialoglycoprotein receptor single-chain variable-fragment-targeted melittin. Biotechnol Appl Biochem 58:405–411CrossRefPubMed Zhao X, Yu Z, Dai W, Yao Z, Zhou W, Zhou J, Yang Y, Zhu Y, Chen S, Cao L et al (2011) Construction and characterization of an anti-asialoglycoprotein receptor single-chain variable-fragment-targeted melittin. Biotechnol Appl Biochem 58:405–411CrossRefPubMed
98.
go back to reference Zhao H, Feng X, Han W, Diao Y, Han D, Tian X, Gao Y, Liu S, Zhu S, Yao C et al (2013) Enhanced binding to and killing of hepatocellular carcinoma cells in vitro by melittin when linked with a novel targeting peptide screened from phage display. J Pept Sci 19:639–650CrossRefPubMed Zhao H, Feng X, Han W, Diao Y, Han D, Tian X, Gao Y, Liu S, Zhu S, Yao C et al (2013) Enhanced binding to and killing of hepatocellular carcinoma cells in vitro by melittin when linked with a novel targeting peptide screened from phage display. J Pept Sci 19:639–650CrossRefPubMed
100.
go back to reference Vago R, Collico V, Zuppone S, Prosperi D, Colombo M (2016) Nanoparticle-mediated delivery of suicide genes in cancer therapy. Pharmacol Res 111:619–641CrossRefPubMed Vago R, Collico V, Zuppone S, Prosperi D, Colombo M (2016) Nanoparticle-mediated delivery of suicide genes in cancer therapy. Pharmacol Res 111:619–641CrossRefPubMed
101.
go back to reference Soman NR, Lanza GM, Heuser JM, Schlesinger PH, Wickline SA (2008) Synthesis and characterization of stable fluorocarbon nanostructures as drug delivery vehicles for cytolytic peptides. Nano Lett 8:1131–1136CrossRefPubMedPubMedCentral Soman NR, Lanza GM, Heuser JM, Schlesinger PH, Wickline SA (2008) Synthesis and characterization of stable fluorocarbon nanostructures as drug delivery vehicles for cytolytic peptides. Nano Lett 8:1131–1136CrossRefPubMedPubMedCentral
102.
go back to reference Pan H, Soman NR, Schlesinger PH, Lanza GM, Wickline SA (2011) Cytolytic peptide nanoparticles (‘NanoBees’) for cancer therapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol 3:318–327CrossRefPubMed Pan H, Soman NR, Schlesinger PH, Lanza GM, Wickline SA (2011) Cytolytic peptide nanoparticles (‘NanoBees’) for cancer therapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol 3:318–327CrossRefPubMed
103.
go back to reference Soman NR, Baldwin SL, Hu G, Marsh JN, Lanza GM, Heuser JE, Arbeit JM, Wickline SA, Schlesinger PH (2009) Molecularly targeted nanocarriers deliver the cytolytic peptide melittin specifically to tumor cells in mice, reducing tumor growth. J Clin Invest 119:2830–2842CrossRefPubMedPubMedCentral Soman NR, Baldwin SL, Hu G, Marsh JN, Lanza GM, Heuser JE, Arbeit JM, Wickline SA, Schlesinger PH (2009) Molecularly targeted nanocarriers deliver the cytolytic peptide melittin specifically to tumor cells in mice, reducing tumor growth. J Clin Invest 119:2830–2842CrossRefPubMedPubMedCentral
104.
go back to reference Jallouk AP, Palekar RU, Marsh JN, Pan H, Pham CT, Schlesinger PH, Wickline SA (2015) Delivery of a protease-activated cytolytic peptide prodrug by perfluorocarbon nanoparticles. Bioconjug Chem 26:1640–1650CrossRefPubMedPubMedCentral Jallouk AP, Palekar RU, Marsh JN, Pan H, Pham CT, Schlesinger PH, Wickline SA (2015) Delivery of a protease-activated cytolytic peptide prodrug by perfluorocarbon nanoparticles. Bioconjug Chem 26:1640–1650CrossRefPubMedPubMedCentral
105.
go back to reference Lavignac N, Lazenby M, Franchini J, Ferruti P, Duncan R (2005) Synthesis and preliminary evaluation of poly(amidoamine)-melittin conjugates as endosomolytic polymers and/or potential anticancer therapeutics. Int J Pharm 300:102–112CrossRefPubMed Lavignac N, Lazenby M, Franchini J, Ferruti P, Duncan R (2005) Synthesis and preliminary evaluation of poly(amidoamine)-melittin conjugates as endosomolytic polymers and/or potential anticancer therapeutics. Int J Pharm 300:102–112CrossRefPubMed
106.
go back to reference Schaffert D, Kiss M, Rodl W, Shir A, Levitzki A, Ogris M, Wagner E (2011) Poly(I:C)-mediated tumor growth suppression in EGF-receptor overexpressing tumors using EGF-polyethylene glycol-linear polyethylenimine as carrier. Pharm Res 28:731–741CrossRefPubMed Schaffert D, Kiss M, Rodl W, Shir A, Levitzki A, Ogris M, Wagner E (2011) Poly(I:C)-mediated tumor growth suppression in EGF-receptor overexpressing tumors using EGF-polyethylene glycol-linear polyethylenimine as carrier. Pharm Res 28:731–741CrossRefPubMed
107.
108.
go back to reference Zetterberg MM, Reijmar K, Pranting M, Engstrom A, Andersson DI, Edwards K (2011) PEG-stabilized lipid disks as carriers for amphiphilic antimicrobial peptides. J Control Release 156:323–328CrossRefPubMed Zetterberg MM, Reijmar K, Pranting M, Engstrom A, Andersson DI, Edwards K (2011) PEG-stabilized lipid disks as carriers for amphiphilic antimicrobial peptides. J Control Release 156:323–328CrossRefPubMed
109.
go back to reference Gao J, Xie C, Zhang M, Wei X, Yan Z, Ren Y, Ying M, Lu W (2016) RGD-modified lipid disks as drug carriers for tumor targeted drug delivery. Nanoscale 8:7209–7216CrossRefPubMed Gao J, Xie C, Zhang M, Wei X, Yan Z, Ren Y, Ying M, Lu W (2016) RGD-modified lipid disks as drug carriers for tumor targeted drug delivery. Nanoscale 8:7209–7216CrossRefPubMed
110.
go back to reference Yang L, Cui F, Shi K, Cun D, Wang R (2009) Design of high payload PLGA nanoparticles containing melittin/sodium dodecyl sulfate complex by the hydrophobic ion-pairing technique. Drug Dev Ind Pharm 35:959–968CrossRefPubMed Yang L, Cui F, Shi K, Cun D, Wang R (2009) Design of high payload PLGA nanoparticles containing melittin/sodium dodecyl sulfate complex by the hydrophobic ion-pairing technique. Drug Dev Ind Pharm 35:959–968CrossRefPubMed
112.
go back to reference Yang WS, Park SO, Yoon AR, Yoo JY, Kim MK, Yun CO, Kim CW (2006) Suicide cancer gene therapy using pore-forming toxin, streptolysin O. Mol Cancer Ther 5:1610–1619CrossRefPubMed Yang WS, Park SO, Yoon AR, Yoo JY, Kim MK, Yun CO, Kim CW (2006) Suicide cancer gene therapy using pore-forming toxin, streptolysin O. Mol Cancer Ther 5:1610–1619CrossRefPubMed
113.
go back to reference Ramamoorth M, Narvekar A (2015) Non viral vectors in gene therapy—an overview. J Clin Diagn Res 9:GE01-06 Ramamoorth M, Narvekar A (2015) Non viral vectors in gene therapy—an overview. J Clin Diagn Res 9:GE01-06
Metadata
Title
Application of bee venom and its main constituent melittin for cancer treatment
Authors
Cui-cui Liu
Ding-jun Hao
Qian Zhang
Jing An
Jing-jing Zhao
Bo Chen
Ling-ling Zhang
Hao Yang
Publication date
01-12-2016
Publisher
Springer Berlin Heidelberg
Published in
Cancer Chemotherapy and Pharmacology / Issue 6/2016
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-016-3160-1

Other articles of this Issue 6/2016

Cancer Chemotherapy and Pharmacology 6/2016 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine