483
Views
1
CrossRef citations to date
0
Altmetric
Reviews

Cellular targets and molecular activity mechanisms of bee venom in cancer: recent trends and developments

, , , , , ORCID Icon, , , , & ORCID Icon show all
Pages 1382-1395 | Received 14 Aug 2021, Accepted 27 Dec 2021, Published online: 15 Feb 2022

References

  • Aggarwal, V., et al., 2019. Role of reactive oxygen species in cancer progression: molecular mechanisms and recent advancements. Biomolecules, 9 (11), 735.
  • Alalawy, A.I., et al., 2020. Effectual anticancer potentiality of loaded bee venom onto fungal chitosan nanoparticles. International journal of polymer science, 2020, 1–9.
  • Ali, M., 2012. Studies on bee venom and its medical uses. International journal of advanced research Technol, 1, 69–83.
  • Alizadehnohi, M., et al., 2012. The synergistic cytotoxic effect of cisplatin and honey bee venom on human ovarian cancer cell line A2780cp. Journal of venom research, 3, 22–27.
  • Almunia, C., et al., 2013. Bee venom phospholipase A2, a good “chauffeur” for delivering tumor antigen to the MHC I and MHC II peptide-loading compartments of the dendritic cells: the case of NY-ESO-1. PLOS One, 8 (6), e67645.
  • Amin, M., Abdel-Raheem, I., and Madkor, H., 2008. Wound healing and anti-inflammatory activities of bee venom-chitosan blend films. Journal of drug delivery science and technology, 18 (6), 424–430.
  • An, H.-J., et al., 2016. The protective effect of melittin on renal fibrosis in an animal model of unilateral ureteral obstruction. Molecules, 21 (9), 1137.
  • Bačkorová, M., et al., 2012. Lichen secondary metabolites are responsible for induction of apoptosis in HT-29 and A2780 human cancer cell lines. Toxicology in vitro, 26 (3), 462–468.
  • Badr-Eldin, S.M., et al., 2020. Cytotoxic and pro-apoptotic effects of a sub-toxic concentration of fluvastatin on OVCAR3 ovarian cancer cells after its optimized formulation to melittin nano-conjugates. Frontiers in pharmacology, 11, 642171.
  • Badr, G., et al., 2016. Bee venom accelerates wound healing in diabetic mice by suppressing activating transcription factor-3 (ATF-3) and inducible nitric oxide synthase (iNOS)-mediated oxidative stress and recruiting bone marrow-derived endothelial progenitor cells. Journal of cellular physiology, 231 (10), 2159–2171.
  • Badria, F., et al., 2017. Evaluate the cytotoxic activity of honey, propolis, and bee venom from different localities in Egypt against liver, breast, and colorectal cancer. Journal of apitherapy, 2 (1), 1–4.
  • Bae, G.-S., et al., 2013. Apamin attenuated cerulein-induced acute pancreatitis by inhibition of JNK pathway in mice. Digestive diseases and sciences, 58 (10), 2908–2917.
  • Bagyalakshmi, B., Priyadarshini, S.L., and Balamurugan, A., 2019. Anticancer activity of bee venom against lung cancer cell line (A549 cells) enhanced by iron oxide nanoparticles synthesized from Syzygium aromaticum. Journal of drug delivery and therapeutics, 9, 248–254.
  • Bellik, Y., 2015. Bee venom: its potential use in alternative medicine. Anti-infective agents, 13 (1), 3–16.
  • Bray, F., et al., 2018. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. A cancer journal for clinicians, 68 (6), 394–424.
  • Celis, J.E., and Heitor, M., 2019. Towards a mission-oriented approach to cancer in Europe: an unmet need in cancer research policy. Molecular oncology, 13 (3), 502–510.
  • Chen, J., and Guan, S.-M., 2017. Bee Venom and Pain. In: L.J. Cruz, S. Luo and P. Gopalakrishnakone, eds. Toxins and drug discovery. Dordrecht: Springer Netherlands, 253–294.
  • Chen, J., and Lariviere, W.R., 2010. The nociceptive and anti-nociceptive effects of bee venom injection and therapy: a double-edged sword. Progress in neurobiology, 92 (2), 151–183.
  • Cho, H.-J., et al., 2010. Bee venom suppresses PMA-mediated MMP-9 gene activation via JNK/p38 and NF-kappaB-dependent mechanisms. Journal of ethnopharmacology, 127 (3), 662–668.
  • Choi, K.E., 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, 6 (8), 2210–2228.
  • Cichocka-Jarosz, E., 2012. Hymenoptera venom allergy in humans. Folia Medica Cracoviensia, 52, 43-60.
  • Cooney, J., et al., 2011. Benefits of exercise in rheumatoid arthritis. Journal of aging research, 2011, 1–14.
  • Davies, J., et al., 2019. Recent developments in advanced imaging in gout. Therapeutic advances in musculoskeletal disease, 11, 1759720X19844429.
  • El Bakary, N.M., et al., 2020. Role of bee venom and melittin on restraining angiogenesis and metastasis in γ-irradiated solid Ehrlich carcinoma-bearing mice. Integrative cancer therapies, 19, 1534735420944476.
  • Fujita, Y., Abe, R., and Shimizu, H., 2008. Clinical approaches toward tumor angiogenesis: past, present and future. Current pharmaceutical design, 14 (36), 3820–3834.
  • Gajski, G., and Garaj-Vrhovac, V., 2009. Radioprotective effects of honeybee venom (Apis mellifera) against 915-mhz microwave radiation-induced DNA damage in Wistar rat lymphocytes: in vitro study. International journal of toxicology, 28 (2), 88–98.
  • Gajski, G., and Garaj-Vrhovac, V., 2011. Bee venom induced cytogenetic damage and decreased cell viability in human white blood cells after treatment in vitro: a multi-biomarker approach. Environmental toxicology and pharmacology, 32 (2), 201–211.
  • Gao, J., et al., 2016. RGD-modified lipid disks as drug carriers for tumor targeted drug delivery. Nanoscale, 8 (13), 7209–7216.
  • Glass, C.K., et al., 2010. Mechanisms underlying inflammation in neurodegeneration. Cell, 140 (6), 918–934.
  • Hanahan, D., and Folkman, J., 1996. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell, 86 (3), 353–364.
  • Hanahan, D., and Weinberg, R.A., 2000. The hallmarks of cancer. Cell, 100 (1), 57–70.
  • Hanahan, D., and Weinberg, R.A., 2011. Hallmarks of cancer: the next generation. Cell, 144 (5), 646–674.
  • Hatef, M., and Balali-Mood, M., 1995. Collection and standardization of honey bee venom in Iran. Journal of natural toxins, 4, 139.
  • Hematyar, M., et al., 2018. Synergistic co-delivery of doxorubicin and melittin using functionalized magnetic nanoparticles for cancer treatment: loading and in vitro release study by LC-MS/MS. Artificial cells, nanomedicine, and biotechnology, 46 (sup3), S1226–S1235.
  • Hong, S.-J., et al., 2005. Bee venom induces apoptosis through caspase-3 activation in synovial fibroblasts of patients with rheumatoid arthritis. Toxicon, 46 (1), 39–45.
  • Huh, J.-E., et al., 2010. Bee venom inhibits tumor angiogenesis and metastasis by inhibiting tyrosine phosphorylation of VEGFR-2 in LLC-tumor-bearing mice. Cancer letters, 292 (1), 98–110.
  • Huh, J.-E., et al., 2012. Melittin suppresses VEGF-A-induced tumor growth by blocking VEGFR-2 and the COX-2-mediated MAPK signaling pathway. Journal of natural products, 75 (11), 1922–1929.
  • Ip, S.-W., et al., 2008. Bee venom induced cell cycle arrest and apoptosis in human cervical epidermoid carcinoma Ca Ski cells. Anticancer research, 28, 833–842.
  • Jallouk, A.P., et al., 2015. Delivery of a protease-activated cytolytic peptide prodrug by perfluorocarbon nanoparticles. Bioconjugate chemistry, 26 (8), 1640–1650.
  • Jang, H.-S., et al., 2005. Effects of bee venom on the pro-inflammatory responses in RAW264.7 macrophage cell line. Journal of ethnopharmacology, 99 (1), 157–160.
  • Jeong, Y.-J., et al., 2019. Bee venom suppresses egf-induced epithelial-mesenchymal transition and tumor invasion in lung cancer cells. The American journal of chinese medicine, 47 (8), 1869–1883.
  • Jin, H., et al., 2017. Melittin-containing hybrid peptide hydrogels for enhanced photothermal therapy of glioblastoma. ACS applied materials & interfaces, 9 (31), 25755–25766.
  • Jo, M., et al., 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. Toxicology and applied pharmacology, 258 (1), 72–81.
  • Jones, S.L., Jones, H.R., and Thrasyvoulou, A., 2011. Disseminating research about bee products. A review of articles published in the journal of apicultural research over the past fifty years. Journal of apiproduct & apimedical science, 3 (3), 105–116.
  • Kim, J.-Y., et al., 2017. Apamin suppresses biliary fibrosis and activation of hepatic stellate cells. International journal of molecular medicine, 39 (5), 1188–1194.
  • Kim, J.-Y., Leem, J., and Hong, H.-L., 2021. Melittin ameliorates endotoxin-induced acute kidney injury by inhibiting inflammation, oxidative stress, and cell death in mice. Oxidative medicine and cellular longevity, 2021, 8843051.
  • Kim, S.-J., et al., 2012. The protective effect of apamin on LPS/fat-induced atherosclerotic mice. Evidence-based complementary and alternative medicine, 2012, 305454.
  • Kim, S.H., et al., 2013. Bee venom effects on ubiquitin proteasome system in hSOD1 G85R-expressing NSC34 motor neuron cells. BMC complementary and alternative medicine, 13 (1), 179.
  • Kim, W.-H., et al., 2018. Anti-inflammatory effect of melittin on Porphyromonas gingivalis LPS-stimulated human keratinocytes. Molecules, 23 (2), 332.
  • Kim, Y., et al., 2019. Bee venom alleviates atopic dermatitis symptoms through the upregulation of decay-accelerating factor (DAF/CD55). Toxins, 11 (5), 239.
  • Koburova, K., Michailova, S., and Shkenderov, S., 1985. Further investigation on the antiinflammatory properties of adolapin–bee venom polypeptide. Acta Physiologica et Pharmacologica Bulgarica, 11 (2), 50–55.
  • Kocyigit, A., Guler, E.M., and Kaleli, S., 2019. Anti-inflammatory and antioxidative properties of honey bee venom on Freund's complete adjuvant-induced arthritis model in rats. Toxicon, 161, 4–11.
  • Koh, P.S., et al., 2013. Clinical effectiveness of bee venom acupuncture and physiotherapy in the treatment of adhesive capsulitis: a randomized controlled trial. Journal of shoulder and elbow surgery, 22 (8), 1053–1062.
  • Kollipara, P.S., et al., 2014. Co-culture with NK-92MI cells enhanced the anti-cancer effect of bee venom on NSCLC cells by inactivation of NF-κB. Archives of pharmacal research, 37 (3), 379–389.
  • Kong, G.-M., et al., 2016. Melittin induces human gastric cancer cell apoptosis via activation of mitochondrial pathway. World journal of gastroenterology, 22 (11), 3186.
  • Kurek-Górecka, A., et al., 2020. Bee venom in wound healing. Molecules, 26 (1), 148.
  • Kwon, Y.B., et al., 2002. The water-soluble fraction of bee venom produces antinociceptive and anti-inflammatory effects on rheumatoid arthritis in rats. Life Sciences, 71 (2), 191–204.
  • Lai, H., et al., 2017. Polypeptide-grafted nanodiamonds for controlled release of melittin to treat breast cancer. ACS macro letters, 6 (8), 796–801.
  • Lee, G., and Bae, H., 2016a. Anti-inflammatory applications of melittin, a major component of bee venom: detailed mechanism of action and adverse effects. Molecules, 21 (5), 616.
  • Lee, G., and Bae, H., 2016b. Bee venom phospholipase A2: yesterday's enemy becomes today's friend. Toxins, 8 (2), 48.
  • Lee, J.-D., et al., 2005. An overview of bee venom acupuncture in the treatment of arthritis. Evidence-based complementary and alternative medicine, 2 (1), 79–84.
  • Lee, W.-R., et al., 2014. The protective effects of melittin on propionibacterium acnes-induced inflammatory responses in vitro and in vivo. The journal of investigative dermatology, 134 (7), 1922–1930.
  • Lee, Y.J., et al., 2020a. Anti-inflammatory effect of bee venom in phthalic anhydride-induced atopic dermatitis animal model. Inflammopharmacology, 28 (1), 253–263.
  • Lee, Y.M., et al., 2020b. Apamin from bee venom suppresses inflammation in a murine model of gouty arthritis. Journal of ethnopharmacology, 257, 112860.
  • Lin, T.-Y., and Hsieh, C.-L., 2020. Clinical applications of bee venom acupoint injection. Toxins, 12 (10), 618.
  • Liu, C.-C., et al., 2016. Application of bee venom and its main constituent melittin for cancer treatment. Cancer chemotherapy and pharmacology, 78 (6), 1113–1130.
  • Liu, H., et al., 2019. Co-delivery of bee venom melittin and a photosensitizer with an organic-inorganic hybrid nanocarrier for photodynamic therapy and immunotherapy. ACS Nano, 13 (11), 12638–12652.
  • Liu, S., et al., 2008. Melittin prevents liver cancer cell metastasis through inhibition of the Rac1-dependent pathway. Hepatology, 47 (6), 1964–1973.
  • Liu, X., et al., 2002. Effect of honey bee venom on proliferation of K1735M2 mouse melanoma cells in-vitro and growth of murine B16 melanomas in-vivo. The journal of pharmacy and pharmacology, 54 (8), 1083–1089.
  • Lokeshwar, V.B., and Selzer, M.G., 2009. Hyaluronidase: both a tumor promoter and suppressor. Hyaluronan in cancer biology, Academic Press, Cambridge, Massachusetts, USA, 189–206.
  • Maher, S., and Mcclean, S., 2008. Melittin exhibits necrotic cytotoxicity in gastrointestinal cells which is attenuated by cholesterol. Biochemical pharmacology, 75 (5), 1104–1114.
  • Mansoori, B., et al., 2017. The different mechanisms of cancer drug resistance: a brief review. Advanced pharmaceutical bulletin, 7 (3), 339–348.
  • Matusiewicz, M., et al., 2014. Matrix metalloproteinase-9: its interplay with angiogenic factors in inflammatory bowel diseases. Disease markers, 2014, 643645.
  • Mcatee, C.O., Barycki, J.J., and Simpson, M.A., 2014. Emerging roles for hyaluronidase in cancer metastasis and therapy. Advances in cancer research, 123, 1–34.
  • Moghaddam, F.D., et al., 2020. Delivery of melittin loaded niosomes for breast cancer treatment: an in-vitro and in-vivo evaluation of anti-cancer effect.
  • Mohamed, W.A., Abd-Elhakim, Y.M., and Ismail, S.A., 2019. Involvement of the anti-inflammatory, anti-apoptotic, and anti-secretory activity of bee venom in its therapeutic effects on acetylsalicylic acid-induced gastric ulceration in rats. Toxicology, 419, 11–23.
  • Mohammadi-Rad, M., Ghasemi, N., and Aliomrani, M., 2019. Evaluation of apamin effects on myelination process in C57BL/6 mice model of multiple sclerosis. Research in pharmaceutical sciences, 14 (5), 424–431.
  • Moon, D.-O., et al., 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. International immunopharmacology, 6 (12), 1796–1807.
  • Mueller, U.R., 2007. Cardiovascular disease and anaphylaxis. Current opinion in allergy and clinical immunology, 7 (4), 337–341.
  • Ong, P.Y., and Leung, D.Y., 2016. Bacterial and viral infections in atopic dermatitis: a comprehensive review. Clinical reviews in allergy & immunology, 51 (3), 329–337.
  • Oršolić, N., 2009. Potentiation of bleomycin lethality in HeLa and V79 cells by bee venom. Archives of industrial hygiene and toxicology, 60 (3), 317–326.
  • Oršolić, N., 2012. Bee venom in cancer therapy. Cancer metastasis reviews, 31 (1–2), 173–194.
  • Ozturk, A., et al., 2017. Comparison of inflammatory cytokine release from nasal epithelial cells of non-atopic non-rhinitic, allergic rhinitic and polyp subjects and effects of diesel exhaust particles in vitro. Allergologia et Immunopathologia, 45 (5), 473–481.
  • Pan, H., et al., 2011. Cytolytic peptide nanoparticles ('NanoBees') for cancer therapy. wiley interdisciplinary reviews-nanomedicine and nanobiotechnology, 3 (3), 318–327.
  • Park, H.G., et al., 2018a. Honeybee (Apis cerana) vitellogenin acts as an antimicrobial and antioxidant agent in the body and venom. Developmental and comparative immunology, 85, 51–60.
  • Park, H.J., et al., 2004. Antiarthritic effect of bee venom: Inhibition of inflammation mediator generation by suppression of NF-kappaB through interaction with the p50 subunit. Arthritis and rheumatism, 50 (11), 3504–3515.
  • Park, H.J., et al., 2007. Melittin inhibits inflammatory target gene expression and mediator generation via interaction with IkappaB kinase. Biochemical pharmacology, 73 (2), 237–247.
  • Park, J.H., et al., 2010. Melittin suppresses PMA-induced tumor cell invasion by inhibiting NF-kappaB and AP-1-dependent MMP-9 expression. Molecules and cells, 29 (2), 209–215.
  • Park, J.H., et al., 2015. Risk associated with bee venom therapy: a systematic review and meta-analysis. PLOS one, 10 (5), e0126971.
  • Park, M.-H., et al., 2018b. Preparation and characterization of bee venom-loaded PLGA particles for sustained release. Pharmaceutical development and technology, 23 (9), 857–864.
  • Pascoal, A., et al., 2019. An overview of the bioactive compounds, therapeutic properties and toxic effects of apitoxin. Food and chemical toxicology,134, 110864–110811.
  • Pavel, C.I., et al., 2014. Comparison between local and commercial royal jelly—use of antioxidant activity and 10-hydroxy-2-decenoic acid as quality parameter. Journal of apicultural research, 53 (1), 116–123.
  • Pucca, M.B., et al., 2019. Bee updated: Current knowledge on bee venom and bee envenoming therapy. Frontiers in immunology, 10, 2090.
  • Qi, J., et al., 2020. Graphene oxide-based magnetic nanocomposites for the delivery of melittin to cervical cancer HeLa cells. Nanotechnology, 31 (6), 065102.
  • Rady, I., et al., 2017. Melittin, a major peptide component of bee venom, and its conjugates in cancer therapy. Cancer letters, 402, 16–31.
  • Raghuraman, H., and Chattopadhyay, A., 2007. Melittin: a membrane-active peptide with diverse functions. Bioscience reports, 27 (4–5), 189–223.
  • Rojiani, M.V., et al., 2010. Expression of MMP-2 correlates with increased angiogenesis in CNS metastasis of lung carcinoma. International journal of clinical and experimental pathology, 3 (8), 775–781.
  • Safaeinejad, Z., Nabiuni, M., and Nazari, Z., 2013. Potentiation of a novel palladium (II) complex lethality with bee venom on the human T-cell acute lymphoblastic leukemia cell line (MOLT-4). Journal of venomous animals and toxins including tropical diseases, 19 (1), 25.
  • Shin, B.-C., et al., 2012. Bee venom acupuncture for chronic low back pain: a randomised, sham-controlled, triple-blind clinical trial. European journal of integrative medicine, 4 (3), e271–e280.
  • Shin, D., Choi, W., and Bae, H., 2018. Bee venom phospholipase A2 alleviate house dust mite-induced atopic dermatitis-like skin lesions by the CD206 mannose receptor. Toxins, 10 (4), 146.
  • Sisakht, M., et al., 2017. Bee venom induces apoptosis and suppresses matrix metaloprotease-2 expression in human glioblastoma cells. Revista Brasileira de Farmacognosia, 27 (3), 324–328.
  • Son, D.J., et al., 2007. Therapeutic application of anti-arthritis, pain-releasing, and anti-cancer effects of bee venom and its constituent compounds. Pharmacology & therapeutics, 115 (2), 246–270.
  • Stuhlmeier, K.M., 2007. Apis mellifera venom and melittin block neither NF-kappa B-p50-DNA interactions nor the activation of NF-kappa B, instead they activate the transcription of proinflammatory genes and the release of reactive oxygen intermediates. Journal of immunology, 179 (1), 655–664.
  • Tu, W.-C., et al., 2008. Honeybee venom induces calcium-dependent but caspase-independent apoptotic cell death in human melanoma A2058 cells. Toxicon, 52 (2), 318–329.
  • Ulus, G., et al., 2018. The anti-angiogenic potential of (±) gossypol in comparison to suramin. Cytotechnology, 70 (6), 1537–1550.
  • Varinska, L., et al., 2010. Antiangogenic effect of selected phytochemicals. Die Pharmazie-an international journal of pharmaceutical sciences, 65, 57–63.
  • Varol, M., 2016. The importance of metal-based drugs in medicinal inorganic chemistry to improve life quality of patients. Journal of applied pharmacy, 08 (01), 2.
  • Varol, M., 2017. Angiogenesis as an important target in cancer therapies. In: A.A. Hasan Arapgirlioglu, Robert L. Elliott, and Edward Turgeon, eds. Researches on science and art in 21st century Turkey. Turkey: Gece Publishing, 1971–1981.
  • Varol, M., 2018. Anti-breast cancer and anti-angiogenic potential of a lichen-derived small-molecule: barbatolic acid. Cytotechnology, 70 (6), 1565–1573.
  • Varol, M., 2019a. Lichens as a promising source of unique and functional small molecules for human health and well-being. Studies in natural products chemistry. Elsevier, Amsterdam, 425–458.
  • Varol, M., 2019b. Parietin as an efficient and promising anti-angiogenic and apoptotic small-molecule from Xanthoria parietina. Revista Brasileira de Farmacognosia, 29 (6), 728–734.
  • Varol, M., 2020a. Natural remedies and functional foods as angiogenesis modulators. Functional foods in cancer prevention and therapy. Elsevier, Amsterdam, 1–31.
  • Varol, M., 2020b. ROS and oxidative stress in cancer: recent advances. Drug targets in cellular processes of cancer: from nonclinical to preclinical models, Springer, Singapore, 109–138.
  • Wattanakul, K., et al., 2019. Oligopeptide-side chained alginate nanocarrier for melittin-targeted chemotherapy. Polymer journal, 51 (8), 771–780.
  • Wehbe, R., et al., 2019. Bee venom: overview of main compounds and bioactivities for therapeutic interests. Molecules, 24 (16), 2997.
  • WHO, 2020. WHO report on cancer: setting priorities, investing wisely and providing care for all. World Health Organization, Geneva. https://apps.who.int/iris/handle/10665/330745.
  • Wu, X., et al., 2015. Melittin induces PTCH1 expression by down-regulating MeCP2 in human hepatocellular carcinoma SMMC-7721 cells. Toxicology and applied pharmacology, 288 (1), 74–83.
  • Yazdian-Robati, R., et al., 2019. Smart aptamer-modified calcium carbonate nanoparticles for controlled release and targeted delivery of epirubicin and melittin into cancer cells in vitro and in vivo. Drug development and industrial pharmacy, 45 (4), 603–610.
  • You, C.E., et al., 2016. Effects of emollient containing bee venom on atopic dermatitis: a double-blinded, randomized, base-controlled, multicenter study of 136 patients. Annals of dermatology, 28 (5), 593–599.
  • You, W., and Henneberg, M., 2018. Cancer incidence increasing globally: the role of relaxed natural selection. Evolutionary applications, 11 (2), 140–152.
  • Zhang, H., et al., 2014. Melittin restores PTEN expression by down-regulating HDAC2 in human hepatocelluar carcinoma HepG2 cells. PloS one, 9 (5), e95520.
  • Zhang, Z., et al., 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. Oncology letters, 11 (1), 610–618.
  • Zhu, H., et al., 2014. Melittin radiosensitizes esophageal squamous cell carcinoma with induction of apoptosis in vitro and in vivo. Tumour biology, 35 (9), 8699–8705.

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.