Selective suppression of tumor cells by a tumor-specific bicistronic lentiviral vector

Selective suppression of tumor cells by a tumor-specific bicistronic lentiviral vector

Lentiviral gene delivery is now considered as a major candidate in the future of cancer therapy. To avoid common side-effects associated with methods of cancer therapy, the survivin promoter shows great promise due to its high expression level in multiple cancers. In this research, using a 2A peptide, Noxa was coexpressed with hemagglutinin neuraminidase (HN) driven by the survivin promoter (Surp). Coding genes of Noxa and HN were connected to the upstream and downstream of a 2A peptide using SOE PCR. After cloning of Noxa-2A-HN and Surp into the lentiviral vector, the final construct was transfected to SK-BR3 tumor cells and human embryonic kidney 293T cells. Quantitative real-time PCR implied efficient transfection and integration of the lentiviral vector in target cells. Western blotting analysis was performed to verify the expression of Noxa and HN and we determined cell viability by MTT calorimetric assay after transfection. Our results showed that the expression of Noxa and HN, though not utterly specific, was relatively higher in tumor cells and led to the selective suppression of tumor cells, which eventually resulted in a cell survival reduction of 76%. In conclusion, the novel pLN2AH-Surp lentiviral vector proved the potential for further in vivo and clinical experiments.

___

  • Albert MC, Brinkmann K, Kashkar H (2014). Noxa and cancer therapy. Mol Cell Oncol 1: e29906. Ambrosini G, Adida CF, Altieri DC (1997). A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med 3: 917-921.
  • Bao R, Connolly DC, Murphy M, Green J, Weinstein JK, Pisarcik DA, Hamilton TC (2002). Activation of cancer-specific gene expression by the survivin promoter. J Natl Canc Inst 94: 522- 528.
  • Chen JS, Liu JC, Shen L, Rau KM, Kuo HP, Li YM, Shi D, Lee YC, Chang KJ, Hung MC (2004). Cancer-specific activation of the survivin promoter and its potential use in gene therapy. Cancer Gene Ther 11: 740-747.
  • de Felipe P, Martín V, Cortés ML, Ryan M, Izquierdo M (1999). Use of the 2A sequence from foot-and-mouth disease virus in the generation of retroviral vectors for gene therapy. Gene Ther 6: 198-208
  • Dong D, Gao J, Sun Y, Long Y, Li M, Zhang D, Gong J, Xu L, Li L, Qin S et al. (2015). Adenovirus-mediated co-expression of the TRAIL and HN genes inhibits growth and induces apoptosis in Marek’s disease tumor cell line MSB-1. Cancer Cell Int 15: 20.
  • Durocher Y, Perret S, Kamen A (2002). High-level and highthroughput recombinant protein production by transient transfection of suspension-growing human 293-EBNA1 cells. Nucleic Acids Res 30: E9.
  • He D, Sun L, Li C, Hu N, Sheng Y, Chen Z, Li X, Chi B, Jin N (2014). Anti-tumor effects of an oncolytic adenovirus expressing hemagglutinin-neuraminidase of Newcastle disease virus in vitro and in vivo. Viruses 6: 856-874.
  • Ji H, Chi B, He D, Li D, Hu N, Wang K, Sheng Y, Wang H, Jin N (2013). Antitumor effects of Newcastle disease virus hemagglutininneuraminidase used as a molecular adjuvant. Chem Res Chinese U 29: 270-274.
  • Kafri T, Blömer U, Peterson DA, Gage FH, Verma IM (1997). Sustained expression of genes delivered directly into liver and muscle by lentiviral vectors. Nat Genet 17: 314-317.
  • Kafri T, Praag HV, Gage FH, Verma IM (2000). Lentiviral vectors: regulated gene expression. Mol Ther 1: 516-521.
  • Levine BL, Humeau LM, Boyer J, MacGregor RR, Rebello T, Lu X, Binder GK, Slepushkin V, Lemiale F, Mascola JR et al. (2006). Gene transfer in humans using a conditionally replicating lentiviral vector. P Natl Acad Sci USA 103: 17372-17377.
  • Li M, Wu Y, Qiu Y, Yao Z, Liu S, Liu Y, Shi J, Zheng D (2012). 2A peptide-based, lentivirus-mediated anti-death receptor 5 chimeric antibody expression prevents tumor growth in nude mice. Mol Ther 20: 46-53.
  • Liechtenstein T, Perez-Janices N, Escors D (2013). Lentiviral vectors for cancer immunotherapy and clinical applications. Cancers (Basel) 5: 815-837.
  • Luke GA (2012). Translating 2A Research into Practice: Innovations in Biotechnology. 1st ed. Rijeka, Croatia: InTech.
  • Mao Y, Yan R, Li A, Zhang Y, Li J, Du H, Chen B, Wei W, Zhang Y, Sumners C et al. (2015). Lentiviral Vectors mediate long-term and high efficiency transgene expression in HEK 293T cells. Int J Med Sci 12: 407-415.
  • Minskaia E, Ryan MD (2013). Protein coexpression using FMDV 2A: effect of “linker” residues. Biomed Res Int 2013: 12.
  • Miyoshi H, Takahashi M, Gage FH, Verma IM (1997). Stable and efficient gene transfer into the retina using an HIV-based lentiviral vector. P Natl Acad Sci 94: 10319-10323.
  • Nakajima W, Tanaka N (2011). Noxa induces apoptosis in oncogeneexpressing cells through catch-and-release mechanism operating between Puma and Mcl-1. Biochem Biophys Res Commun 413: 643-648.
  • Naldini L, Blömer U, Gage FH, Trono D, Verma IM (1996). Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector. P Natl Acad Sci 93: 11382-11388.
  • Ni J, Galani IE, Cerwenka A, Schirrmacher V, Fournier P (2011). Antitumor vaccination by Newcastle disease virus hemagglutinin-neuraminidase plasmid DNA application: changes in tumor microenvironment and activation of innate anti-tumor immunity. Vaccine 29: 1185-1193.
  • Ni J, Schirrmacher V, Fournier P (2010). The hemagglutininneuraminidase gene of Newcastle disease virus: a powerful molecular adjuvant for DNA anti-tumor vaccination. Vaccine 28: 6891-6900.
  • Salmon P, Trono D (2007). Production and titration of lentiviral vectors. Curr Protoc Hum Genet 12: 10.
  • Shibue T, Suzuki S, Okamoto H, Yoshida H, Ohba Y, Takaoka A, Taniguchi T (2006). Differential contribution of Puma and Noxa in dual regulation of p53-mediated apoptotic pathways. EMBO J 25: 4952-4962.
  • Shibue T, Takeda K, Oda E, Tanaka H, Murasawa H, Takaoka A, Morishita Y, Akira S, Taniguchi T, Tanaka N (2003). Integral role of Noxa in p53-mediated apoptotic response. Genes Dev 17: 2233-2238.
  • Suzuki S, Nakasato M, Shibue T, Koshima I, Taniguchi T (2009). Therapeutic potential of proapoptotic molecule Noxa in the selective elimination of tumor cells. Cancer Sci 100: 759-769.
  • Wang R, Wang X, Li B, Lin F, Dong K, Gao P, Zhang HZ (2009). Tumor-specific adenovirus-mediated PUMA gene transfer using the survivin promoter enhances radiosensitivity of breast cancer cells in vitro and in vivo. Breast Cancer Res Treat 117: 45-54.
  • Yamamoto T, Tanigawa N (2001). The role of survivin as a new target of diagnosis and treatment in human cancer. Med Electron Microsc 34: 207-212.
  • Yang B, Jeang J, Yang A, Wu T, Hung CF (2014). DNA vaccine for cancer immunotherapy. Hum Vaccin Immunother 10: 3153- 3164.
  • Yang S, Cohen CJ, Peng PD, Zhao Y, Cassard L, Yu Z, Zheng Z, Jones S, Restifo NP, Rosenberg SA et al. (2008). Development of optimal bicistronic lentiviral vectors facilitates high-level TCR gene expression and robust tumor cell recognition. Gene Ther 15: 1411-1423.
  • Zhang LN, Li JY, Xu W (2013). A review of the role of Puma, Noxa and Bim in the tumorigenesis, therapy and drug resistance of chronic lymphocytic leukemia. Cancer Gene Ther 20: 1-7.
  • Zhao J, Pettigrew GJ, Thomas J, Vandenberg JI, Delriviere L, Bolton EM, Carmichael A, Martin JL, Marber MS, Lever AM (2002). Lentiviral vectors for delivery of genes into neonatal and adult ventricular cardiac myocytes in vitro and in vivo. Basic Res Cardiol 97: 348-358.