1,006
Views
37
CrossRef citations to date
0
Altmetric
Review Article

Targeted polymeric nanoparticles for cancer gene therapy

, , &
Pages 627-641 | Received 01 Apr 2015, Accepted 02 May 2015, Published online: 10 Jun 2015

References

  • Viola JR, El-Andaloussi S, Oprea, II, Smith CI. Non-viral nanovectors for gene delivery: factors that govern successful therapeutics. Expert Opin Drug Deliv 2010;7:721–35
  • Jafari M, Soltani M, Naahidi S, et al. Nonviral approach for targeted nucleic acid delivery. Curr Med Chem 2012;19:197–208
  • Rice J, Ottensmeier CH, Stevenson FK. DNA vaccines: precision tools for activating effective immunity against cancer. Nat Rev Cancer 2008;8:108–20
  • Zarogoulidis P, Darwiche K, Sakkas A, et al. Suicide gene therapy for cancer – current strategies. J Genet Syndr Gene Ther 2013;4:1–13 . doi: http://dx.doi.org/10.4172/2157-7412.1000139
  • Yadav S, van Vlerken LE, Little SR, Amiji MM. Evaluations of combination MDR-1 gene silencing and paclitaxel administration in biodegradable polymeric nanoparticle formulations to overcome multidrug resistance in cancer cells. Cancer Chemother Pharmacol 2009;63:711–22
  • Prabha S, Labhasetwar V. Nanoparticle-mediated wild-type p53 gene delivery results in sustained antiproliferative activity in breast cancer cells. Mol Pharm 2004;1:211–19
  • Pack DW, Hoffman AS, Pun S, Stayton PS. Design and development of polymers for gene delivery. Nat Rev Drug Discov 2005;4:581–93
  • Petros RA, DeSimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nat Rev Drug Discov 2010;9:615–27
  • Torchilin V. Tumor delivery of macromolecular drugs based on the EPR effect. Adv Drug Deliv Rev 2011;63:131–5
  • Harris JM, Martin NE, Modi M. Pegylation: a novel process for modifying pharmacokinetics. Clin Pharmacokinet 2001;40:539–51
  • van Vlerken LE, Vyas TK, Amiji MM. Poly(ethylene glycol)-modified nanocarriers for tumor-targeted and intracellular delivery. Pharm Res 2007;24:1405–14
  • Ogris M, Brunner S, Schuller S, et al. PEGylated DNA/transferrin-PEI complexes: reduced interaction with blood components, extended circulation in blood and potential for systemic gene delivery. Gene Ther 1999;6:595–605
  • Boylan NJ, Suk JS, Lai SK, et al. Highly compacted DNA nanoparticles with low MW PEG coatings: in vitro, ex vivo and in vivo evaluation. J Control Release 2012;157:72–9
  • Verma A, Stellacci F. Effect of surface properties on nanoparticle-cell interactions. Small 2010;6:12–21
  • Khan JA, Pillai B, Das TK, et al. Molecular effects of uptake of gold nanoparticles in HeLa cells. Chembiochem 2007;8:1237–40
  • Dutta D, Sundaram SK, Teeguarden JG, et al. Adsorbed proteins influence the biological activity and molecular targeting of nanomaterials. Toxicol Sci 2007;100:303–15
  • Chithrani BD, Ghazani AA, Chan WC. Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells. Nano Lett 2006;6:662–8
  • Patel PC, Giljohann DA, Daniel WL, et al. Scavenger receptors mediate cellular uptake of polyvalent oligonucleotide-functionalized gold nanoparticles. Bioconjug Chem 2010;21:2250–6
  • Li J, Ge Z, Liu S. PEG-sheddable polyplex micelles as smart gene carriers based on MMP-cleavable peptide-linked block copolymers. Chem Commun (Camb) 2013;49:6974–6
  • Zhou J, Liu J, Cheng CJ, et al. Biodegradable poly(amine-co-ester) terpolymers for targeted gene delivery. Nat Mater 2012;11:82–90
  • Peng SF, Yang MJ, Su CJ, et al. Effects of incorporation of poly(gamma-glutamic acid) in chitosan/DNA complex nanoparticles on cellular uptake and transfection efficiency. Biomaterials 2009;30:1797–808
  • Kurosaki T, Kitahara T, Fumoto S, et al. Ternary complexes of pDNA, polyethylenimine, and gamma-polyglutamic acid for gene delivery systems. Biomaterials 2009;30:2846–53
  • Green JJ, Chiu E, Leshchiner ES, et al. Electrostatic ligand coatings of nanoparticles enable ligand-specific gene delivery to human primary cells. Nano Lett 2007;7:874–9
  • Gu J, Wang X, Jiang X, et al. Self-assembled carboxymethyl poly (l-histidine) coated poly (beta-amino ester)/DNA complexes for gene transfection. Biomaterials 2012;33:644–58
  • Ito T, Yoshihara C, Hamada K, Koyama Y. DNA/polyethyleneimine/hyaluronic acid small complex particles and tumor suppression in mice. Biomaterials 2010;31:2912–18
  • Ando S, Putnam D, Pack DW, Langer R. PLGA microspheres containing plasmid DNA: preservation of supercoiled DNA via cryopreparation and carbohydrate stabilization. J Pharm Sci 1999;88:126–30
  • Hsu YY, Hao T, Hedley ML. Comparison of process parameters for microencapsulation of plasmid DNA in poly(d,l-lactic-co-glycolic) acid microspheres. J Drug Target 1999;7:313–23
  • Labhasetwar V, Bonadio J, Goldstein S, et al. A DNA controlled-release coating for gene transfer: transfection in skeletal and cardiac muscle. J Pharm Sci 1998;87:1347–50
  • Wang D, Robinson DR, Kwon GS, Samuel J. Encapsulation of plasmid DNA in biodegradable poly(d,l-lactic-co-glycolic acid) microspheres as a novel approach for immunogene delivery. J Control Release 1999;57:9–18
  • Tinsley-Bown AM, Fretwell R, Dowsett AB, et al. Formulation of poly(d,l-lactic-co-glycolic acid) microparticles for rapid plasmid DNA delivery. J Control Release 2000;66:229–41
  • Walter E, Dreher D, Kok M, et al. Hydrophilic poly(d,l-lactide-co-glycolide) microspheres for the delivery of DNA to human-derived macrophages and dendritic cells. J Control Release 2001;76:149–68
  • Benoit MA, Ribet C, Distexhe J, et al. Studies on the potential of microparticles entrapping pDNA-poly(aminoacids) complexes as vaccine delivery systems. J Drug Target 2001;9:253–66
  • Hedley ML, Curley J, Urban R. Microspheres containing plasmid-encoded antigens elicit cytotoxic T-cell responses. Nat Med 1998;4:365–8
  • Lunsford L, McKeever U, Eckstein V, Hedley ML. Tissue distribution and persistence in mice of plasmid DNA encapsulated in a PLGA-based microsphere delivery vehicle. J Drug Target 2000;8:39–50
  • Cohen H, Levy RJ, Gao J, et al. Sustained delivery and expression of DNA encapsulated in polymeric nanoparticles. Gene Ther 2000;7:1896–905
  • Fields RJ, Cheng CJ, Quijano E, et al. Surface modified poly(beta amino ester)-containing nanoparticles for plasmid DNA delivery. J Control Release 2012;164:41–8
  • Bivas-Benita M, Romeijn S, Junginger HE, Borchard G. PLGA-PEI nanoparticles for gene delivery to pulmonary epithelium. Eur J Pharm Biopharm 2004;58:1–6
  • Wagner E, Plank C, Zatloukal K, et al. Influenza virus hemagglutinin HA-2 N-terminal fusogenic peptides augment gene transfer by transferrin-polylysine-DNA complexes: toward a synthetic virus-like gene-transfer vehicle. Proc Natl Acad Sci USA 1992;89:7934–8
  • Wagner E, Cotten M, Foisner R, Birnstiel ML. Transferrin-polycation-DNA complexes: the effect of polycations on the structure of the complex and DNA delivery to cells. Proc Natl Acad Sci USA 1991;88:4255–9
  • Meyer M, Dohmen C, Philipp A, et al. Synthesis and biological evaluation of a bioresponsive and endosomolytic siRNA-polymer conjugate. Mol Pharm 2009;6:752–62
  • Rojanarata T, Opanasopit P, Techaarpornkul S, et al. Chitosan-thiamine pyrophosphate as a novel carrier for siRNA delivery. Pharm Res 2008;25:2807–14
  • Mao HQ, Roy K, Troung-Le VL, et al. Chitosan-DNA nanoparticles as gene carriers: synthesis, characterization and transfection efficiency. J Control Release 2001;70:399–421
  • Cohen JL, Schubert S, Wich PR, et al. Acid-degradable cationic dextran particles for the delivery of siRNA therapeutics. Bioconjug Chem 2011;22:1056–65
  • Cohen JA, Beaudette TT, Cohen JL, et al. Acetal-modified dextran microparticles with controlled degradation kinetics and surface functionality for gene delivery in phagocytic and non-phagocytic cells. Adv Mater 2010;22:3593–7
  • Han SE, Kang H, Shim GY, et al. Cationic derivatives of biocompatible hyaluronic acids for delivery of siRNA and antisense oligonucleotides. J Drug Target 2009;17:123–32
  • Forrest ML, Koerber JT, Pack DW. A degradable polyethylenimine derivative with low toxicity for highly efficient gene delivery. Bioconjug Chem 2003;14:934–40
  • Boussif O, Lezoualc'h F, Zanta MA, et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc Natl Acad Sci USA 1995;92:7297–301
  • Abdallah B, Hassan A, Benoist C, et al. A powerful nonviral vector for in vivo gene transfer into the adult mammalian brain: polyethylenimine. Hum Gene Ther 1996;7:1947–54
  • Mastorakos P, Kambhampati SP, Mishra MK, et al. Hydroxyl PAMAM dendrimer-based gene vectors for transgene delivery to human retinal pigment epithelial cells. Nanoscale 2015;7:3845–56
  • Chen J, Wu C, Oupicky D. Bioreducible hyperbranched poly(amido amine)s for gene delivery. Biomacromolecules 2009;10:2921–7
  • Bayele HK, Sakthivel T, O'Donell M, et al. Versatile peptide dendrimers for nucleic acid delivery. J Pharm Sci 2005;94:446–57
  • Shmueli RB, Sunshine JC, Xu Z, et al. Gene delivery nanoparticles specific for human microvasculature and macrovasculature. Nanomedicine 2012;8:1200–7
  • Guerrero-Cazares H, Tzeng SY, Young NP, et al. Biodegradable polymeric nanoparticles show high efficacy and specificity at DNA delivery to human glioblastoma in vitro and in vivo. ACS Nano 2014;8:5141–53
  • Poon Z, Lee JB, Morton SW, Hammond PT. Controlling in vivo stability and biodistribution in electrostatically assembled nanoparticles for systemic delivery. Nano Lett 2011;11:2096–103
  • Jeong GJ, Byun HM, Kim JM, et al. Biodistribution and tissue expression kinetics of plasmid DNA complexed with polyethylenimines of different molecular weight and structure. J Control Release 2007;118:118–25
  • Harris TJ, Green JJ, Fung PW, et al. Tissue-specific gene delivery via nanoparticle coating. Biomaterials 2010;31:998–1006
  • Kim J, Sunshine JC, Green JJ. Differential polymer structure tunes mechanism of cellular uptake and transfection routes of poly(beta-amino ester) polyplexes in human breast cancer cells. Bioconjug Chem 2014;25:43–51
  • Bishop CJ, Ketola TM, Tzeng SY, et al. The effect and role of carbon atoms in poly(beta-amino ester)s for DNA binding and gene delivery. J Am Chem Soc 2013;135:6951–7
  • Bhise NS, Gray RS, Sunshine JC, et al. The relationship between terminal functionalization and molecular weight of a gene delivery polymer and transfection efficacy in mammary epithelial 2-D cultures and 3-D organotypic cultures. Biomaterials 2010;31:8088–96
  • Tzeng SY, Higgins LJ, Pomper MG, Green JJ. Student award winner in the Ph.D. category for the 2013 society for biomaterials annual meeting and exposition, April 10–13, 2013, Boston, Massachusetts: biomaterial-mediated cancer-specific DNA delivery to liver cell cultures using synthetic poly(beta-amino ester)s. J Biomed Mater Res A 2013;101:1837–45
  • Wagner E, Zenke M, Cotten M, et al. Transferrin-polycation conjugates as carriers for DNA uptake into cells. Proc Natl Acad Sci USA 1990;87:3410–14
  • Kircheis R, Kichler A, Wallner G, et al. Coupling of cell-binding ligands to polyethylenimine for targeted gene delivery. Gene therapy 1997;4:409–18
  • Hildebrandt IJ, Iyer M, Wagner E, Gambhir SS. Optical imaging of transferrin targeted PEI//DNA complexes in living subjects. Gene Ther 2003;10:758–64
  • Bellocq NC, Pun SH, Jensen GS, Davis ME. Transferrin-containing, cyclodextrin polymer-based particles for tumor-targeted gene delivery. Bioconjug Chem 2003;14:1122–32
  • Davis ME, Pun SH, Bellocq NC, et al. Self-assembling nucleic acid delivery vehicles via linear, water-soluble, cyclodextrin-containing polymers. Curr Med Chem 2004;11:179–97
  • Huang R-Q, Qu Y-H, Ke W-L, et al. Efficient gene delivery targeted to the brain using a transferrin-conjugated polyethyleneglycol-modified polyamidoamine dendrimer. FASEB J 2007;21:1117–25
  • Davis ME. The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic. Mol Pharm 2009;6:659–68
  • Davis ME, Zuckerman JE, Choi CHJ, et al. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 2010;464:1067–70
  • Zhang C, Gao S, Jiang W, et al. Targeted minicircle DNA delivery using folate–poly(ethylene glycol)–polyethylenimine as non-viral carrier. Biomaterials 2010;31:6075–86
  • Lee H, Lytton-Jean AKR, Chen Y, et al. Molecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNA delivery. Nat Nanotechnol 2012;7:389–93
  • Blessing T, Kursa M, Holzhauser R, et al. Different strategies for formation of PEGylated EGF-conjugated PEI/DNA complexes for targeted gene delivery. Bioconjug Chem 2001;12:529–37
  • Klutz K, Schaffert D, Willhauck MJ, et al. Epidermal growth factor receptor-targeted (131)I-therapy of liver cancer following systemic delivery of the sodium iodide symporter gene. Mol Ther 2011;19:676–85
  • Green JJ, Chiu E, Leshchiner ES, et al. Electrostatic ligand coatings of nanoparticles enable ligand-specific gene delivery to human primary cells. Nano Lett 2007;7:874–9
  • Sakae M, Ito T, Yoshihara C, et al. Highly efficient in vivo gene transfection by plasmid/PEI complexes coated by anionic PEG derivatives bearing carboxyl groups and RGD peptide. Biomed Pharmacother 2008;62:448–53
  • Needham CJ, Williams AK, Chew SA, et al. Engineering a polymeric gene delivery vector based on poly(ethylenimine) and hyaluronic acid. Biomacromolecules 2012;13:1429–37
  • Choi KY, Jeon EJ, Yoon HY, et al. Theranostic nanoparticles based on PEGylated hyaluronic acid for the diagnosis, therapy and monitoring of colon cancer. Biomaterials 2012;33:6186–93
  • Chiu S-J, Ueno NT, Lee RJ. Tumor-targeted gene delivery via anti-HER2 antibody (trastuzumab, Herceptin®) conjugated polyethylenimine. J Control Release 2004;97:357–69
  • Moffatt S, Papasakelariou C, Wiehle S, Cristiano R. Successful in vivo tumor targeting of prostate-specific membrane antigen with a highly efficient J591//PEI//DNA molecular conjugate. Gene Ther 2006;13:761–72
  • Yao YD, Sun TM, Huang SY, et al. Targeted delivery of PLK1-siRNA by ScFv suppresses Her2+ breast cancer growth and metastasis. Sci Transl Med 2012;4:130ra48–ra000048
  • Akinc A, Querbes W, De S, et al. Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based mechanisms. Mol Ther 2010;18:1357–64
  • Danhier F, Feron O, Préat V. To exploit the tumor microenvironment: passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J Control Release 2010;148:135–46
  • Kunath K, von Harpe A, Fischer D, et al. Low-molecular-weight polyethylenimine as a non-viral vector for DNA delivery: comparison of physicochemical properties, transfection efficiency and in vivo distribution with high-molecular-weight polyethylenimine. J Control Release 2003;89:113–25
  • Ogris M, Walker G, Blessing T, et al. Tumor-targeted gene therapy: strategies for the preparation of ligand-polyethylene glycol-polyethylenimine/DNA complexes. J Control Release 2003;91:173–81
  • Ross JF, Chaudhuri PK, Ratnam M. Differential regulation of folate receptor isoforms in normal and malignant tissues in vivo and in established cell lines. Physiologic and clinical implications. Cancer 1994;73:2432–43
  • Jones HE, Gee JMW, Hutcheson IR, et al. Growth factor receptor interplay and resistance in cancer. Endocr Relat Cancer 2006;13:S45–51
  • Chen J, Zhou J, Lu J, et al. Significance of CD44 expression in head and neck cancer: a systemic review and meta-analysis. BMC Cancer 2014;14:15
  • Cheng Z, Al Zaki A, Hui JZ, et al. Multifunctional nanoparticles: cost versus benefit of adding targeting and imaging capabilities. Science 2012;338:903–10
  • Bhatia S, Menezes ME, Das SK, et al. Innovative approaches for enhancing cancer gene therapy. Discov Med 2013;15:309–17
  • Alberts B, Johnson A, Lewis J, et al. Molecular biology of the cell. New York: Garland Science; 2002
  • Maston GA, Evans SK, Green MR. Transcriptional regulatory elements in the human genome. Annu Rev Genomics Hum Genet 2006;7:29–59
  • Minn I, Menezes ME, Sarkar S, et al. Molecular-genetic imaging of cancer. Adv Cancer Res 2014;124:131–69
  • Struhl K. Yeast transcriptional regulatory mechanisms. Annu Rev Genet 1995;29:651–74
  • Lee TI, Young RA. Transcription of eukaryotic protein-coding genes. Annu Rev Genet 2000;34:77–137
  • Buratowski S. The basics of basal transcription by RNA polymerase II. Cell 1994;77:1–3
  • Farnham PJ. Insights from genomic profiling of transcription factors. Nat Rev Genet 2009;10:605–16
  • Adryan B, Teichmann SA. Computational identification of site-specific transcription factors in Drosophila. Fly 2007;1:142–5
  • Carlberg C, Molnár F. Transcription factors. Mechanisms of gene regulation. Dordrecht: Springer; 2014:55–70
  • Papadakis ED, Nicklin SA, Baker AH, White SJ. Promoters and control elements: designing expression cassettes for gene therapy. Curr Gene Ther 2004;4:89–113
  • Robson T, Hirst DG. Transcriptional targeting in cancer gene therapy. BioMed Res Int 2003;2003:110–37
  • Schaffer DV, Fidelman NA, Dan N, Lauffenburger DA. Vector unpacking as a potential barrier for receptor‐mediated polyplex gene delivery. Biotechnol Bioeng 2000;67:598–606
  • Vile RG, Hart IR. In vitro and in vivo targeting of gene expression to melanoma cells. Cancer Res 1993;53:962–7
  • Bazzoni F, Regalia E. Triggering of antitumor activity through melanoma-specific transduction of a constitutively active tumor necrosis factor (TNF) R1 chimeric receptor in the absence of TNF-α. Cancer Res 2001;61:1050–7
  • Delmas V, Beermann F, Martinozzi S, et al. β-Catenin induces immortalization of melanocytes by suppressing p16INK4a expression and cooperates with N-Ras in melanoma development. Genes Dev 2007;21:2923–35
  • Gallagher SJ, Rambow F, Kumasaka M, et al. Beta-catenin inhibits melanocyte migration but induces melanoma metastasis. Oncogene 2013;32:2230–8
  • ŠEstÁKovÁ B, Vachtenheim J. Distinct co-regulation of endogenous versus transfected MITF-dependent tyrosinase promoter. Folia Biologica (Praha) 2006;52:161–6
  • Pang S, Taneja S, Dardashti K, et al. Prostate tissue specificity of the prostate-specific antigen promoter isolated from a patient with prostate cancer. Human Gene Ther 1995;6:1417–26
  • O'Keefe DS, Uchida A, Bacich DJ, et al. Prostate‐specific suicide gene therapy using the prostate‐specific membrane antigen promoter and enhancer. Prostate 2000;45:149–57
  • Zhang J, Thomas TZ, Kasper S, Matusik RJ. A small composite probasin promoter confers high levels of prostate-specific gene expression through regulation by androgens and glucocorticoids in vitro and in vivo 1. Endocrinology 2000;141:4698–710
  • Song J, Pang S, Lu Y, et al. Gene silencing in androgen-responsive prostate cancer cells from the tissue-specific prostate-specific antigen promoter. Cancer Res 2004;64:7661–3
  • Nettelbeck DM, Jérôme V, Müller R. Gene therapy: designer promoters for tumour targeting. Trends Genet 2000;16:174–81
  • Harrington KJ, Linardakis E, Vile RG. Transcriptional control: an essential component of cancer gene therapy strategies? Adv Drug Deliv Rev 2000;44:167–84
  • Su Z-Z, Shi Y, Fisher PB. Subtraction hybridization identifies a transformation progression-associated gene PEG-3 with sequence homology to a growth arrest and DNA damage-inducible gene. Proc Natl Acad Sci USA 1997;94:9125–30
  • Su Z-Z, Sarkar D, Emdad L, et al. Targeting gene expression selectively in cancer cells by using the progression-elevated gene-3 promoter. Proc Natl Acad Sci USA 2005;102:1059–64
  • Hyo-eun CB, Gabrielson KL, Laterra J, et al. Tumor-specific imaging through progression elevated gene-3 promoter-driven gene expression. Nat Med 2011;17:123–9
  • Pan CX, Koeneman KS. A novel tumor-specific gene therapy for bladder cancer. Med Hypotheses 1999;53:130–5
  • Ambrosini G, Adida C, Altieri DC. A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med 1997;3:917–21
  • Ambrosini G, Adida C, Sirugo G, Altieri DC. Induction of apoptosis and inhibition of cell proliferation by survivin gene targeting. J Biol Chem 1998;273:11177–82
  • Lee S-G, Su Z-Z, Emdad L, et al. Astrocyte elevated gene-1 (AEG-1) is a target gene of oncogenic Ha-ras requiring phosphatidylinositol 3-kinase and c-Myc. Proc Natl Acad Sci USA 2006;103:17390–5
  • Bhatnagar A, Wang Y, Mease RC, et al. AEG-1 promoter-mediated imaging of prostate cancer. Cancer Res 2014;74:5772–81
  • Hu BG, Liu LP, Chen GG, et al. Therapeutic efficacy of improved α-fetoprotein promoter-mediated tBid delivered by folate-PEI600-cyclodextrin nanopolymer vector in hepatocellular carcinoma. Exp Cell Res 2014;324:183–91
  • Huang Y-H, Zugates GT, Peng W, et al. Nanoparticle-delivered suicide gene therapy effectively reduces ovarian tumor burden in mice. Cancer Res 2009;69:6184–91
  • Gao P, Wang R, Shen JJ, et al. Hypoxia‐inducible enhancer/α‐fetoprotein promoter‐driven RNA interference targeting STK15 suppresses proliferation and induces apoptosis in human hepatocellular carcinoma cells. Cancer Sci 2008;99:2209–17
  • Jaggar RT, Chan HY, Harris AL, Bicknell R. Endothelial cell-specific expression of tumor necrosis factor-α from the KDR or E-selectin promoters following retroviral delivery. Hum Gene Ther 1997;8:2239–47
  • Harris JD, Gutierrez AA, Hurst HC, et al. Gene therapy for cancer using tumour-specific prodrug activation. Gene Ther 1994;1:170–5
  • Parr MJ, Manome Y, Tanaka T, et al. Tumor-selective transgene expression in vivo mediated by an E2F-responsive adenoviral vector. Nat Med 1997;3:1145–9
  • Goverdhana S, Puntel M, Xiong W, et al. Regulatable gene expression systems for gene therapy applications: progress and future challenges. Mol Ther 2005;12:189–211
  • Voellmy R. Gene switches for deliberate regulation of transgene expression: recent advances in system development and uses. J Genet Syndr Gene Ther 2011;2:107
  • Makarov SS. Gene therapy for rheumatoid arthritis: preclinical studies. In Evans CH, Robbins PD, eds. Gene therapy in inflammatory diseases. Basel: Birkhäuser; 2000:13--34
  • Bateman BT, Donegan NP, Jarry TM, et al. Evaluation of a tetracycline-inducible promoter in Staphylococcus aureus in vitro and in vivo and its application in demonstrating the role of sigB in microcolony formation. Infect Immun 2001;69:7851–7
  • Anton M, Gomaa IEO, von Lukowicz T, et al. Optimization of radiation controlled gene expression by adenoviral vectors in vitro. Cancer Gene Ther 2005;12:640–6
  • Xiong J, Sun WJ, Wang WF, et al. Novel, chimeric, cancer‐specific, and radiation‐inducible gene promoters for suicide gene therapy of cancer. Cancer 2012;118:536–48
  • Kawashita Y, Ohtsuru A, Kaneda Y, et al. Regression of hepatocellular carcinoma in vitro and in vivo by radiosensitizing suicide gene therapy under the inducible and spatial control of radiation. Hum Gene Ther 1999;10:1509–19
  • Manome Y, Kunieda T, Wen PY, et al. Transgene expression in malignant glioma using a replication-defective adenoviral vector containing the Egr-1 promoter: activation by ionizing radiation or uptake of radioactive iododeoxyuridine. Hum Gene Ther 1998;9:1409–17
  • Buchholz DR. Tet-on binary systems for tissue-specific and inducible transgene expression In: Hoppler S, Vize PD, eds. Xenopus protocol: post-genomic approaches. 2nd ed. New York: Humana Press; 2012:265–75
  • Das B, Brown DD. Controlling transgene expression to study Xenopus laevis metamorphosis. Proc Natl Acad Sci USA 2004;101:4839–42
  • Smith-Arica JR, Morelli AE, Larregina AT, et al. Cell-type-specific and regulatable transgenesis in the adult brain: adenovirus-encoded combined transcriptional targeting and inducible transgene expression. Mol Ther 2000;2:579–87
  • Datta R, Rubin E, Sukhatme V, et al. Ionizing radiation activates transcription of the EGR1 gene via CArG elements. Proc Natl Acad Sci 1992;89:10149–53
  • Cooper TA, Mattox W. The regulation of splice-site selection, and its role in human disease. Am J Hum Genet 1997;61:259–66
  • Zheng Z-M. Regulation of alternative RNA splicing by exon definition and exon sequences in viral and mammalian gene expression. J Biomed Sci 2004;11:278–94
  • Hayes GM, Carpenito C, Davis PD, et al. Alternative splicing as a novel of means of regulating the expression of therapeutic genes. Cancer Gene Ther 2002;9:133–41
  • Günthert U, Hofmann M, Rudy W, et al. A new variant of glycoprotein CD44 confers metastatic potential to rat carcinoma cells. Cell 1991;65:13–24
  • Mochizuki H, Nishi T, Bruner JM, et al. Alternative splicing of neurofibromatosis type 1 gene transcript in malignant brain tumors: PCR analysis of frozen‐section mRNA. Mol Carcinog 1992;6:83–7
  • Knapinska AM, Irizarry-Barreto P, Adusumalli S, et al. Molecular mechanisms regulating mRNA stability: physiological and pathological significance. Curr Genomics 2005;6:471–86
  • Tuschl T. RNA interference and small interfering RNAs. Chembiochem 2001;2:239–45
  • Zeng Y, Yi R, Cullen BR. MicroRNAs and small interfering RNAs can inhibit mRNA expression by similar mechanisms. Proc Natl Acad Sci USA 2003;100:9779–84
  • Devi GR. siRNA-based approaches in cancer therapy. Cancer Gene Ther 2006;13:819–29
  • Grzmil M, Thelen P, Hemmerlein B, et al. Bax inhibitor-1 is overexpressed in prostate cancer and its specific down-regulation by RNA interference leads to cell death in human prostate carcinoma cells. Am J Pathol 2003;163:543–52
  • Akar U, Chaves-Reyez A, Barria M, et al. Silencing of Bcl-2 expression by small interfering RNA induces autophagic cell death in MCF-7 breast cancer cells. Autophagy 2008;4:669–79
  • Pecot CV, Wu SY, Bellister S, et al. Therapeutic silencing of KRAS using systemically delivered siRNAs. Mol Cancer Ther 2014;13:2876–85
  • Butz K, Ristriani T, Hengstermann A, et al. siRNA targeting of the viral E6 oncogene efficiently kills human papillomavirus-positive cancer cells. Oncogene 2003;22:5938–45
  • Urban-Klein B, Werth S, Abuharbeid S, et al. RNAi-mediated gene-targeting through systemic application of polyethylenimine (PEI)-complexed siRNA in vivo. Gene Ther 2005;12:461–6
  • Yokota J, Yamamoto T, Miyajima N, et al. Genetic alterations of the c-erbB-2 oncogene occur frequently in tubular adenocarcinoma of the stomach and are often accompanied by amplification of the v-erbA homologue. Oncogene 1988;2:283–7
  • Menard S, Casalini P, Campiglio M, et al. HER2 overexpression in various tumor types, focussing on its relationship to the development of invasive breast cancer. Ann Oncol 2001;12:S15–19
  • Slamon DJ, Godolphin W, Jones LA, et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 1989;244:707–12
  • Bader AG, Brown D, Winkler M. The promise of microRNA replacement therapy. Cancer Res 2010;70:7027–30
  • Barh D, Malhotra R, Ravi B, Sindhurani P. MicroRNA let-7: an emerging next-generation cancer therapeutic. Curr Oncol 2010;17:70–80
  • Welch C, Chen Y, Stallings RL. MicroRNA-34a functions as a potential tumor suppressor by inducing apoptosis in neuroblastoma cells. Oncogene 2007;26:5017–22
  • Chang T-C, Wentzel EA, Kent OA, et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell 2007;26:745–52
  • Li N, Fu H, Tie Y, et al. miR-34a inhibits migration and invasion by down-regulation of c-Met expression in human hepatocellular carcinoma cells. Cancer Lett 2009;275:44–53
  • Chen Y, Zhu X, Zhang X, et al. Nanoparticles modified with tumor-targeting scFv deliver siRNA and miRNA for cancer therapy. Mol Ther 2010;18:1650–6
  • Ohno S-i, Takanashi M, Sudo K, et al. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol Ther 2013;21:185–91
  • Zhao XD, Zhang W, Liang HJ, Ji WY. Overexpression of miR-155 promotes proliferation and invasion of human laryngeal squamous cell carcinoma via targeting SOCS1 and STAT3. PLoS One 2013;8:e56395
  • Wang Z, Rao DD, Senzer N, Nemunaitis J. RNA interference and cancer therapy. Pharm Res 2011;28:2983–95
  • Meric F, Hunt KK. Translation initiation in cancer: a novel target for therapy 1 FM is supported by The University of Texas MD Anderson Cancer Center Physician-Scientist Program and by NIH Grant 1KO8-CA 91895-01. KKH is supported by Department of Defense Award DAMD-17-97-1-7162. 1. Mol Cancer Ther 2002;1:971–9
  • Darveau A, Pelletier J, Sonenberg N. Differential efficiencies of in vitro translation of mouse c-myc transcripts differing in the 5′untranslated region. Proc Natl Acad Sci USA 1985;82:2315–19
  • DeFatta RJ, Li Y, De Benedetti A. Selective killing of cancer cells based on translational control of a suicide gene. Cancer Gene Ther 2002;9:573–8
  • Koromilas AE, Lazaris-Karatzas A, Sonenberg N. mRNAs containing extensive secondary structure in their 5'non-coding region translate efficiently in cells overexpressing initiation factor eIF-4E. EMBO J 1992;11:4153
  • Crew JP, Fuggle S, Bicknell R, et al. Eukaryotic initiation factor-4E in superficial and muscle invasive bladder cancer and its correlation with vascular endothelial growth factor expression and tumour progression. Br J Cancer 2000;82:161–6
  • Kerekatte V, Smiley K, Hu B, et al. The proto‐oncogene/translation factor eIF4E: a survey of its expression in breast carcinomas. Int J Cancer 1995;64:27–31
  • Wang R, Geng J, Wang JH, et al. Overexpression of eukaryotic initiation factor 4E (eIF4E) and its clinical significance in lung adenocarcinoma. Lung Cancer 2009;66:237–44
  • Kevil C, Carter P, Hu B, DeBenedetti A. Translational enhancement of FGF-2 by eIF-4 factors, and alternate utilization of CUG and AUG codons for translation initiation. Oncogene 1995;11:2339–48
  • Wang Z, Sun Y. Targeting p53 for novel anticancer therapy. Transl Oncol 2010;3:1–12
  • Roth JA. Adenovirus p53 gene therapy. Expert Opin Biol Ther 2006;6:55–61
  • Lemke J, Von Karstedt S, Zinngrebe J, Walczak H. Getting TRAIL back on track for cancer therapy. Cell Death Differ 2014;21:1350–64
  • Walczak H, Miller RE, Ariail K, et al. Tumoricidal activity of tumor necrosis factor–related apoptosis–inducing ligand in vivo. Nat Med 1999;5:157–63
  • Ashkenazi A. Directing cancer cells to self-destruct with pro-apoptotic receptor agonists. Nat Rev Drug Discov 2008;7:1001–12
  • Komarova S, Kawakami Y, Stoff-Khalili MA, et al. Mesenchymal progenitor cells as cellular vehicles for delivery of oncolytic adenoviruses. Mol Cancer Ther 2006;5:755–66
  • Studeny M, Marini FC, Champlin RE, et al. Bone marrow-derived mesenchymal stem cells as vehicles for interferon-β delivery into tumors. Cancer Res 2002;62:3603–8
  • Aboody KS, Brown A, Rainov NG, et al. Neural stem cells display extensive tropism for pathology in adult brain: evidence from intracranial gliomas. Proc Natl Acad Sci USA 2000;97:12846–51
  • Lombardo Y, Scopelliti A, Cammareri P, et al. Bone morphogenetic protein 4 induces differentiation of colorectal cancer stem cells and increases their response to chemotherapy in mice. Gastroenterology 2011;140:297–309
  • Zhang L, Sun H, Zhao F, et al. BMP4 administration induces differentiation of CD133+ hepatic cancer stem cells, blocking their contributions to hepatocellular carcinoma. Cancer Res 2012;72:4276–85
  • Piccirillo SGM, Reynolds BA, Zanetti N, et al. Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature 2006;444:761–5
  • Li Q, Wijesekera O, Salas SJ, et al. Mesenchymal stem cells from human fat engineered to secrete BMP4 are nononcogenic, suppress brain cancer, and prolong survival. Clin Cancer Res 2014;20:2375–87
  • Hanna N, Ohana P, Konikoff FM, et al. Phase 1/2a, dose-escalation, safety, pharmacokinetic and preliminary efficacy study of intratumoral administration of BC-819 in patients with unresectable pancreatic cancer. Cancer Gene Ther 2012;19:374–81
  • Buscail L, Bournet B, Vernejoul F, et al. First-in-man phase I clinical trial of gene therapy for advanced pancreatic cancer: safety, biodistribution and preliminary clinical findings. Mol Ther 2015;23:779–89
  • Alvarez RD, Sill MW, Davidson SA, et al. A phase II trial of intraperitoneal EGEN-001, an IL-12 plasmid formulated with PEG–PEI–cholesterol lipopolymer in the treatment of persistent or recurrent epithelial ovarian, fallopian tube or primary peritoneal cancer: A Gynecologic Oncology Group study. Gynecol Oncol 2014;133:433–8
  • Francis SM, Taylor CA, Tang T, et al. SNS01-T modulation of eIF5A inhibits B-cell cancer progression and synergizes with bortezomib and lenalidomide. Mol Ther 2014;22:1643–52
  • Knop K, Hoogenboom R, Fischer D, Schubert US. Poly(ethylene glycol) in drug delivery: pros and cons as well as potential alternatives. Angew Chem (International ed in English) 2010;49:6288–308
  • Yin H, Kanasty RL, Eltoukhy AA, et al. Non-viral vectors for gene-based therapy. Nat Rev Genet 2014;15:541–55

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.