Skip to main content

Hepatic Gene Therapy

  • Chapter
  • First Online:
Molecular Pathology of Liver Diseases

Part of the book series: Molecular Pathology Library ((MPLB,volume 5))

Abstract

Around the turn of the new millennium, after its 20-year history, gene therapy experienced both an exciting success [1] and an unexpected failure [2, 3], which significantly changed our perspectives on gene therapy from “versatile therapy” that we expected would soon become available to cure difficult-to-treat diseases to “potentially effective therapy” that would surely be superior over conventional therapies, but still needs further refinement towards clinical applications. Since then, within less than a decade, exciting discoveries and the development of emerging technologies pertinent to gene therapy have occurred, re-inspiring a greater-than-ever interest to gene delivery approaches. As for hepatic gene transfer, we have already become able to deliver genes of interest to target cells in the liver at extremely high efficiency and with minimum toxicity, at least in mice. With the contemporary hepatic gene delivery methods in our hands, any disease can be effectively treated or even cured in animal models, as long as the right therapeutic targets have been identified in the liver. At the current stage of the development of new molecular therapeutics, in addition to seeking new breakthroughs, it is critical to further refine the technologies, to understand the underlying mechanisms of action, to explore the methods to minimize undesired reactions and side effects, and importantly to further the knowledge of disease pathogenesis. With this introduction, this chapter provides an overview of contemporary methods and applications of hepatic gene transfer with an emphasis on the underlying mechanisms of action of each gene delivery approach.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 299.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile G, et al. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science. 2000;288(5466):669–72.

    PubMed  CAS  Google Scholar 

  2. Raper SE, Chirmule N, Lee FS, et al. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab. 2003;80(1–2):148–58.

    PubMed  CAS  Google Scholar 

  3. Hacein-Bey-Abina S, Von Kalle C, Schmidt M, et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science. 2003;302(5644):415–9.

    PubMed  CAS  Google Scholar 

  4. Ferber S, Halkin A, Cohen H, et al. Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia. Nat Med. 2000;6(5):568–72.

    PubMed  CAS  Google Scholar 

  5. Liu F, Song Y, Liu D. Hydrodynamics-based transfection in animals by systemic administration of plasmid DNA. Gene Ther. 1999;6(7):1258–66.

    PubMed  CAS  Google Scholar 

  6. Zhang G, Budker V, Wolff JA. High levels of foreign gene expression in hepatocytes after tail vein injections of naked plasmid DNA. Hum Gene Ther. 1999;10(10):1735–7.

    PubMed  CAS  Google Scholar 

  7. Lau AH, Thomson AW. Dendritic cells and immune regulation in the liver. Gut. 2003;52(2):307–14.

    PubMed  CAS  Google Scholar 

  8. Manno CS, Pierce GF, Arruda VR, et al. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat Med. 2006;12(3):342–7.

    PubMed  CAS  Google Scholar 

  9. Raper SE, Yudkoff M, Chirmule N, et al. A pilot study of in vivo liver-directed gene transfer with an adenoviral vector in partial ornithine transcarbamylase deficiency. Hum Gene Ther. 2002;13(1):163–75.

    PubMed  CAS  Google Scholar 

  10. Di Paolo NC, Shayakhmetov DM. Immune responses to adenoviral vectors. In: Herzog RW, editor. Gene therapy immunology. Hoboken, NJ: Wiley-Blackwell; 2009. p. 57–84.

    Google Scholar 

  11. Yamaguchi T, Kawabata K, Koizumi N, et al. Role of MyD88 and TLR9 in the innate immune response elicited by serotype 5 adenoviral vectors. Hum Gene Ther. 2007;18(8):753–62.

    PubMed  CAS  Google Scholar 

  12. Di Paolo NC, Miao EA, Iwakura Y, et al. Virus binding to a plasma membrane receptor triggers interleukin-1 alpha-mediated proinflammatory macrophage response in vivo. Immunity. 2009;31(1):110–21.

    PubMed  Google Scholar 

  13. Cao O, Furlan-Freguia C, Arruda VR, Herzog RW. Emerging role of regulatory T cells in gene transfer. Curr Gene Ther. 2007;7(5):381–90.

    PubMed  CAS  Google Scholar 

  14. Mingozzi F, Hasbrouck NC, Basner-Tschakarjan E, et al. Modulation of tolerance to the transgene product in a nonhuman primate model of AAV-mediated gene transfer to liver. Blood. 2007;110(7):2334–41.

    PubMed  CAS  Google Scholar 

  15. LoDuca PA, Hoffman BE, Herzog RW. Hepatic gene transfer as a means of tolerance induction to transgene products. Curr Gene Ther. 2009;9(2):104–14.

    PubMed  CAS  Google Scholar 

  16. Luth S, Huber S, Schramm C, et al. Ectopic expression of neural autoantigen in mouse liver suppresses experimental autoimmune neuroinflammation by inducing antigen-specific Tregs. J Clin Invest. 2008;118(10):3403–10.

    PubMed  Google Scholar 

  17. Stone D, Liu Y, Shayakhmetov D, Li ZY, Ni S, Lieber A. Adenovirus-platelet interaction in blood causes virus sequestration to the reticuloendothelial system of the liver. J Virol. 2007;81(9):4866–71.

    PubMed  CAS  Google Scholar 

  18. Snoeys J, Lievens J, Wisse E, et al. Species differences in transgene DNA uptake in hepatocytes after adenoviral transfer correlate with the size of endothelial fenestrae. Gene Ther. 2007;14(7):604–12.

    PubMed  CAS  Google Scholar 

  19. Leopold PL, Crystal RG. Intracellular trafficking of adenovirus: many means to many ends. Adv Drug Deliv Rev. 2007;59(8):810–21.

    PubMed  CAS  Google Scholar 

  20. 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 U S A. 1995;92(16):7297–301.

    PubMed  CAS  Google Scholar 

  21. Thomas CE, Storm TA, Huang Z, Kay MA. Rapid uncoating of vector genomes is the key to efficient liver transduction with pseudotyped adeno-associated virus vectors. J Virol. 2004;78(6):3110–22.

    PubMed  CAS  Google Scholar 

  22. Zhong L, Zhou X, Li Y, et al. Single-polarity recombinant adeno-associated virus 2 vector-mediated transgene expression in vitro and in vivo: mechanism of transduction. Mol Ther. 2008;16(2):290–5.

    PubMed  CAS  Google Scholar 

  23. Chen ZY, He CY, Meuse L, Kay MA. Silencing of episomal transgene expression by plasmid bacterial DNA elements in vivo. Gene Ther. 2004;11(10):856–64.

    PubMed  CAS  Google Scholar 

  24. Garrison BS, Yant SR, Mikkelsen JG, Kay MA. Postintegrative gene silencing within the Sleeping Beauty transposition system. Mol Cell Biol. 2007;27(24):8824–33.

    PubMed  CAS  Google Scholar 

  25. Schiedner G, Morral N, Parks RJ, et al. Genomic DNA transfer with a high-capacity adenovirus vector results in improved in vivo gene expression and decreased toxicity. Nat Genet. 1998;18(2):180–3.

    PubMed  CAS  Google Scholar 

  26. Brown BD, Shi CX, Powell S, Hurlbut D, Graham FL, Lillicrap D. Helper-dependent adenoviral vectors mediate therapeutic factor VIII expression for several months with minimal accompanying toxicity in a canine model of severe hemophilia A. Blood. 2004;103(3):804–10.

    PubMed  CAS  Google Scholar 

  27. Chuah MK, Schiedner G, Thorrez L, et al. Therapeutic factor VIII levels and negligible toxicity in mouse and dog models of hemophilia A following gene therapy with high-capacity adenoviral vectors. Blood. 2003;101(5):1734–43.

    PubMed  CAS  Google Scholar 

  28. Ehrhardt A, Xu H, Dillow AM, Bellinger DA, Nichols TC, Kay MA. A gene-deleted adenoviral vector results in phenotypic correction of canine hemophilia B without liver toxicity or thrombocytopenia. Blood. 2003;102(7):2403–11.

    PubMed  CAS  Google Scholar 

  29. McCormack Jr WM, Seiler MP, Bertin TK, et al. Helper-dependent adenoviral gene therapy mediates long-term correction of the clotting defect in the canine hemophilia A model. J Thromb Haemost. 2006;4(6):1218–25.

    PubMed  CAS  Google Scholar 

  30. Balague C, Zhou J, Dai Y, et al. Sustained high-level expression of full-length human factor VIII and restoration of clotting activity in hemophilic mice using a minimal adenovirus vector. Blood. 2000;95(3):820–8.

    PubMed  CAS  Google Scholar 

  31. Kiang A, Hartman ZC, Liao S, et al. Fully deleted adenovirus persistently expressing GAA accomplishes long-term skeletal muscle glycogen correction in tolerant and nontolerant GSD-II mice. Mol Ther. 2006;13(1):127–34.

    PubMed  CAS  Google Scholar 

  32. Koeberl DD, Sun B, Bird A, Chen YT, Oka K, Chan L. Efficacy of helper-dependent adenovirus vector-mediated gene therapy in murine glycogen storage disease type Ia. Mol Ther. 2007;15(7):1253–8.

    PubMed  CAS  Google Scholar 

  33. Mian A, McCormack Jr WM, Mane V, et al. Long-term correction of ornithine transcarbamylase deficiency by WPRE-mediated overexpression using a helper-dependent adenovirus. Mol Ther. 2004;10(3):492–9.

    PubMed  CAS  Google Scholar 

  34. Nomura S, Merched A, Nour E, Dieker C, Oka K, Chan L. Low-density lipoprotein receptor gene therapy using helper-dependent adenovirus produces long-term protection against atherosclerosis in a mouse model of familial hypercholesterolemia. Gene Ther. 2004;11(20):1540–8.

    PubMed  CAS  Google Scholar 

  35. Smith T, Idamakanti N, Kylefjord H, et al. In vivo hepatic adenoviral gene delivery occurs independently of the coxsackievirus-adenovirus receptor. Mol Ther. 2002;5(6):770–9.

    PubMed  CAS  Google Scholar 

  36. Waddington SN, McVey JH, Bhella D, et al. Adenovirus serotype 5 hexon mediates liver gene transfer. Cell. 2008;132(3):397–409.

    PubMed  CAS  Google Scholar 

  37. Tao N, Gao GP, Parr M, et al. Sequestration of adenoviral vector by Kupffer cells leads to a nonlinear dose response of transduction in liver. Mol Ther. 2001;3(1):28–35.

    PubMed  CAS  Google Scholar 

  38. Parks RJ, Bramson JL, Wan Y, Addison CL, Graham FL. Effects of stuffer DNA on transgene expression from helper-dependent adenovirus vectors. J Virol. 1999;73(10):8027–34.

    PubMed  CAS  Google Scholar 

  39. Brunetti-Pierri N, Stapleton GE, Palmer DJ, et al. Pseudo-hydrodynamic delivery of helper-dependent adenoviral vectors into non-human primates for liver-directed gene therapy. Mol Ther. 2007;15(4):732–40.

    PubMed  CAS  Google Scholar 

  40. Gao GP, Alvira MR, Wang L, Calcedo R, Johnston J, Wilson JM. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc Natl Acad Sci U S A. 2002;99(18):11854–9.

    PubMed  CAS  Google Scholar 

  41. Nakai H, Fuess S, Storm TA, Muramatsu S, Nara Y, Kay MA. Unrestricted hepatocyte transduction with adeno-associated virus serotype 8 vectors in mice. J Virol. 2005;79(1):214–24.

    PubMed  CAS  Google Scholar 

  42. McEachern KA, Nietupski JB, Chuang WL, et al. AAV8-mediated expression of glucocerebrosidase ameliorates the storage pathology in the visceral organs of a mouse model of Gaucher disease. J Gene Med. 2006;8(6):719–29.

    PubMed  CAS  Google Scholar 

  43. Ziegler RJ, Cherry M, Barbon CM, et al. Correction of the biochemical and functional deficits in fabry mice following AAV8-mediated hepatic expression of alpha-galactosidase A. Mol Ther. 2007;15(3):492–500.

    PubMed  CAS  Google Scholar 

  44. Barbon CM, Ziegler RJ, Li C, et al. AAV8-mediated hepatic expression of acid sphingomyelinase corrects the metabolic defect in the visceral organs of a mouse model of Niemann-Pick disease. Mol Ther. 2005;12(3):431–40.

    PubMed  CAS  Google Scholar 

  45. Harding CO, Gillingham MB, Hamman K, et al. Complete correction of hyperphenylalaninemia following liver-directed, recombinant AAV2/8 vector-mediated gene therapy in murine phenylketonuria. Gene Ther. 2006;13(5):457–62.

    PubMed  CAS  Google Scholar 

  46. Mount JD, Herzog RW, Tillson DM, et al. Sustained phenotypic correction of hemophilia B dogs with a factor IX null mutation by liver-directed gene therapy. Blood. 2002;99(8):2670–6.

    PubMed  CAS  Google Scholar 

  47. Nathwani AC, Gray JT, McIntosh J, et al. Safe and efficient transduction of the liver after peripheral vein infusion of self-complementary AAV vector results in stable therapeutic expression of human FIX in nonhuman primates. Blood. 2007;109(4):1414–21.

    PubMed  CAS  Google Scholar 

  48. Sarkar R, Mucci M, Addya S, et al. Long-term efficacy of adeno-associated virus serotypes 8 and 9 in hemophilia a dogs and mice. Hum Gene Ther. 2006;17(4):427–39.

    PubMed  CAS  Google Scholar 

  49. Sarkar R, Tetreault R, Gao G, et al. Total correction of hemophilia A mice with canine FVIII using an AAV 8 serotype. Blood. 2004;103(4):1253–60.

    PubMed  CAS  Google Scholar 

  50. Chen CC, Ko TM, Ma HI, et al. Long-term inhibition of hepatitis B virus in transgenic mice by double-stranded adeno-associated virus 8-delivered short hairpin RNA. Gene Ther. 2007;14(1):11–9.

    PubMed  Google Scholar 

  51. Grimm D, Streetz KL, Jopling CL, et al. Fatality in mice due to oversaturation of cellular microRNA/short hairpin RNA pathways. Nature. 2006;441(7092):537–41.

    PubMed  CAS  Google Scholar 

  52. Niemeyer GP, Herzog RW, Mount J, et al. Long-term correction of inhibitor-prone hemophilia B dogs treated with liver-directed AAV2-mediated factor IX gene therapy. Blood. 2009;113(4):797–806.

    PubMed  CAS  Google Scholar 

  53. Snyder RO, Miao C, Meuse L, et al. Correction of hemophilia B in canine and murine models using recombinant adeno-associated viral vectors. Nat Med. 1999;5(1):64–70.

    PubMed  CAS  Google Scholar 

  54. Wang L, Takabe K, Bidlingmaier SM, Charles III R, Verma IM. Sustained correction of bleeding disorder in hemophilia B mice by gene therapy. Proc Natl Acad Sci U S A. 1999;96(7):3906–10.

    PubMed  CAS  Google Scholar 

  55. Zhong L, Li B, Mah CS, et al. Next generation of adeno-associated virus 2 vectors: point mutations in tyrosines lead to high-efficiency transduction at lower doses. Proc Natl Acad Sci U S A. 2008;105(22):7827–32.

    PubMed  CAS  Google Scholar 

  56. Akache B, Grimm D, Pandey K, Yant SR, Xu H, Kay MA. The 37/67-kilodalton laminin receptor is a receptor for adeno-associated virus serotypes 8, 2, 3, and 9. J Virol. 2006;80(19):9831–6.

    PubMed  CAS  Google Scholar 

  57. McCarty DM, Monahan PE, Samulski RJ. Self-complementary recombinant adeno-associated virus (scAAV) vectors promote efficient transduction independently of DNA synthesis. Gene Ther. 2001;8(16):1248–54.

    PubMed  CAS  Google Scholar 

  58. McCarty DM, Fu H, Monahan PE, Toulson CE, Naik P, Samulski RJ. Adeno-associated virus terminal repeat (TR) mutant generates self-complementary vectors to overcome the rate-limiting step to transduction in vivo. Gene Ther. 2003;10(26):2112–8.

    PubMed  CAS  Google Scholar 

  59. Wang Z, Ma HI, Li J, Sun L, Zhang J, Xiao X. Rapid and highly efficient transduction by double-stranded adeno-associated virus vectors in vitro and in vivo. Gene Ther. 2003;10(26):2105–11.

    PubMed  CAS  Google Scholar 

  60. Nakai H, Yant SR, Storm TA, Fuess S, Meuse L, Kay MA. Extrachromosomal recombinant adeno-associated virus vector genomes are primarily responsible for stable liver transduction in vivo. J Virol. 2001;75(15):6969–76.

    PubMed  CAS  Google Scholar 

  61. Chen ZY, He CY, Ehrhardt A, Kay MA. Minicircle DNA vectors devoid of bacterial DNA result in persistent and high-level transgene expression in vivo. Mol Ther. 2003;8(3):495–500.

    PubMed  CAS  Google Scholar 

  62. Mingozzi F, Liu YL, Dobrzynski E, et al. Induction of immune tolerance to coagulation factor IX antigen by in vivo hepatic gene transfer. J Clin Invest. 2003;111(9):1347–56.

    PubMed  CAS  Google Scholar 

  63. Nakai H, Montini E, Fuess S, Storm TA, Grompe M, Kay MA. AAV serotype 2 vectors preferentially integrate into active genes in mice. Nat Genet. 2003;34(3):297–302.

    PubMed  CAS  Google Scholar 

  64. Inagaki K, Lewis SM, Wu X, et al. DNA palindromes with a modest arm length of greater, similar 20 base pairs are a significant target for recombinant adeno-associated virus vector integration in the liver, muscles, and heart in mice. J Virol. 2007;81(20):11290–303.

    PubMed  CAS  Google Scholar 

  65. Inagaki K, Piao C, Kotchey NM, Wu X, Nakai H. Frequency and spectrum of genomic integration of recombinant adeno-associated virus serotype 8 vector in neonatal mouse liver. J Virol. 2008;82(19):9513–24.

    PubMed  CAS  Google Scholar 

  66. Donsante A, Miller DG, Li Y, et al. AAV vector integration sites in mouse hepatocellular carcinoma. Science. 2007;317(5837):477.

    PubMed  CAS  Google Scholar 

  67. Bell P, Wang L, Lebherz C, et al. No evidence for tumorigenesis of AAV vectors in a large-scale study in mice. Mol Ther. 2005;12(2):299–306.

    PubMed  CAS  Google Scholar 

  68. Chowdhury JR, Grossman M, Gupta S, Chowdhury NR, Baker Jr JR, Wilson JM. Long-term improvement of hypercholesterolemia after ex vivo gene therapy in LDLR-deficient rabbits. Science. 1991;254(5039):1802–5.

    PubMed  CAS  Google Scholar 

  69. Grossman M, Raper SE, Kozarsky K, et al. Successful ex vivo gene therapy directed to liver in a patient with familial hypercholesterolaemia. Nat Genet. 1994;6(4):335–41.

    PubMed  CAS  Google Scholar 

  70. Ma X, Liu Y, Tittiger M, et al. Improvements in mucopolysaccharidosis I mice after adult retroviral vector-mediated gene therapy with immunomodulation. Mol Ther. 2007;15(5):889–902.

    PubMed  CAS  Google Scholar 

  71. Powell JS, Ragni MV, White II GC, et al. Phase 1 trial of FVIII gene transfer for severe hemophilia A using a retroviral construct administered by peripheral intravenous infusion. Blood. 2003;102(6):2038–45.

    PubMed  CAS  Google Scholar 

  72. Ponder KP, Haskins ME. Gene therapy for mucopolysaccharidosis. Expert Opin Biol Ther. 2007;7(9):1333–45.

    PubMed  CAS  Google Scholar 

  73. Traas AM, Wang P, Ma X, et al. Correction of clinical manifestations of canine mucopolysaccharidosis I with neonatal retroviral vector gene therapy. Mol Ther. 2007;15(8):1423–31.

    PubMed  CAS  Google Scholar 

  74. Mango RL, Xu L, Sands MS, et al. Neonatal retroviral vector-mediated hepatic gene therapy reduces bone, joint, and cartilage disease in mucopolysaccharidosis VII mice and dogs. Mol Genet Metab. 2004;82(1):4–19.

    PubMed  CAS  Google Scholar 

  75. Bellodi-Privato M, Aubert D, Pichard V, Myara A, Trivin F, Ferry N. Successful gene therapy of the Gunn rat by in vivo neonatal hepatic gene transfer using murine oncoretroviral vectors. Hepatology. 2005;42(2):431–8.

    PubMed  CAS  Google Scholar 

  76. Bosch A, McCray Jr PB, Chang SM, et al. Proliferation induced by keratinocyte growth factor enhances in vivo retroviral-mediated gene transfer to mouse hepatocytes. J Clin Invest. 1996;98(12):2683–7.

    PubMed  CAS  Google Scholar 

  77. Patijn GA, Lieber A, Schowalter DB, Schwall R, Kay MA. Hepatocyte growth factor induces hepatocyte proliferation in vivo and allows for efficient retroviral-mediated gene transfer in mice. Hepatology. 1998;28(3):707–16.

    PubMed  CAS  Google Scholar 

  78. Hacein-Bey-Abina S, Garrigue A, Wang GP, et al. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest. 2008;118(9):3132–42.

    PubMed  CAS  Google Scholar 

  79. Howe SJ, Mansour MR, Schwarzwaelder K, et al. Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. J Clin Invest. 2008;118(9):3143–50.

    PubMed  CAS  Google Scholar 

  80. Wu X, Li Y, Crise B, Burgess SM. Transcription start regions in the human genome are favored targets for MLV integration. Science. 2003;300(5626):1749–51.

    PubMed  CAS  Google Scholar 

  81. Tittiger ME, Ma X, Xu L, Ponder KP. Neonatal intravenous injection of a gammaretroviral vector has a low incidence of tumor induction in mice. Hum Gene Ther. 2008;19(11)1317–23.

    Google Scholar 

  82. Naldini L, Blomer U, Gage FH, Trono D, Verma IM. Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector. Proc Natl Acad Sci U S A. 1996;93(21):11382–8.

    PubMed  CAS  Google Scholar 

  83. Kafri T, Blomer U, Peterson DA, Gage FH, Verma IM. Sustained expression of genes delivered directly into liver and muscle by lentiviral vectors. Nat Genet. 1997;17(3):314–7.

    PubMed  CAS  Google Scholar 

  84. Nguyen TH, Oberholzer J, Birraux J, Majno P, Morel P, Trono D. Highly efficient lentiviral vector-mediated transduction of nondividing, fully reimplantable primary hepatocytes. Mol Ther. 2002;6(2):199–209.

    PubMed  CAS  Google Scholar 

  85. Nguyen TH, Birraux J, Wildhaber B, et al. Ex vivo lentivirus transduction and immediate transplantation of uncultured hepatocytes for treating hyperbilirubinemic Gunn rat. Transplantation. 2006;82(6):794–803.

    PubMed  Google Scholar 

  86. Menzel O, Birraux J, Wildhaber BE, Jond C, Lasne F, Habre W, Trono D, Nguyen TH, Chardot C. Biosafety in ex vivo gene therapy and conditional ablation of lentivirally transduced hepatocytes in nonhuman primates. Mol Ther. 2009;17(10):1754–60. Epub 2009 Jun 30.

    Google Scholar 

  87. Brown BD, Cantore A, Annoni A, et al. A microRNA-regulated lentiviral vector mediates stable correction of hemophilia B mice. Blood. 2007;110(13):4144–52.

    PubMed  CAS  Google Scholar 

  88. Follenzi A, Battaglia M, Lombardo A, Annoni A, Roncarolo MG, Naldini L. Targeting lentiviral vector expression to hepatocytes limits transgene-specific immune response and establishes long-term expression of human antihemophilic factor IX in mice. Blood. 2004;103(10):3700–9.

    PubMed  CAS  Google Scholar 

  89. Park F, Ohashi K, Kay MA. Therapeutic levels of human factor VIII and IX using HIV-1-based lentiviral vectors in mouse liver. Blood. 2000;96(3):1173–6.

    PubMed  CAS  Google Scholar 

  90. High KA. Update on progress and hurdles in novel genetic therapies for hemophilia. Hematology Am Soc Hematol Educ Program. 2007:466–472.

    Google Scholar 

  91. Waddington SN, Nivsarkar MS, Mistry AR, et al. Permanent phenotypic correction of hemophilia B in immunocompetent mice by prenatal gene therapy. Blood. 2004;104(9):2714–21.

    PubMed  CAS  Google Scholar 

  92. Yoshimitsu M, Sato T, Tao K, et al. Bioluminescent imaging of a marking transgene and correction of Fabry mice by neonatal injection of recombinant lentiviral vectors. Proc Natl Acad Sci U S A. 2004;101(48):16909–14.

    PubMed  CAS  Google Scholar 

  93. van der Wegen P, Louwen R, Imam AM, et al. Successful treatment of UGT1A1 deficiency in a rat model of Crigler-Najjar disease by intravenous administration of a liver-specific lentiviral vector. Mol Ther. 2006;13(2):374–81.

    PubMed  Google Scholar 

  94. Di Domenico C, Di Napoli D, Gonzalez YRE, Lombardo A, Naldini L, Di Natale P. Limited transgene immune response and long-term expression of human alpha-L-iduronidase in young adult mice with mucopolysaccharidosis type I by liver-directed gene therapy. Hum Gene Ther. 2006;17(11):1112–21.

    PubMed  Google Scholar 

  95. Di Natale P, Di Domenico C, Gargiulo N, et al. Treatment of the mouse model of mucopolysaccharidosis type IIIB with lentiviral-NAGLU vector. Biochem J. 2005;388(Pt 2):639–46.

    PubMed  Google Scholar 

  96. Kobayashi H, Carbonaro D, Pepper K, et al. Neonatal gene therapy of MPS I mice by intravenous injection of a lentiviral vector. Mol Ther. 2005;11(5):776–89.

    PubMed  CAS  Google Scholar 

  97. McIntyre C, Derrick Roberts AL, Ranieri E, Clements PR, Byers S, Anson DS. Lentiviral-mediated gene therapy for murine mucopolysaccharidosis type IIIA. Mol Genet Metab. 2008;93(4):411–8.

    PubMed  CAS  Google Scholar 

  98. Di Domenico C, Villani GR, Di Napoli D, et al. Gene therapy for a mucopolysaccharidosis type I murine model with lentiviral-IDUA vector. Hum Gene Ther. 2005;16(1):81–90.

    PubMed  Google Scholar 

  99. Brown BD, Gentner B, Cantore A, et al. Endogenous microRNA can be broadly exploited to regulate transgene expression according to tissue, lineage and differentiation state. Nat Biotechnol. 2007;25(12):1457–67.

    PubMed  CAS  Google Scholar 

  100. VandenDriessche T, Thorrez L, Naldini L, et al. Lentiviral vectors containing the human immunodeficiency virus type-1 central polypurine tract can efficiently transduce nondividing hepatocytes and antigen-presenting cells in vivo. Blood. 2002;100(3):813–22.

    PubMed  CAS  Google Scholar 

  101. Brown BD, Sitia G, Annoni A, et al. In vivo administration of lentiviral vectors triggers a type I interferon response that restricts hepatocyte gene transfer and promotes vector clearance. Blood. 2007;109(7):2797–805.

    PubMed  CAS  Google Scholar 

  102. Selden C, Mellor N, Rees M, et al. Growth factors improve gene expression after lentiviral transduction in human adult and fetal hepatocytes. J Gene Med. 2007;9(2):67–76.

    PubMed  CAS  Google Scholar 

  103. Park F, Ohashi K, Chiu W, Naldini L, Kay MA. Efficient lentiviral transduction of liver requires cell cycling in vivo. Nat Genet. 2000;24(1):49–52.

    PubMed  CAS  Google Scholar 

  104. Park F, Kay MA. Modified HIV-1 based lentiviral vectors have an effect on viral transduction efficiency and gene expression in vitro and in vivo. Mol Ther. 2001;4(3):164–73.

    PubMed  CAS  Google Scholar 

  105. Follenzi A, Ailles LE, Bakovic S, Geuna M, Naldini L. Gene transfer by lentiviral vectors is limited by nuclear translocation and rescued by HIV-1 pol sequences. Nat Genet. 2000;25(2):217–22.

    PubMed  CAS  Google Scholar 

  106. Zennou V, Petit C, Guetard D, Nerhbass U, Montagnier L, Charneau P. HIV-1 genome nuclear import is mediated by a central DNA flap. Cell. 2000;101(2):173–85.

    PubMed  CAS  Google Scholar 

  107. Schroder AR, Shinn P, Chen H, Berry C, Ecker JR, Bushman F. HIV-1 integration in the human genome favors active genes and local hotspots. Cell. 2002;110(4):521–9.

    PubMed  CAS  Google Scholar 

  108. Montini E, Cesana D, Schmidt M, et al. The genotoxic potential of retroviral vectors is strongly modulated by vector design and integration site selection in a mouse model of HSC gene therapy. J Clin Invest. 2009;119(4):964–75.

    PubMed  CAS  Google Scholar 

  109. Montini E, Cesana D, Schmidt M, et al. Hematopoietic stem cell gene transfer in a tumor-prone mouse model uncovers low genotoxicity of lentiviral vector integration. Nat Biotechnol. 2006;24(6):687–96.

    PubMed  CAS  Google Scholar 

  110. Themis M, Waddington SN, Schmidt M, et al. Oncogenesis following delivery of a nonprimate lentiviral gene therapy vector to fetal and neonatal mice. Mol Ther. 2005;12(4):763–71.

    PubMed  CAS  Google Scholar 

  111. Yanez-Munoz RJ, Balaggan KS, MacNeil A, et al. Effective gene therapy with nonintegrating lentiviral vectors. Nat Med. 2006;12(3):348–53.

    PubMed  CAS  Google Scholar 

  112. Bayer M, Kantor B, Cockrell A, et al. A large U3 deletion causes increased in vivo expression from a nonintegrating lentiviral vector. Mol Ther. 2008;16(12):1968–76.

    PubMed  CAS  Google Scholar 

  113. Wolff JA, Malone RW, Williams P, et al. Direct gene transfer into mouse muscle in vivo. Science. 1990;247(4949 Pt 1):1465–8.

    PubMed  CAS  Google Scholar 

  114. Hickman MA, Malone RW, Lehmann-Bruinsma K, et al. Gene expression following direct injection of DNA into liver. Hum Gene Ther. 1994;5(12):1477–83.

    PubMed  CAS  Google Scholar 

  115. Kanemura H, Iimuro Y, Takeuchi M, et al. Hepatocyte growth factor gene transfer with naked plasmid DNA ameliorates dimethylnitrosamine-induced liver fibrosis in rats. Hepatol Res. 2008;38(9):930–9.

    PubMed  CAS  Google Scholar 

  116. Horiguchi K, Hirano T, Ueki T, Hirakawa K, Fujimoto J. Treating liver cirrhosis in dogs with hepatocyte growth factor gene therapy via the hepatic artery. J Hepatobiliary Pancreat Surg. 2009;16(2):171–7.

    PubMed  Google Scholar 

  117. Perales JC, Ferkol T, Beegen H, Ratnoff OD, Hanson RW. Gene transfer in vivo: sustained expression and regulation of genes introduced into the liver by receptor-targeted uptake. Proc Natl Acad Sci U S A. 1994;91(9):4086–90.

    PubMed  CAS  Google Scholar 

  118. Kren BT, Unger GM, Sjeklocha L, et al. Nanocapsule-delivered Sleeping Beauty mediates therapeutic Factor VIII expression in liver sinusoidal endothelial cells of hemophilia A mice. J Clin Invest. 2009;119(7):2086–99.

    PubMed  CAS  Google Scholar 

  119. Hashida M, Nishikawa M, Yamashita F, Takakura Y. Cell-specific delivery of genes with glycosylated carriers. Adv Drug Deliv Rev. 2001;52(3):187–96.

    PubMed  CAS  Google Scholar 

  120. Kim SI, Shin D, Lee H, Ahn BY, Yoon Y, Kim M. Targeted delivery of siRNA against hepatitis C virus by apolipoprotein A-I-bound cationic liposomes. J Hepatol. 2009;50(3):479–88.

    PubMed  CAS  Google Scholar 

  121. Yamada T, Iwasaki Y, Tada H, et al. Nanoparticles for the delivery of genes and drugs to human hepatocytes. Nat Biotechnol. 2003;21(8):885–90.

    PubMed  CAS  Google Scholar 

  122. Sato Y, Murase K, Kato J, et al. Resolution of liver cirrhosis using vitamin A-coupled liposomes to deliver siRNA against a collagen-specific chaperone. Nat Biotechnol. 2008;26(4):431–42.

    PubMed  CAS  Google Scholar 

  123. Wong SC, Wakefield D, Klein J, et al. Hepatocyte targeting of nucleic acid complexes and liposomes by a T7 phage p17 peptide. Mol Pharm. 2006;3(4):386–97.

    PubMed  CAS  Google Scholar 

  124. Rozema DB, Lewis DL, Wakefield DH, et al. Dynamic PolyConjugates for targeted in vivo delivery of siRNA to hepatocytes. Proc Natl Acad Sci U S A. 2007;104(32):12982–7.

    PubMed  CAS  Google Scholar 

  125. Kaneda Y, Iwai K, Uchida T. Increased expression of DNA cointroduced with nuclear protein in adult rat liver. Science. 1989;243(4889):375–8.

    PubMed  CAS  Google Scholar 

  126. Liu L, Zern MA, Lizarzaburu ME, Nantz MH, Wu J. Poly(cationic lipid)-mediated in vivo gene delivery to mouse liver. Gene Ther. 2003;10(2):180–7.

    PubMed  CAS  Google Scholar 

  127. Li S, Huang L. In vivo gene transfer via intravenous administration of cationic lipid-protamine-DNA (LPD) complexes. Gene Ther. 1997;4(9):891–900.

    PubMed  CAS  Google Scholar 

  128. Soutschek J, Akinc A, Bramlage B, et al. (2004) Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs. Nature. 2004;432(7014):173–8.

    PubMed  CAS  Google Scholar 

  129. Zimmermann TS, Lee AC, Akinc A, et al. RNAi-mediated gene silencing in non-human primates. Nature. 2006;441(7089):111–4.

    PubMed  CAS  Google Scholar 

  130. Judge AD, Bola G, Lee AC, MacLachlan I. Design of noninflammatory synthetic siRNA mediating potent gene silencing in vivo. Mol Ther. 2006;13(3):494–505.

    PubMed  CAS  Google Scholar 

  131. Krutzfeldt J, Rajewsky N, Braich R, et al. Silencing of microRNAs in vivo with ‘antagomirs’. Nature. 2005;438(7068):685–9.

    PubMed  Google Scholar 

  132. Jopling CL, Yi M, Lancaster AM, Lemon SM, Sarnow P. Modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA. Science. 2005;309(5740):1577–81.

    PubMed  CAS  Google Scholar 

  133. Okuyama T, Huber RM, Bowling W, et al. Liver-directed gene therapy: a retroviral vector with a complete LTR and the ApoE enhancer-alpha 1-antitrypsin promoter dramatically increases expression of human alpha 1-antitrypsin in vivo. Hum Gene Ther. 1996;7(5):637–45.

    PubMed  CAS  Google Scholar 

  134. Miao CH, Ohashi K, Patijn GA, et al. Inclusion of the hepatic locus control region, an intron, and untranslated region increases and stabilizes hepatic factor IX gene expression in vivo but not in vitro. Mol Ther. 2000;1(6):522–32.

    PubMed  CAS  Google Scholar 

  135. Jacobs F, Snoeys J, Feng Y, et al. Direct comparison of hepatocyte-specific expression cassettes following adenoviral and nonviral hydrodynamic gene transfer. Gene Ther. 2008;15(8):594–603.

    PubMed  CAS  Google Scholar 

  136. Kramer MG, Barajas M, Razquin N, et al. In vitro and in vivo comparative study of chimeric liver-specific promoters. Mol Ther. 2003;7(3):375–85.

    PubMed  CAS  Google Scholar 

  137. Riu E, Chen ZY, Xu H, He CY, Kay MA. Histone modifications are associated with the persistence or silencing of vector-mediated transgene expression in vivo. Mol Ther. 2007;15(7):1348–55.

    PubMed  CAS  Google Scholar 

  138. Chen ZY, Yant SR, He CY, Meuse L, Shen S, Kay MA. Linear DNAs concatemerize in vivo and result in sustained transgene expression in mouse liver. Mol Ther. 2001;3(3):403–10.

    PubMed  CAS  Google Scholar 

  139. Li S, Wu SP, Whitmore M, et al. Effect of immune response on gene transfer to the lung via systemic administration of cationic lipidic vectors. Am J Physiol. 1999;276(5 Pt 1):L796–804.

    PubMed  CAS  Google Scholar 

  140. Tan Y, Li S, Pitt BR, Huang L. The inhibitory role of CpG immunostimulatory motifs in cationic lipid vector-mediated transgene expression in vivo. Hum Gene Ther. 1999;10(13):2153–61.

    PubMed  CAS  Google Scholar 

  141. Tousignant JD, Gates AL, Ingram LA, et al. Comprehensive analysis of the acute toxicities induced by systemic administration of cationic lipid:plasmid DNA complexes in mice. Hum Gene Ther. 2000;11(18):2493–513.

    PubMed  CAS  Google Scholar 

  142. Zhao H, Hemmi H, Akira S, Cheng SH, Scheule RK, Yew NS. Contribution of Toll-like receptor 9 signaling to the acute inflammatory response to nonviral vectors. Mol Ther. 2004;9(2):241–8.

    PubMed  CAS  Google Scholar 

  143. Yew NS, Zhao H, Przybylska M, et al. CpG-depleted plasmid DNA vectors with enhanced safety and long-term gene expression in vivo. Mol Ther. 2002;5(6):731–8.

    PubMed  CAS  Google Scholar 

  144. Miao CH, Ye X, Thompson AR. High-level factor VIII gene expression in vivo achieved by nonviral liver-specific gene therapy vectors. Hum Gene Ther. 2003;14(14):1297–305.

    PubMed  CAS  Google Scholar 

  145. Wooddell CI, Reppen T, Wolff JA, Herweijer H. Sustained liver-specific transgene expression from the albumin promoter in mice following hydrodynamic plasmid DNA delivery. J Gene Med. 2008;10(5):551–63.

    PubMed  CAS  Google Scholar 

  146. Hodges BL, Scheule RK. Hydrodynamic delivery of DNA. Expert Opin Biol Ther. 2003;3(6):911–8.

    PubMed  CAS  Google Scholar 

  147. Brunetti-Pierri N, Palmer DJ, Mane V, Finegold M, Beaudet AL, Ng P. Increased hepatic transduction with reduced systemic dissemination and proinflammatory cytokines following hydrodynamic injection of helper-dependent adenoviral vectors. Mol Ther. 2005;12(1):99–106.

    PubMed  CAS  Google Scholar 

  148. Yant SR, Meuse L, Chiu W, Ivics Z, Izsvak Z, Kay MA. Somatic integration and long-term transgene expression in normal and haemophilic mice using a DNA transposon system. Nat Genet. 2000;25(1):35–41.

    PubMed  CAS  Google Scholar 

  149. Miao CH. A novel gene expression system: non-viral gene transfer for hemophilia as model systems. Adv Genet. 2005;54:143–77.

    PubMed  CAS  Google Scholar 

  150. Ohlfest JR, Frandsen JL, Fritz S, et al. Phenotypic correction and long-term expression of factor VIII in hemophilic mice by immunotolerization and nonviral gene transfer using the Sleeping Beauty transposon system. Blood. 2005;105(7):2691–8.

    PubMed  CAS  Google Scholar 

  151. Olivares EC, Hollis RP, Chalberg TW, Meuse L, Kay MA, Calos MP. Site-specific genomic integration produces therapeutic Factor IX levels in mice. Nat Biotechnol. 2002;20(11):1124–8.

    PubMed  CAS  Google Scholar 

  152. Montini E, Held PK, Noll M, et al. In vivo correction of murine tyrosinemia type I by DNA-mediated transposition. Mol Ther. 2002;6(6):759–69.

    PubMed  CAS  Google Scholar 

  153. Aronovich EL, Bell JB, Belur LR, et al. Prolonged expression of a lysosomal enzyme in mouse liver after Sleeping Beauty transposon-mediated gene delivery: implications for non-viral gene therapy of mucopolysaccharidoses. J Gene Med. 2007;9(5):403–15.

    PubMed  CAS  Google Scholar 

  154. Held PK, Olivares EC, Aguilar CP, Finegold M, Calos MP, Grompe M. In vivo correction of murine hereditary tyrosinemia type I by phiC31 integrase-mediated gene delivery. Mol Ther. 2005;11(3):399–408.

    PubMed  CAS  Google Scholar 

  155. McCaffrey AP, Nakai H, Pandey K, et al. Inhibition of hepatitis B virus in mice by RNA interference. Nat Biotechnol. 2003;21(6):639–44.

    PubMed  CAS  Google Scholar 

  156. Zender L, Hutker S, Liedtke C, et al. Caspase 8 small interfering RNA prevents acute liver failure in mice. Proc Natl Acad Sci U S A. 2003;100(13):7797–802.

    PubMed  CAS  Google Scholar 

  157. He CX, Shi D, Wu WJ, et al. Insulin expression in livers of diabetic mice mediated by hydrodynamics-based administration. World J Gastroenterol. 2004;10(4):567–72.

    PubMed  CAS  Google Scholar 

  158. Yazawa H, Murakami T, Li HM, et al. Hydrodynamics-based gene delivery of naked DNA encoding fetal liver kinase-1 gene effectively suppresses the growth of pre-existing tumors. Cancer Gene Ther. 2006;13(11):993–1001.

    PubMed  CAS  Google Scholar 

  159. Chen HW, Lee YP, Chung YF, et al. Inducing long-term survival with lasting anti-tumor immunity in treating B cell lymphoma by a combined dendritic cell-based and hydrodynamic plasmid-encoding IL-12 gene therapy. Int Immunol. 2003;15(3):427–35.

    PubMed  CAS  Google Scholar 

  160. Jiang J, Yamato E, Miyazaki J. Intravenous delivery of naked plasmid DNA for in vivo cytokine expression. Biochem Biophys Res Commun. 2001;289(5):1088–92.

    PubMed  CAS  Google Scholar 

  161. He Y, Pimenov AA, Nayak JV, Plowey J, Falo Jr LD, Huang L. Intravenous injection of naked DNA encoding secreted flt3 ligand dramatically increases the number of dendritic cells and natural killer cells in vivo. Hum Gene Ther. 2000;11(4):547–54.

    PubMed  CAS  Google Scholar 

  162. Ortaldo JR, Winkler-Pickett RT, Bere Jr EW, Watanabe M, Murphy WJ, Wiltrout RH. In vivo hydrodynamic delivery of cDNA encoding IL-2: rapid, sustained redistribution, activation of mouse NK cells, and therapeutic potential in the absence of NKT cells. J Immunol. 2005;175(2):693–9.

    PubMed  CAS  Google Scholar 

  163. Yang PL, Althage A, Chung J, Chisari FV. Hydrodynamic injection of viral DNA: a mouse model of acute hepatitis B virus infection. Proc Natl Acad Sci U S A. 2002;99(21):13825–30.

    PubMed  CAS  Google Scholar 

  164. Suda T, Suda K, Liu D. Computer-assisted hydrodynamic gene delivery. Mol Ther. 2008;16(6):1098–104.

    PubMed  CAS  Google Scholar 

  165. Kamimura K, Suda T, Xu W, Zhang G, Liu D. Image-guided, lobe-specific hydrodynamic gene delivery to swine liver. Mol Ther. 2009;17(3):491–9.

    PubMed  CAS  Google Scholar 

  166. Zhang G, Gao X, Song YK, et al. Hydroporation as the mechanism of hydrodynamic delivery. Gene Ther. 2004;11(8):675–82.

    PubMed  CAS  Google Scholar 

  167. Kobayashi N, Nishikawa M, Hirata K, Takakura Y. Hydrodynamics-based procedure involves transient hyperpermeability in the hepatic cellular membrane: implication of a nonspecific process in efficient intracellular gene delivery. J Gene Med. 2004;6(5):584–92.

    PubMed  CAS  Google Scholar 

  168. Crespo A, Peydro A, Dasi F, et al. Hydrodynamic liver gene transfer mechanism involves transient sinusoidal blood stasis and massive hepatocyte endocytic vesicles. Gene Ther. 2005;12(11):927–35.

    PubMed  CAS  Google Scholar 

  169. Budker V, Budker T, Zhang G, Subbotin V, Loomis A, Wolff JA. Hypothesis: naked plasmid DNA is taken up by cells in vivo by a receptor-mediated process. J Gene Med. 2000;2(2):76–88.

    PubMed  CAS  Google Scholar 

  170. Herweijer H, Wolff JA. Gene therapy progress and prospects: hydrodynamic gene delivery. Gene Ther. 2007;14(2):99–107.

    PubMed  CAS  Google Scholar 

  171. Suda T, Liu D. Hydrodynamic gene delivery: its principles and applications. Mol Ther. 2007;15(12):2063–9.

    PubMed  CAS  Google Scholar 

  172. Ivics Z, Hackett PB, Plasterk RH, Izsvak Z. Molecular reconstruction of Sleeping Beauty, a Tc1-like transposon from fish, and its transposition in human cells. Cell. 1997;91(4):501–10.

    PubMed  CAS  Google Scholar 

  173. Thyagarajan B, Olivares EC, Hollis RP, Ginsburg DS, Calos MP. Site-specific genomic integration in mammalian cells mediated by phage phiC31 integrase. Mol Cell Biol. 2001;21(12):3926–34.

    PubMed  CAS  Google Scholar 

  174. Huang X, Wilber AC, Bao L, et al. Stable gene transfer and expression in human primary T cells by the Sleeping Beauty transposon system. Blood. 2006;107(2):483–91.

    PubMed  CAS  Google Scholar 

  175. Ehrhardt A, Xu H, Huang Z, Engler JA, Kay MA. A direct comparison of two nonviral gene therapy vectors for somatic integration: in vivo evaluation of the bacteriophage integrase phiC31 and the Sleeping Beauty transposase. Mol Ther. 2005;11(5):695–706.

    PubMed  CAS  Google Scholar 

  176. Aronovich EL, Bell JB, Khan SA, et al. Systemic correction of storage disease in MPS I NOD/SCID mice using the sleeping beauty transposon system. Mol Ther. 2009;17(7):1136–44.

    PubMed  CAS  Google Scholar 

  177. Yant SR, Ehrhardt A, Mikkelsen JG, Meuse L, Pham T, Kay MA. Transposition from a gutless adeno-transposon vector stabilizes transgene expression in vivo. Nat Biotechnol. 2002;20(10):999–1005.

    PubMed  CAS  Google Scholar 

  178. Ehrhardt A, Yant SR, Giering JC, Xu H, Engler JA, Kay MA. Somatic integration from an adenoviral hybrid vector into a hot spot in mouse liver results in persistent transgene expression levels in vivo. Mol Ther. 2007;15(1):146–56.

    PubMed  CAS  Google Scholar 

  179. Vink CA, Gaspar HB, Gabriel R, et al. Sleeping beauty transposition from nonintegrating lentivirus. Mol Ther. 2009;17(7):1197–204.

    PubMed  CAS  Google Scholar 

  180. Staunstrup NH, Moldt B, Mates L, et al. Hybrid lentivirus-transposon vectors with a random integration profile in human cells. Mol Ther. 2009;17(7):1205–14.

    PubMed  CAS  Google Scholar 

  181. Yant SR, Wu X, Huang Y, Garrison B, Burgess SM, Kay MA. High-resolution genome-wide mapping of transposon integration in mammals. Mol Cell Biol. 2005;25(6):2085–94.

    PubMed  CAS  Google Scholar 

  182. Mates L, Chuah MK, Belay E, et al. Molecular evolution of a novel hyperactive Sleeping Beauty transposase enables robust stable gene transfer in vertebrates. Nat Genet. 2009;41(6):753–61.

    PubMed  CAS  Google Scholar 

  183. Chalberg TW, Portlock JL, Olivares EC, et al. Integration specificity of phage phiC31 integrase in the human genome. J Mol Biol. 2006;357(1):28–48.

    PubMed  CAS  Google Scholar 

  184. Ehrhardt A, Engler JA, Xu H, Cherry AM, Kay MA. Molecular analysis of chromosomal rearrangements in mammalian cells after phiC31-mediated integration. Hum Gene Ther. 2006;17(11):1077–94.

    PubMed  CAS  Google Scholar 

  185. Liu J, Jeppesen I, Nielsen K, Jensen TG. Phi c31 integrase induces chromosomal aberrations in primary human fibroblasts. Gene Ther. 2006;13(15):1188–90.

    PubMed  CAS  Google Scholar 

  186. Keravala A, Lee S, Thyagarajan B, et al. Mutational derivatives of PhiC31 integrase with increased efficiency and specificity. Mol Ther. 2009;17(1):112–20.

    PubMed  CAS  Google Scholar 

  187. Carroll D. Progress and prospects: zinc-finger nucleases as gene therapy agents. Gene Ther. 2008;15(22):1463–8.

    PubMed  CAS  Google Scholar 

  188. Porteus MH. Mammalian gene targeting with designed zinc finger nucleases. Mol Ther. 2006;13(2):438–46.

    PubMed  CAS  Google Scholar 

  189. Russell DW, Hirata RK. Human gene targeting by viral vectors. Nat Genet. 1998;18(4):325–30.

    PubMed  CAS  Google Scholar 

  190. Chamberlain JR, Schwarze U, Wang PR, et al. Gene targeting in stem cells from individuals with osteogenesis imperfecta. Science. 2004;303(5661):1198–201.

    PubMed  CAS  Google Scholar 

  191. Hockemeyer D, Soldner F, Beard C, et al. Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases. Nat Biotechnol. 2009;27(9):851–7.

    PubMed  CAS  Google Scholar 

  192. Miller DG, Wang PR, Petek LM, Hirata RK, Sands MS, Russell DW. Gene targeting in vivo by adeno-associated virus vectors. Nat Biotechnol. 2006;24(8):1022–6.

    PubMed  CAS  Google Scholar 

  193. de Semir D, Aran JM. Targeted gene repair: the ups and downs of a promising gene therapy approach. Curr Gene Ther. 2006;6(4):481–504.

    PubMed  Google Scholar 

  194. Kren BT, Bandyopadhyay P, Steer CJ. In vivo site-directed mutagenesis of the factor IX gene by chimeric RNA/DNA oligonucleotides. Nat Med. 1998;4(3):285–90.

    PubMed  CAS  Google Scholar 

  195. Kren BT, Parashar B, Bandyopadhyay P, Chowdhury NR, Chowdhury JR, Steer CJ. Correction of the UDP-glucuronosyltransferase gene defect in the gunn rat model of crigler-najjar syndrome type I with a chimeric oligonucleotide. Proc Natl Acad Sci U S A. 1999;96(18):10349–54.

    PubMed  CAS  Google Scholar 

  196. Taubes G. Gene therapy. The strange case of chimeraplasty. Science. 2002;298(5601):2116–20.

    PubMed  CAS  Google Scholar 

  197. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998;391(6669):806–11.

    PubMed  CAS  Google Scholar 

  198. Kim DH, Rossi JJ. Strategies for silencing human disease using RNA interference. Nat Rev Genet. 2007;8(3):173–84.

    PubMed  CAS  Google Scholar 

  199. Castanotto D, Rossi JJ. The promises and pitfalls of RNA-interference-based therapeutics. Nature. 2009;457(7228):426–33.

    PubMed  CAS  Google Scholar 

  200. McCaffrey AP, Meuse L, Pham TT, Conklin DS, Hannon GJ, Kay MA. RNA interference in adult mice. Nature. 2002;418(6893):38–9.

    PubMed  CAS  Google Scholar 

  201. Giering JC, Grimm D, Storm TA, Kay MA. Expression of shRNA from a tissue-specific pol II promoter is an effective and safe RNAi therapeutic. Mol Ther. 2008;16(9):1630–6.

    PubMed  CAS  Google Scholar 

  202. Doege H, Grimm D, Falcon A, et al. Silencing of hepatic fatty acid transporter protein 5 in vivo reverses diet-induced non-alcoholic fatty liver disease and improves hyperglycemia. J Biol Chem. 2008;283(32):22186–92.

    PubMed  CAS  Google Scholar 

  203. Song E, Lee SK, Wang J, et al. RNA interference targeting Fas protects mice from fulminant hepatitis. Nat Med. 2003;9(3):347–51.

    PubMed  CAS  Google Scholar 

  204. Judge AD, Sood V, Shaw JR, Fang D, McClintock K, MacLachlan I. Sequence-dependent stimulation of the mammalian innate immune response by synthetic siRNA. Nat Biotechnol. 2005;23(4):457–62.

    PubMed  CAS  Google Scholar 

  205. Hornung V, Guenthner-Biller M, Bourquin C, et al. Sequence-specific potent induction of IFN-alpha by short interfering RNA in plasmacytoid dendritic cells through TLR7. Nat Med. 2005;11(3):263–70.

    PubMed  CAS  Google Scholar 

  206. Jackson AL, Burchard J, Schelter J, et al. Widespread siRNA “off-target” transcript silencing mediated by seed region sequence complementarity. RNA. 2006;12(7):1179–87.

    PubMed  CAS  Google Scholar 

  207. Burchard J, Jackson AL, Malkov V, et al. MicroRNA-like off-target transcript regulation by siRNAs is species specific. RNA. 2009;15(2):308–15.

    PubMed  CAS  Google Scholar 

  208. Judge A, MacLachlan I. Overcoming the innate immune response to small interfering RNA. Hum Gene Ther. 2008;19(2):111–24.

    PubMed  CAS  Google Scholar 

  209. Sun X, Rogoff HA, Li CJ. Asymmetric RNA duplexes mediate RNA interference in mammalian cells. Nat Biotechnol. 2008;26(12):1379–82.

    PubMed  CAS  Google Scholar 

  210. McBride JL, Boudreau RL, Harper SQ, et al. Artificial miRNAs mitigate shRNA-mediated toxicity in the brain: implications for the therapeutic development of RNAi. Proc Natl Acad Sci U S A. 2008;105(15):5868–73.

    PubMed  CAS  Google Scholar 

  211. Ely A, Naidoo T, Mufamadi S, Crowther C, Arbuthnot P. Expressed anti-HBV primary microRNA shuttles inhibit viral replication efficiently in vitro and in vivo. Mol Ther. 2008;16(6):1105–12.

    PubMed  CAS  Google Scholar 

  212. Herzog RW. Immune responses to AAV capsid: are mice not humans after all? Mol Ther. 2007;15(4):649–50.

    PubMed  CAS  Google Scholar 

  213. Lu H, Chen L, Wang J, et al. Complete correction of hemophilia A with adeno-associated viral vectors containing a full-size expression cassette. Hum Gene Ther. 2008;19(6):648–54.

    PubMed  CAS  Google Scholar 

  214. Ye X, Loeb KR, Stafford DW, Thompson AR, Miao CH. Complete and sustained phenotypic correction of hemophilia B in mice following hepatic gene transfer of a high-expressing human factor IX plasmid. J Thromb Haemost. 2003;1(1):103–11.

    PubMed  CAS  Google Scholar 

  215. Sands MS, Davidson BL. Gene therapy for lysosomal storage diseases. Mol Ther. 2006;13(5):839–49.

    PubMed  CAS  Google Scholar 

  216. Liu Y, Xu L, Hennig AK, et al. Liver-directed neonatal gene therapy prevents cardiac, bone, ear, and eye disease in mucopolysaccharidosis I mice. Mol Ther. 2005;11(1):35–47.

    PubMed  CAS  Google Scholar 

  217. Xu L, Mango RL, Sands MS, Haskins ME, Ellinwood NM, Ponder KP. Evaluation of pathological manifestations of disease in mucopolysaccharidosis VII mice after neonatal hepatic gene therapy. Mol Ther. 2002;6(6):745–58.

    PubMed  CAS  Google Scholar 

  218. Nguyen TH, Bellodi-Privato M, Aubert D, et al. Therapeutic lentivirus-mediated neonatal in vivo gene therapy in hyperbilirubinemic Gunn rats. Mol Ther. 2005;12(5):852–9.

    PubMed  CAS  Google Scholar 

  219. Daly TM, Vogler C, Levy B, Haskins ME, Sands MS. Neonatal gene transfer leads to widespread correction of pathology in a murine model of lysosomal storage disease. Proc Natl Acad Sci U S A. 1999;96(5):2296–300.

    PubMed  CAS  Google Scholar 

  220. Ding Z, Georgiev P, Thony B. Administration-route and gender-independent long-term therapeutic correction of phenylketonuria (PKU) in a mouse model by recombinant adeno-associated virus 8 pseudotyped vector-mediated gene transfer. Gene Ther. 2006;13(7):587–93.

    PubMed  CAS  Google Scholar 

  221. Bataller R, Brenner DA. Liver fibrosis. J Clin Invest. 2005;115(2):209–18.

    PubMed  CAS  Google Scholar 

  222. Iredale J. Defining therapeutic targets for liver fibrosis: exploiting the biology of inflammation and repair. Pharmacol Res. 2008;58(2):129–36.

    PubMed  CAS  Google Scholar 

  223. Friedman SL, Roll FJ, Boyles J, Bissell DM. Hepatic lipocytes: the principal collagen-producing cells of normal rat liver. Proc Natl Acad Sci U S A. 1985;82(24):8681–5.

    PubMed  CAS  Google Scholar 

  224. Forbes SJ, Russo FP, Rey V, et al. A significant proportion of myofibroblasts are of bone marrow origin in human liver fibrosis. Gastroenterology. 2004;126(4):955–63.

    PubMed  Google Scholar 

  225. Siller-Lopez F, Sandoval A, Salgado S, et al. Treatment with human metalloproteinase-8 gene delivery ameliorates experimental rat liver cirrhosis. Gastroenterology. 2004;126(4):1122–33. discussion 1949.

    PubMed  CAS  Google Scholar 

  226. Iimuro Y, Brenner DA. Matrix metalloproteinase gene delivery for liver fibrosis. Pharm Res. 2008;25(2):249–58.

    PubMed  CAS  Google Scholar 

  227. Iimuro Y, Nishio T, Morimoto T, et al. Delivery of matrix metalloproteinase-1 attenuates established liver fibrosis in the rat. Gastroenterology. 2003;124(2):445–58.

    PubMed  CAS  Google Scholar 

  228. Garcia-Banuelos J, Siller-Lopez F, Miranda A, Aguilar LK, Aguilar-Cordova E, Armendariz-Borunda J. Cirrhotic rat livers with extensive fibrosis can be safely transduced with clinical-grade adenoviral vectors. Evidence of cirrhosis reversion. Gene Ther. 2002;9(2):127–34.

    PubMed  CAS  Google Scholar 

  229. Bueno M, Salgado S, Beas-Zarate C, Armendariz-Borunda J. Urokinase-type plasminogen activator gene therapy in liver cirrhosis is mediated by collagens gene expression down-regulation and up-regulation of MMPs, HGF and VEGF. J Gene Med. 2006;8(11):1291–9.

    PubMed  CAS  Google Scholar 

  230. Roderfeld M, Weiskirchen R, Wagner S, et al. Inhibition of hepatic fibrogenesis by matrix metalloproteinase-9 mutants in mice. FASEB J. 2006;20(3):444–54.

    PubMed  CAS  Google Scholar 

  231. Jiang W, Wang JY, Yang CQ, Liu WB, Wang YQ, He BM. Effects of a plasmid expressing antisense tissue inhibitor of metalloproteinase-1 on liver fibrosis in rats. Chin Med J (Engl). 2005;118(3):192–7.

    CAS  Google Scholar 

  232. Hu YB, Li DG, Lu HM. Modified synthetic siRNA targeting tissue inhibitor of metalloproteinase-2 inhibits hepatic fibrogenesis in rats. J Gene Med. 2007;9(3):217–29.

    PubMed  CAS  Google Scholar 

  233. Qi Z, Atsuchi N, Ooshima A, Takeshita A, Ueno H. Blockade of type beta transforming growth factor signaling prevents liver fibrosis and dysfunction in the rat. Proc Natl Acad Sci U S A. 1999;96(5):2345–9.

    PubMed  CAS  Google Scholar 

  234. Arias M, Sauer-Lehnen S, Treptau J, et al. Adenoviral expression of a transforming growth factor-beta1 antisense mRNA is effective in preventing liver fibrosis in bile-duct ligated rats. BMC Gastroenterol. 2003;3:29.

    PubMed  Google Scholar 

  235. Kinoshita K, Iimuro Y, Otogawa K, et al. Adenovirus-mediated expression of BMP-7 suppresses the development of liver fibrosis in rats. Gut. 2007;56(5):706–14.

    PubMed  CAS  Google Scholar 

  236. George J, Tsutsumi M. siRNA-mediated knockdown of connective tissue growth factor prevents N-nitrosodimethylamine-induced hepatic fibrosis in rats. Gene Ther. 2007;14(10):790–803.

    PubMed  CAS  Google Scholar 

  237. Chen SW, Zhang XR, Wang CZ, Chen WZ, Xie WF, Chen YX. RNA interference targeting the platelet-derived growth factor receptor beta subunit ameliorates experimental hepatic fibrosis in rats. Liver Int. 2008;28(10):1446–57.

    PubMed  CAS  Google Scholar 

  238. Xia JL, Dai C, Michalopoulos GK, Liu Y. Hepatocyte growth factor attenuates liver fibrosis induced by bile duct ligation. Am J Pathol. 2006;168(5):1500–12.

    PubMed  CAS  Google Scholar 

  239. Ozawa S, Uchiyama K, Nakamori M, et al. Combination gene therapy of HGF and truncated type II TGF-beta receptor for rat liver cirrhosis after partial hepatectomy. Surgery. 2006;139(4):563–73.

    PubMed  Google Scholar 

  240. Ke B, Lipshutz GS, Kupiec-Weglinski JW. Gene therapy in liver ischemia and reperfusion injury. Curr Pharm Des. 2006;12(23):2969–75.

    PubMed  CAS  Google Scholar 

  241. Ritter T, Kupiec-Weglinski JW. Gene therapy for the prevention of ischemia/reperfusion injury in organ transplantation. Curr Gene Ther. 2005;5(1):101–9.

    PubMed  CAS  Google Scholar 

  242. Lehmann TG, Wheeler MD, Schoonhoven R, Bunzendahl H, Samulski RJ, Thurman RG. Delivery of Cu/Zn-superoxide dismutase genes with a viral vector minimizes liver injury and improves survival after liver transplantation in the rat. Transplantation. 2000;69(6):1051–7.

    PubMed  CAS  Google Scholar 

  243. Wheeler MD, Katuna M, Smutney OM, et al. Comparison of the effect of adenoviral delivery of three superoxide dismutase genes against hepatic ischemia-reperfusion injury. Hum Gene Ther. 2001;12(18):2167–77.

    PubMed  CAS  Google Scholar 

  244. He SQ, Zhang YH, Venugopal SK, et al. Delivery of antioxidative enzyme genes protects against ischemia/reperfusion-induced liver injury in mice. Liver Transpl. 2006;12(12):1869–79.

    PubMed  Google Scholar 

  245. Coito AJ, Buelow R, Shen XD, et al. Heme oxygenase-1 gene transfer inhibits inducible nitric oxide synthase expression and protects genetically fat Zucker rat livers from ischemia-reperfusion injury. Transplantation. 2002;74(1):96–102.

    PubMed  CAS  Google Scholar 

  246. Ke B, Buelow R, Shen XD, et al. Heme oxygenase 1 gene transfer prevents CD95/Fas ligand-mediated apoptosis and improves liver allograft survival via carbon monoxide signaling pathway. Hum Gene Ther. 2002;13(10):1189–99.

    PubMed  CAS  Google Scholar 

  247. Berberat PO, Katori M, Kaczmarek E, et al. Heavy chain ferritin acts as an antiapoptotic gene that protects livers from ischemia reperfusion injury. FASEB J. 2003;17(12):1724–6.

    PubMed  CAS  Google Scholar 

  248. Bilbao G, Contreras JL, Eckhoff DE, et al. Reduction of ischemia-reperfusion injury of the liver by in vivo adenovirus-mediated gene transfer of the antiapoptotic Bcl-2 gene. Ann Surg. 1999;230(2):185–93.

    PubMed  CAS  Google Scholar 

  249. Harada H, Wakabayashi G, Takayanagi A, et al. Transfer of the interleukin-1 receptor antagonist gene into rat liver abrogates hepatic ischemia-reperfusion injury. Transplantation. 2002;74(10):1434–41.

    PubMed  CAS  Google Scholar 

  250. Ke B, Shen XD, Gao F, et al. Gene therapy for liver transplantation using adenoviral vectors: CD40-CD154 blockade by gene transfer of CD40Ig protects rat livers from cold ischemia and reperfusion injury. Mol Ther. 2004;9(1):38–45.

    PubMed  CAS  Google Scholar 

  251. Kolodka TM, Finegold M, Moss L, Woo SL. Gene therapy for diabetes mellitus in rats by hepatic expression of insulin. Proc Natl Acad Sci U S A. 1995;92(8):3293–7.

    PubMed  CAS  Google Scholar 

  252. Nett PC, Sollinger HW, Alam T. Hepatic insulin gene therapy in insulin-dependent diabetes mellitus. Am J Transplant. 2003;3(10):1197–203.

    PubMed  CAS  Google Scholar 

  253. Thule PM, Liu JM. Regulated hepatic insulin gene therapy of STZ-diabetic rats. Gene Ther. 2000;7(20):1744–52.

    PubMed  CAS  Google Scholar 

  254. Hay CW, Docherty K. Enhanced expression of a furin-cleavable proinsulin. J Mol Endocrinol. 2003;31(3):597–607.

    PubMed  CAS  Google Scholar 

  255. Thule PM, Liu J, Phillips LS. Glucose regulated production of human insulin in rat hepatocytes. Gene Ther. 2000;7(3):205–14.

    PubMed  CAS  Google Scholar 

  256. Chen R, Meseck M, McEvoy RC, Woo SL. Glucose-stimulated and self-limiting insulin production by glucose 6-phosphatase promoter driven insulin expression in hepatoma cells. Gene Ther. 2000;7(21):1802–9.

    PubMed  CAS  Google Scholar 

  257. Burkhardt BR, Parker MJ, Zhang YC, Song S, Wasserfall CH, Atkinson MA. Glucose transporter-2 (GLUT2) promoter mediated transgenic insulin production reduces hyperglycemia in diabetic mice. FEBS Lett. 2005;579(25):5759–64.

    PubMed  CAS  Google Scholar 

  258. Burkhardt BR, Loiler SA, Anderson JA, et al. Glucose-responsive expression of the human insulin promoter in HepG2 human hepatoma cells. Ann NY Acad Sci. 2003;1005:237–41.

    PubMed  CAS  Google Scholar 

  259. Olson DE, Paveglio SA, Huey PU, Porter MH, Thule PM. Glucose-responsive hepatic insulin gene therapy of spontaneously diabetic BB/Wor rats. Hum Gene Ther. 2003;14(15):1401–13.

    PubMed  CAS  Google Scholar 

  260. Chen R, Meseck ML, Woo SL. Auto-regulated hepatic insulin gene expression in type 1 diabetic rats. Mol Ther. 2001;3(4):584–90.

    PubMed  CAS  Google Scholar 

  261. Auricchio A, Gao GP, Yu QC, et al. Constitutive and regulated expression of processed insulin following in vivo hepatic gene transfer. Gene Ther. 2002;9(14):963–71.

    PubMed  CAS  Google Scholar 

  262. Kojima H, Fujimiya M, Matsumura K, et al. NeuroD-betacellulin gene therapy induces islet neogenesis in the liver and reverses diabetes in mice. Nat Med. 2003;9(5):596–603.

    PubMed  CAS  Google Scholar 

  263. Yechoor V, Liu V, Espiritu C, et al. Neurogenin3 is sufficient for transdetermination of hepatic progenitor cells into neo-islets in vivo but not transdifferentiation of hepatocytes. Dev Cell. 2009;16(3):358–73.

    PubMed  CAS  Google Scholar 

  264. Song YD, Lee EJ, Yashar P, Pfaff LE, Kim SY, Jameson JL. Islet cell differentiation in liver by combinatorial expression of transcription factors neurogenin-3, BETA2, and RIPE3b1. Biochem Biophys Res Commun. 2007;354(2):334–9.

    PubMed  CAS  Google Scholar 

  265. Li H, Li X, Lam KS, Tam S, Xiao W, Xu R. Adeno-associated virus-mediated pancreatic and duodenal homeobox gene-1 expression enhanced differentiation of hepatic oval stem cells to insulin-producing cells in diabetic rats. J Biomed Sci. 2008;15(4):487–97.

    PubMed  Google Scholar 

  266. Wang AY, Ehrhardt A, Xu H, Kay MA. Adenovirus transduction is required for the correction of diabetes using Pdx-1 or Neurogenin-3 in the liver. Mol Ther. 2007;15(2):255–63.

    PubMed  CAS  Google Scholar 

  267. Arbuthnot P, Longshaw V, Naidoo T, Weinberg MS. Opportunities for treating chronic hepatitis B and C virus infection using RNA interference. J Viral Hepat. 2007;14(7):447–59.

    PubMed  CAS  Google Scholar 

  268. Uprichard SL, Boyd B, Althage A, Chisari FV. Clearance of hepatitis B virus from the liver of transgenic mice by short hairpin RNAs. Proc Natl Acad Sci U S A. 2005;102(3):773–8.

    PubMed  CAS  Google Scholar 

  269. Crowther C, Ely A, Hornby J, et al. Efficient inhibition of hepatitis B virus replication in vivo using Peg-modified adenovirus vectors. Hum Gene Ther. 2008;19(11):1325–31.

    PubMed  CAS  Google Scholar 

  270. Carmona S, Jorgensen MR, Kolli S, et al. Controlling HBV replication in vivo by intravenous administration of triggered PEGylated siRNA-nanoparticles. Mol Pharm. 2009;6(3):706–17.

    PubMed  CAS  Google Scholar 

  271. Morrissey DV, Lockridge JA, Shaw L, et al. Potent and persistent in vivo anti-HBV activity of chemically modified siRNAs. Nat Biotechnol. 2005;23(8):1002–7.

    PubMed  CAS  Google Scholar 

  272. Wu HL, Huang LR, Huang CC, et al. RNA interference-mediated control of hepatitis B virus and emergence of resistant mutant. Gastroenterology. 2005;128(3):708–16.

    PubMed  CAS  Google Scholar 

  273. Witting SR, Brown M, Saxena R, Nabinger S, Morral N. Helper-dependent adenovirus-mediated short hairpin RNA expression in the liver activates the interferon response. J Biol Chem. 2008;283(4):2120–8.

    PubMed  CAS  Google Scholar 

  274. Blight KJ, Kolykhalov AA, Rice CM. Efficient initiation of HCV RNA replication in cell culture. Science. 2000;290(5498):1972–4.

    PubMed  CAS  Google Scholar 

  275. Mercer DF, Schiller DE, Elliott JF, et al. Hepatitis C virus replication in mice with chimeric human livers. Nat Med. 2001;7(8):927–33.

    PubMed  CAS  Google Scholar 

  276. Yokota T, Sakamoto N, Enomoto N, et al. Inhibition of intracellular hepatitis C virus replication by synthetic and vector-derived small interfering RNAs. EMBO Rep. 2003;4(6):602–8.

    PubMed  CAS  Google Scholar 

  277. Kapadia SB, Brideau-Andersen A, Chisari FV. Interference of hepatitis C virus RNA replication by short interfering RNAs. Proc Natl Acad Sci U S A. 2003;100(4):2014–8.

    PubMed  CAS  Google Scholar 

  278. Wilson JA, Jayasena S, Khvorova A, et al. RNA interference blocks gene expression and RNA synthesis from hepatitis C replicons propagated in human liver cells. Proc Natl Acad Sci U S A. 2003;100(5):2783–8.

    PubMed  CAS  Google Scholar 

  279. Randall G, Grakoui A, Rice CM. Clearance of replicating hepatitis C virus replicon RNAs in cell culture by small interfering RNAs. Proc Natl Acad Sci U S A. 2003;100(1):235–40.

    PubMed  CAS  Google Scholar 

  280. Wang Q, Contag CH, Ilves H, Johnston BH, Kaspar RL. Small hairpin RNAs efficiently inhibit hepatitis C IRES-mediated gene expression in human tissue culture cells and a mouse model. Mol Ther. 2005;12(3):562–8.

    PubMed  CAS  Google Scholar 

  281. Yokota T, Iijima S, Kubodera T, et al. Efficient regulation of viral replication by siRNA in a non-human primate surrogate model for hepatitis C. Biochem Biophys Res Commun. 2007;361(2):294–300.

    PubMed  CAS  Google Scholar 

  282. Roth JA. Adenovirus p53 gene therapy. Expert Opin Biol Ther. 2006;6(1):55–61.

    PubMed  CAS  Google Scholar 

  283. Anderson SC, Johnson DE, Harris MP, et al. p53 gene therapy in a rat model of hepatocellular carcinoma: intra-arterial delivery of a recombinant adenovirus. Clin Cancer Res. 1998;4(7):1649–59.

    PubMed  CAS  Google Scholar 

  284. Tian G, Liu J, Zhou JS, Chen W. Multiple hepatic arterial injections of recombinant adenovirus p53 and 5-fluorouracil after transcatheter arterial chemoembolization for unresectable hepatocellular carcinoma: a pilot phase II trial. Anticancer Drugs. 2009;20(5):389–95.

    PubMed  CAS  Google Scholar 

  285. Peng Z. Current status of gendicine in China: recombinant human Ad-p53 agent for treatment of cancers. Hum Gene Ther. 2005;16(9):1016–27.

    PubMed  CAS  Google Scholar 

  286. Bischoff JR, Kirn DH, Williams A, et al. An adenovirus mutant that replicates selectively in p53-deficient human tumor cells. Science. 1996;274(5286):373–6.

    PubMed  CAS  Google Scholar 

  287. O’Shea CC, Johnson L, Bagus B, et al. Late viral RNA export, rather than p53 inactivation, determines ONYX-015 tumor selectivity. Cancer Cell. 2004;6(6):611–23.

    PubMed  Google Scholar 

  288. Reid T, Galanis E, Abbruzzese J, et al. Hepatic arterial infusion of a replication-selective oncolytic adenovirus (dl1520): phase II viral, immunologic, and clinical endpoints. Cancer Res. 2002;62(21):6070–9.

    PubMed  CAS  Google Scholar 

  289. Reid T, Galanis E, Abbruzzese J, et al. Intra-arterial administration of a replication-selective adenovirus (dl1520) in patients with colorectal carcinoma metastatic to the liver: a phase I trial. Gene Ther. 2001;8(21):1618–26.

    PubMed  CAS  Google Scholar 

  290. Habib N, Salama H, Abd El Latif Abu Median A, et al. Clinical trial of E1B-deleted adenovirus (dl1520) gene therapy for hepatocellular carcinoma. Cancer Gene Ther. 2002;9(3):254–9.

    PubMed  CAS  Google Scholar 

  291. Makower D, Rozenblit A, Kaufman H, et al. Phase II clinical trial of intralesional administration of the oncolytic adenovirus ONYX-015 in patients with hepatobiliary tumors with correlative p53 studies. Clin Cancer Res. 2003;9(2):693–702.

    PubMed  Google Scholar 

  292. Reid TR, Freeman S, Post L, McCormick F, Sze DY. Effects of Onyx-015 among metastatic colorectal cancer patients that have failed prior treatment with 5-FU/leucovorin. Cancer Gene Ther. 2005;12(8):673–81.

    PubMed  CAS  Google Scholar 

  293. Hernandez-Alcoceba R, Sangro B, Prieto J. Gene therapy of liver cancer. Ann Hepatol. 2007;6(1):5–14.

    PubMed  CAS  Google Scholar 

  294. Conlon TJ, Cossette T, Erger K, et al. Efficient hepatic delivery and expression from a recombinant adeno-associated virus 8 pseudotyped alpha1-antitrypsin vector. Mol Ther. 2005;12(5):867–75.

    PubMed  CAS  Google Scholar 

  295. Beaty RM, Jackson M, Peterson D, et al. Delivery of glucose-6-phosphatase in a canine model for glycogen storage disease, type Ia, with adeno-associated virus (AAV) vectors. Gene Ther. 2002;9(15):1015–22.

    PubMed  CAS  Google Scholar 

  296. Koeberl DD, Pinto C, Sun B, et al. AAV vector-mediated reversal of hypoglycemia in canine and murine glycogen storage disease type Ia. Mol Ther. 2008;16(4):665–72.

    PubMed  CAS  Google Scholar 

  297. Ghosh A, Allamarvdasht M, Pan CJ, et al. Long-term correction of murine glycogen storage disease type Ia by recombinant adeno-associated virus-1-mediated gene transfer. Gene Ther. 2006;13(4):321–9.

    PubMed  CAS  Google Scholar 

  298. Ziegler RJ, Bercury SD, Fidler J, et al. Ability of adeno-associated virus serotype 8-mediated hepatic expression of acid alpha-glucosidase to correct the biochemical and motor function deficits of presymptomatic and symptomatic Pompe mice. Hum Gene Ther. 2008;19(6):609–21.

    PubMed  CAS  Google Scholar 

  299. Sun B, Zhang H, Franco LM, et al. Efficacy of an adeno-associated virus 8-pseudotyped vector in glycogen storage disease type II. Mol Ther. 2005;11(1):57–65.

    PubMed  CAS  Google Scholar 

  300. Sferra TJ, Backstrom K, Wang C, Rennard R, Miller M, Hu Y. Widespread correction of lysosomal storage following intrahepatic injection of a recombinant adeno-associated virus in the adult MPS VII mouse. Mol Ther. 2004;10(3):478–91.

    PubMed  CAS  Google Scholar 

Download references

Acknowledgement

Preparation of this chapter is in part supported by the National Institution of Health (R01 DK078388) and Cystic Fibrosis Foundation (R883-CR02). The author is most grateful to Nicole Kotchey, Frank Park and Christopher Naitza for their invaluable assistance in preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Hiroyuki Nakai .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2011 Springer Science+Business Media, LLC

About this chapter

Cite this chapter

Nakai, H. (2011). Hepatic Gene Therapy. In: Monga, S. (eds) Molecular Pathology of Liver Diseases. Molecular Pathology Library, vol 5. Springer, Boston, MA. https://doi.org/10.1007/978-1-4419-7107-4_23

Download citation

  • DOI: https://doi.org/10.1007/978-1-4419-7107-4_23

  • Published:

  • Publisher Name: Springer, Boston, MA

  • Print ISBN: 978-1-4419-7106-7

  • Online ISBN: 978-1-4419-7107-4

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics