Citation: Hubert E. Blum. Molecular Therapy and Prevention of Liver Diseases .VIROLOGICA SINICA, 2008, 23(2) : 81-92.  http://dx.doi.org/10.1007/s12250-008-2953-8

Molecular Therapy and Prevention of Liver Diseases

  • Corresponding author: Hubert E. Blum, hubert.blum@uniklinikfreiburg.de
  • Received Date: 25 November 2007
    Accepted Date: 24 January 2008
    Available online: 01 April 2008
  • Molecular analyses have become an integral part of biomedical research as well as clinical medicine. The definition of the genetic basis of many human diseases has led to a better understanding of their pathogenesis and has in addition offered new perspectives for their diagnosis, therapy and prevention. Genetically, human diseases can be classified as hereditary monogenic, acquired monogenic and polygenic diseases. Based on this classification, gene therapy is based on six concepts: (1) gene repair, (2) gene substitution, (3) cell therapy, (4) block of gene expression or function, (5) DNA vaccination and (6) gene augmentation. While major advances have been made in all areas of gene therapy during the last years, various delivery, targeting and safety issues need to be addressed before these strategies will enter clinical practice. Nevertheless, gene therapy will eventually become part of the management of patients with various liver diseases, complementing or replacing existing therapeutic and preventive strategies.

  • 加载中
    1. Alt M, Renz R, Hofschneider P H, et al. 1995. Specific inhibition of hepatitis C viral gene expression by antisense phosphorothioate oligodeoxynucleotides. Hepatology, 22: 707-717.

    2. Anderson W F. 1992. Human gene therapy. Science, 256: 808-813.
        doi: 10.1126/science.1589762

    3. Baltimore D. 1988. Intracellular immunization. Nature, 325: 395-396.

    4. Barajas M, Mazzolini G, Genove G, et al. 2001. Gene therapy of orthotopic hepatocellular carcinoma in rats using adenovirus coding for interleukin 12. Hepatology, 33: 52-61.
        doi: 10.1053/jhep.2001.20796

    5. Bartholomew R M, Carmichael E P, Findeis M A, et al. 1995. Targeted delivery of antisense DNA in woodchuck hepatitis virus-infected woodchucks. J Viral Hepatitis, 2: 273-278.
        doi: 10.1111/jvh.1995.2.issue-6

    6. Beck J, Nassal M. 1995. Efficient hammerhead ribozyme-mediated cleavage of the structured hepatitis B virus encapsidation signal in vitro and in cell extracts, but not in intact cells. Nucl Acids Res, 23: 4954-4962.
        doi: 10.1093/nar/23.24.4954

    7. Bilheimer D W, Goldstein J L, Grundy S M, et al. 1984. Liver transplantation to provide low density lipoprotein receptors and lower plasma cholesterol in a child with homozygous familial hypercholesterolemia. N Engl J Med, 311: 1658-1664.
        doi: 10.1056/NEJM198412273112603

    8. Blau H M, Springer M L. 1995. Muscle mediated gene therapy. N Engl J Med, 333: 1554-1556.
        doi: 10.1056/NEJM199512073332308

    9. Blum H E, Galun E, Weizs cker F, et al. 1991. Inhibition of hepatitis B virus by antisense oligodeoxy-nucleotides. Lancet, 337: 1230.

    10. Branch A D. 1996. A hitchhiker's guide to antisense and nonantisense biochemical pathways. Hepatology, 24: 1517-1529.
        doi: 10.1002/(ISSN)1527-3350

    11. Brown M S, Goldstein J L. 1986. A receptor-mediated pathway for cholesterol homeostasis. Science, 232: 34-37.
        doi: 10.1126/science.3513311

    12. Butterfield L H. 2004. Immunotherapeutic strategies for hepatocellular carcinoma. Gastroenterology, 127: S232-241.
        doi: 10.1053/j.gastro.2004.09.038

    13. Butterfield L H, Meng W S, Koh A, et al. 2001. T cell responses to HLA-A*0201-restricted peptides derived fromhuman alpha fetoprotein. J Immunol, 166:5300-5308.
        doi: 10.4049/jimmunol.166.8.5300

    14. Calabretta B. 1991. Inhibition of protooncogene expres-sion by antisense oligodeoxynucleotides: biological and therapeutic implications. Cancer Res, 51: 4505-4510.

    15. Cao G, Kuriyama S, Du P, et al. 1997. Complete regression of established murine hepatocellular carcinoma by in vivo tumor necrosis factor alpha gene transfer [see comments]. Gastroenterology, 112: 501-510.
        doi: 10.1053/gast.1997.v112.pm9024304

    16. Chow Y H, Huang W L, Chi W K, et al. 1997. Imp-rovement of hepatitis B virus DNA vaccines by plasmids coexpressing hepatitis B surface antigen and interleukin-2. J Virol, 71: 169-178.

    17. Davis G L. 1991. Treatment of chronic hepatitis B (editorial; comment).Hepatology, 14:567-569.
        doi: 10.1002/(ISSN)1527-3350

    18. Davis H L, Mancini M, Michel M L, et al.1996. DNA-mediated immunization to hepatitis B surface antigen-longevity of primary response and effect of boost. Vaccine, 14: 910-915.
        doi: 10.1016/0264-410X(95)00255-Y

    19. Davis H L, McCluskie M J, Gerin J L, et al. 1996. DNA vaccine for hepatitis B: Evidence for immunogenicity in chimpanzees and comparison with other vaccines. Proc Natl Acad Sci USA, 93: 7213-7218.
        doi: 10.1073/pnas.93.14.7213

    20. Davis H L, Schirmbeck R, Reimann J, et al. 1995. DNA-mediated immunization in mice induces a potent MHC class Ⅰ-restricted cytotoxic T lymphocyte response to the hepatitis B envelope protein. Human Gene Therapy, 6: 1447-1456.
        doi: 10.1089/hum.1995.6.11-1447

    21. Delaney M A, Goyal S, Seeger C. 1991. Design of modified core genes that inhibit replication of woodchuck hepatitis virus. In: , Viral hepatitis and liver disease (Hollinger F B, Lemon S M, Margolis H. ed. ). Williams & Wilkins, Baltimore, Maryland. p 667-668.

    22. Dougherty W G, Parks T D. 1995. Transgenes and gene suppression: telling us something new? Curr Opin Cell Biol, 7: 399-405.

    23. Dykxhoorn D M, Lieberman J. 2005. The silent revo-lution: RNA interference as basic biology, research tool, and therapeutic. Annu Rev Med, 56: 401-423.
        doi: 10.1146/annurev.med.56.082103.104606

    24. Folgori A, Capone S, Ruggeri L, et al. 2006. A T-cell HCV vaccine eliciting effective immunity against heterologous virus challenge in chimpanzees. Nat Med, 12: 190-197.
        doi: 10.1038/nm1353

    25. Forns X, Payette P J, Ma X. et al. 2000. Vaccination of chimpanzees with plasmid DNA encoding the hepatitis C virus (HCV) envelope E2 protein modified the infection after challenge with homologous monoclonal HCV. Hepatology, 32: 618-625.

    26. Fox I J, Chowdhury J R, Kaufman S S, et al. 1998. Treatment of the Crigler-Najjar syndrome type Ⅰ with hepatocyte transplantation. N Engl J Med, 338: 1422-1426.
        doi: 10.1056/NEJM199805143382004

    27. Friedmann, T. 1989. Progress toward human gene therapy. Science, 244: 1275-1281.
        doi: 10.1126/science.2660259

    28. Geissler M, Mohr L, Weth R, et al. 2001. Immuno-therapy directed against alpha-fetoprotein results in autoimmune liver disease during liver regeneration in mice. Gastroenterology, 121: 931-939.
        doi: 10.1053/gast.2001.28019

    29. Giladi H, Ketzinel-Gilad M, Rivkin L, et al. 2003. Small interfering RNA inhibits hepatitis B virus replication in mice. Mol Ther, 8: 769-776.
        doi: 10.1016/S1525-0016(03)00244-2

    30. Goodarzi G, Gross S C, Tewari A, et al. 1990. Antisense oligodeoxyribonucleotides inhibit the expression of the gene for hepatitis B virus surface antigen. J Gen Virol, 71: 3021-3025.
        doi: 10.1099/0022-1317-71-12-3021

    31. Grimm C F, Ortmann D, Mohr L, et al. 2000. Mouse alpha-fetoprotein-specific DNA-based immunotherapy of hepatocellular carcinoma leads to tumor regression in mice. Gastroenterology, 119: 1104-1112.
        doi: 10.1053/gast.2000.18157

    32. Grimm D, Streetz K L, Jopling C L, et al. 2006. Fatality in mice due to oversaturation of cellular microRNA/short hairpin RNA pathways. Nature, 441: 537-541.
        doi: 10.1038/nature04791

    33. Grompe M. 2001. The pathophysiology and treatment of hereditary tyrosinemia type 1. Semin Liver Dis, 21: 563-571.
        doi: 10.1055/s-2001-19035

    34. Grossman M, Rader D J, Muller D W M, et al. 1995. A pilot study of ex vivo gene therapy for homozygous familial hypercholesterolaemia. NatureMedicine, 1: 1148-1154.

    35. Grossman M, Raper S E, Kozarsky K, et al. 1994. Successful ex vivo gene therapy directed to liver in a patient with familial hypercholesterolaemia. Nat Genet, 6: 335-341.
        doi: 10.1038/ng0494-335

    36. Gutierrez A A, Lemoine N R, Sikora K. 1992. Gene therapy for cancer. Lancet, 339: 715-721.
        doi: 10.1016/0140-6736(92)90606-4

    37. Habib N, Salama H, Abd El Latif Abu Median A, et al. 2002. Clinical trial of E1B-deleted adenovirus (dl1520) gene therapy for hepatocellular carcinoma. Cancer Gene Ther, 9: 254-259.
        doi: 10.1038/sj.cgt.7700431

    38. Hallenbeck P L, Chang Y N, Hay C, et al. 1999. A novel tumor-specific replication-restricted adenoviral vector for gene therapy of hepatocellular carcinoma. Hum Gene Ther, 10:1721-1733.
        doi: 10.1089/10430349950017725

    39. Hanecak R, Brown-Driver V, Fox M C, et al.1996. Antisense oligonucleotide inhibition of hepatitis C virus gene expression in transformed hepatocytes. J Virol, 70: 5203-5212.

    40. Haseloff J, Gerlach W L. 1988. Simple RNA enzymes with new and highly specific endoribonuclease activities. Nature, 334: 585-591.
        doi: 10.1038/334585a0

    41. Heise C, Sampson-Johannes A, Williams A, et al. 1997. ONYX-015, an E1B gene-attenuated adenovirus, causes tumor-specific cytolysis and antitumoral efficacy that can be augmented by standard chemotherapeutic agents.Nature Med, 3: 639-645.
        doi: 10.1038/nm0697-639

    42. Helene C. 1991. Rational design of sequence-specific oncogene inhibitors based on antisense and antigene oligonucleotides. Eur J Cancer, 27: 1466-1471.
        doi: 10.1016/0277-5379(91)90033-A

    43. Helene C, Toulme J J. 1990. Specific regulation of gene expression by antisense, sense and antigene nucleic acids. Biochim Biophys Acta, 1049: 99-125.
        doi: 10.1016/0167-4781(90)90031-V

    44. Herschkowitz I.1987. Functional inactivation of genes by dominant negative mutations. Nature, 329: 219-222.
        doi: 10.1038/329219a0

    45. Hoeg J M, Starzl T E, Brewer H B. 1987. Liver transplantation for treatment of cardiovascular disease: comparison with medication and plasma exchange in homozygous familial hypercholesterolemia. Am J Card, 59: 705-707.
        doi: 10.1016/0002-9149(87)91199-4

    46. Huang H, Chen S H, Kosai K, et al. 1996. Gene therapy for hepatocellular carcinoma: long-term remission of primary and metastatic tumors in mice by interleukin-2 gene therapy in vivo. Gene Therapy, 3: 980-987.

    47. Ido A, Nakata K, Kato Y, et al. 1995. Gene therapy for hepatoma cells using a retrovirus vector carrying herpes simplex virus thymidine kinase gene under the control of human alpha-fetoprotein gene promoter. Cancer Research, 55: 3105-3109.

    48. Ishikawa H, Nakao K, Matsumoto K, et al. 2003. Antiangiogenic gene therapy for hepatocellular carcinoma using angiostatin gene. Hepatology, 37: 696-704.
        doi: 10.1053/jhep.2003.50077

    49. Iwashita Y, Tahara K, Goto S, et al. 2003. A Phase Ⅱ study of autologous dendritic cell-based immunotherapy for patients with unresectable primary liver cancer. Cancer Immunol Immunother, 52: 155-161.

    50. Jones J T, Lee S W, Sullenger B A. 1996. Tagging ribozyme reaction sites to follow trans-splicing in mammalian cells. Nature Med, 2: 643-648.
        doi: 10.1038/nm0696-643

    51. Kanai F, Lan K H, Shiratori Y, et al. 1997. In vivo gene therapy for alpha-fetoprotein-producing hepatocellular carcinoma by adenovirus-mediated transfer of cytosine deaminase gene. Cancer Res, 57: 461-465.

    52. Kanai F, Shiratori Y, Yoshida Y, et al. 1996. Gene therapy for alpha-fetoprotein-producing human hepatoma cells by adenovirus-mediated transfer of the herpes simplex virus thymidine kinase gene. Hepatology, 23: 1359-1368.

    53. Kaneko S, Hallenbeck P, Kotani T, et al. 1995. Adenovirus-mediated gene therapy of hepatocellular carcinoma using cancer-specific gene expression. Cancer Research, 55: 5283-5287.

    54. Kapadia S B, Brideau-Andersen A, Chisari F V. 2003. Interference of hepatitis C virus RNA replication by short interfering RNAs. Proc Natl Acad Sci USA, 100: 2014-2018.
        doi: 10.1073/pnas.252783999

    55. Kay M A, Manno C S, Ragni M V, et al.2000. Evidence for gene transfer and expression of factor Ⅸ in haemophilia B patients treated with an AAV vector. Nat Genet, 24: 257-261.
        doi: 10.1038/73464

    56. Kitabwalla M, Ruprecht R M. 2002. RNA interference-a new weapon against HIV and beyond. N Engl J Med, 347: 1364-1367.
        doi: 10.1056/NEJMcibr022294

    57. Klein C, Bock C T, Wedemeyer H, et al.2003. Inhibition of hepatitis B virus replication in vivo by nucleoside analogues and siRNA. Gastroenterology, 125: 9-18.
        doi: 10.1016/S0016-5085(03)00720-0

    58. Konishi M, Wu C H, Wu G Y. 2003. Inhibition of HBV replication by siRNA in a stable HBV-producing cell line. Hepatology, 38: 842-850.
        doi: 10.1002/(ISSN)1527-3350

    59. Kren B T, Parashar B, Bandyopadhyay P, et al. 1999. 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 USA, 96: 10349-10354.
        doi: 10.1073/pnas.96.18.10349

    60. Kroger A, Ortmann D, Krohne T U, et al. 2001. Growth suppression of the hepatocellular carcinoma cell line Hepa1-6 by an activatable interferon regulatory factor-1 in mice. Cancer Res, 61: 2609-2617.

    61. Krohne T U, Shankara S, Geissler M, et al. 2001. Mechanisms of cell death induced by suicide genes encoding purine nucleoside phosphorylase and thymidine kinase in human hepatocellular carcinoma cells in vitro. Hepatology, 34: 511-518.
        doi: 10.1053/jhep.2001.26749

    62. Kuhöber A, Pudollek H P, Reifenberg K, et al. 1996. DNA immunization induces antibody and cytotoxic T cell responses to hepatitis B core antigen in H-2b mice. J Immunol, 156: 3687-3695.

    63. Kuriyama S, Nakatani T, Masui K, et al. 1995. Bystander effect caused by suicide gene expression indicates the feasibility of gene therapy for hepatocellular carcinoma. Hepatology, 22: 1838-1846.

    64. Lagging L M, Meyer K, Hoft D, et al.1995. Immune responses to plasmid DNA encoding the hepatitis C virus core protein. J Virol, 69: 5859-5863.

    65. Lan N, Howrey R P, Lee S-W, et al. 1998. Ribozyme-mediated repair of sickle -globin mRNAs in erythrocyte precursors. Science, 280: 1593-1596.
        doi: 10.1126/science.280.5369.1593

    66. Lieber A, He C-Y, Polyak S J, et al. 1996. Elimination of hepatitis C virus RNA in infected human hepatocytes by adenovirus-mediated expression of ribozymes. J Virol, 70: 8782-8791.

    67. Liu B L, Robinson M, Han Z Q, et al. 2003. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther, 10: 292-303.

    68. Lowe S W.1997. Progress of the smart bomb cancer virus. Nature Med, 3: 606-608.
        doi: 10.1038/nm0697-606

    69. Major M E, Vitvitski L, Mink M A, et al. 1995. DNA based immunization with chimeric vectors for the induction of immune responses against the hepatitis C virus nucleocapsid. J Virol, 69: 5798-5805.

    70. Mancini M, Hadchouel M, Davis H L, et al. 1996. DNA-mediated immunization in a transgenic mouse model of the hepatitis B surface antigen chronic carrier state. Proc Natl Acad Sci USA, 93: 12496-12501.
        doi: 10.1073/pnas.93.22.12496

    71. McCaffrey A P, Nakai H, Pandey K, et al. 2003. Inhibition of hepatitis B virus in mice by RNA interference. Nat Biotechnol, 21: 639-644.
        doi: 10.1038/nbt824

    72. McDonnell W M, Askari F K. 1996. DNA vaccines. N Engl J Med, 334: 42-45.
        doi: 10.1056/NEJM199601043340110

    73. Michel M L, Davis H L, Schleef M, et al. 1995. DNA-mediated immunization to the hepatitis B surface antigen in mice: aspects of the humoral response mimic hepatitis B viral infection in humans. Proc Natl Acad Sci USA, 92: 5307-5311.
        doi: 10.1073/pnas.92.12.5307

    74. Miller A D. 1992. Human gene therapy comes of age. Nature, 357: 455-460.
        doi: 10.1038/357455a0

    75. Mizutani T, Kato N, Hirota M, et al. 1995. Inhibition of hepatitis C virus replication by antisense oligonucleotide in culture cells. Biochem Biophys Res Commun, 212: 906-911.
        doi: 10.1006/bbrc.1995.2055

    76. Mohr L, Shankara S, Yoon S K, et al.2000. Gene therapy of hepatocellular carcinoma in vitro and in vivo in nude mice by adenoviral transfer of the Escherichia coli purine nucleoside phosphorylase gene. Hepatology, 31: 606-614.
        doi: 10.1002/(ISSN)1527-3350

    77. Morgan R A, Anderson W F.1993. Human gene therapy. Annu Rev Biochem, 62: 191-217.
        doi: 10.1146/annurev.bi.62.070193.001203

    78. Mulligan R C. 1993. The basic science of gene therapy. Science, 260: 926-932.
        doi: 10.1126/science.8493530

    79. Muraca M, Gerunda G, Neri D, et al. 2002. Hepatocyte transplantation as a treatment for glycogen storage disease type 1a. Lancet, 359: 317-318.
        doi: 10.1016/S0140-6736(02)07529-3

    80. Nakazono K, Ito Y, Wu C H, et al. 1996. Inhibition of hepatitis B virus replication by targeted pretreatment of complexed antisense DNA in vitro.Hepatology, 23: 1297-1303.

    81. Offensperger W B, Offensperger S, Walter E, et al. 1993. In vivo inhibition of duck hepatitis B virus replication and gene expression by phosphorothioate modified antisense oligodeoxynucleotides.EMBO J, 12: 1257-1262.

    82. Overturf K, Al-Dhalimy M, Tanguay R, et al. 1996. Hepatocytes corrected by gene therapy are selected in vivo in a murine model of hereditary tyrosinaemia type Ⅰ. Nat Genet, 12: 266-273.
        doi: 10.1038/ng0396-266

    83. Pardoll D M, Beckerleg A. M. 1995. Exposing the immunology of naked DNA vaccines. Immunity, 3: 165-169.
        doi: 10.1016/1074-7613(95)90085-3

    84. Pawlik T M, Nakamura H, Yoon S S, et al. 2000. Oncolysis of diffuse hepatocellular carcinoma by intravascular administration of a replication-competent, genetically engineered herpesvirus. Cancer Res, 60: 2790-2795.

    85. Pei Z, Chu L, Zou W, et al. 2004. An oncolytic adenoviral vector of Smac increases antitumor activity of TRAIL against HCC in human cells and in mice. Hepatology, 39: 1371-1381.
        doi: 10.1002/(ISSN)1527-3350

    86. Qian C, Drozdzik M, Caselmann W H, et al. 2000. The potential of gene therapy in the treatment of hepatocellular carcinoma. J Hepatol, 32: 344-351.
        doi: 10.1016/S0168-8278(00)80082-3

    87. Randall G, Grakoui A, Rice C M. 2003. Clearance of replicating hepatitis C virus replicon RNAs in cell culture by small interfering RNAs. Proc Natl Acad Sci USA, 100: 235-240.
        doi: 10.1073/pnas.0235524100

    88. Rosenberg S A. 1992. Gene therapy for cancer. JAMA, 268: 2416-2419.
        doi: 10.1001/jama.1992.03490170088031

    89. Roth D A, Tawa N E, Jr O'Brien J M, et al. 2001. Nonviral transfer of the gene encoding coagulation factor Ⅷ in patients with severe hemophilia A. N Engl J Med, 344: 1735-1742.
        doi: 10.1056/NEJM200106073442301

    90. Sakamoto N, Wu C H, Wu G Y. 1996. Intracellular cleavage of hepatitis C virus RNA and inhibition of viral protein translation by hammerhead ribozymes. J Clin Invest, 98: 2720-2728.
        doi: 10.1172/JCI119097

    91. Sangro B, Mazzolini G, Ruiz J, et al. 2004. Phase Ⅱ trial of intratumoral injection of an adenovirus encoding interleukin-12 for advanced digestive tumors. J Clin Oncol, 22: 1389-1397.
        doi: 10.1200/JCO.2004.04.059

    92. Sarver N, Cairns S. 1996. Ribozyme trans-splicing and RNA tagging: Following the messenger. Nature Med, 2: 641-642.
        doi: 10.1038/nm0696-641

    93. Scaglioni P, Melegari M, Takahashi M, et al. 1996. Use of dominant negative mutants of the hepadnaviral core protein as antiviral agents. Hepatology, 24:1010-1017.
        doi: 10.1002/(ISSN)1527-3350

    94. Scaglioni P P, Melegari M, Wands J R. 1994. Characterization of hepatitis B virus core mutants that inhibit viral replication. Virology, 205: 112-120.
        doi: 10.1006/viro.1994.1625

    95. Schirmbeck R, Bohm W, Ando K, et al. 1995. Nucleic acid vaccination primes hepatitis B virus surface antigen specific cytotoxic T lymphocytes in nonresponder mice. J Virol, 69: 5929-5934.

    96. Seki T, Kiyosawa K, Inoko H, et al. 1990. Association of autoimmune hepatitis with HLA-Bw54 and DR4 in Japanese patients. Hepatology, 12: 1300-1304.
        doi: 10.1002/(ISSN)1527-3350

    97. Shlomai A, Shaul Y. 2003. Inhibition of hepatitis B virus expression and replication by RNA interference. Hepatology, 37: 764-770.
        doi: 10.1053/jhep.2003.50146

    98. Taylor J A, Naoumov N V. 2005. The potential of RNA interference as a tool in the management of viral hepatitis. J Hepatol, 42: 139-144.

    99. Thompson J D, Macejak D, Couture L, et al. 1995. Ribozymes in gene therapy. Nature Med, 1: 277-278.
        doi: 10.1038/nm0395-277

    100. Tseng B Y, Brown K D. 1994. Antisense oligonucleotide technology in the development of cancer therapeutics. Cancer Gene Ther, 1: 65-71.

    101. Tuerk C, Gold L. 1990. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science, 249: 505-510.
        doi: 10.1126/science.2200121

    102. Uprichard S L, Boyd B, Althage A, et al. 2005. Clearance of hepatitis B virus from the liver of transgenic mice by short hairpin RNAs. Proc Natl Acad Sci USA, 102: 773-778.
        doi: 10.1073/pnas.0409028102

    103. Vidalin O, Major M E, Rayner B, et al. 1996. In vitro inhibition of hepatitis C virus gene expression by chemically modified antisense oligodeoxynucleotides. Antimicrobial Agents & Chemotherapy, 40: 2337-2344.

    104. Vitiello A, Ishioka G, Grey H M, et al. 1995. Development of a lipopeptide based therapeutic vaccine to treat chronic HBV infection. I. Induction of a primary cytotoxic T lymphocyte response in humans. J Clin Invest, 95: 341-349.

    105. von Weizsäcker F, Blum H E, Wands J R. 1992. Cleavage of hepatitis B virus RNA by three ribozymes transcribed from a single DNA template. Biochem Biophys Res Commun, 189: 743-748.
        doi: 10.1016/0006-291X(92)92264-X

    106. von Weizsäcker F, Wieland S, Blum H E. 1996. Inhibition of viral replication by genetically engineered mutants of the duck hepatitis B virus core protein. Hepatology, 24: 294-299.

    107. Wagner R W. 1994. Gene inhibition using antisense oligodeoxynucleotides. Nature, 372: 333-335.
        doi: 10.1038/372333a0

    108. Wakita T, Wands J R. 1994. Specific inhibition of hepatitis C virus expression by antisense oligodeoxy-nucleotides. In vitro model for selection of target sequence. J Biol Chem, 269: 14205-14210.

    109. Wands J R, Lavaissiere L, Moradpour D, et al. 1997. Immunological approach to hepatocellular carcinoma. Journal of Viral Hepatitis, 4 (Suppl. 2): 60-74.

    110. Welch P J, Tritz R, Yei S, et al. 1996. A potential therapeutic application of hairpin ribozymes -in vitro and in vivo studies of gene therapy for hepatitis C virus infection.Gene Therapy, 3: 994-1001.

    111. Williams B R. 2005. Targeting specific cell types with silencing RNA. N Engl J Med, 353: 1410-1411.
        doi: 10.1056/NEJMcibr052492

    112. Wills K N, Huang W M, Harris M P, et al. 1995. Gene therapy for hepatocellular carcinoma: chemosensitivity conferred by adenovirus-mediated transfer of the HSV-1 thymidine kinase gene. Cancer Gene Therapy, 2: 191-197.

    113. Wilson J A, Jayasena S, Khvorova A, et al. 2003. RNA interference blocks gene expression and RNA synthesis from hepatitis C replicons propagated in human liver cells. Proc Natl Acad Sci USA, 100: 2783-2788.
        doi: 10.1073/pnas.252758799

    114. Wu G Y, Wu C H. 1992. Specific inhibition of hepatitis B viral gene expression in vitro by targeted antisense oligonucleotides. J Biol Chem, 267: 12436-12439.

    115. Xu G W, Sun Z T, Forrester K, et al. 1996. Tissue-specific growth suppression and chemosensitivity prom-otion in human hepatocellular carcinoma cells by retroviral-mediated transfer of the wild-type p53 gene. Hepatology, 24: 1264-1268.
        doi: 10.1002/(ISSN)1527-3350

    116. Yao Z, Zhou Y, Feng X, et al. 1996. In vivo inhibition of hepatitis B viral gene expression by antisense phosphoro-thioate oligodeoxynucleotides in athymic nude mice. J Viral Hepatitis, 3: 19-22.
        doi: 10.1111/jvh.1996.3.issue-1

    117. Ying C, De Clercq E, Neyts J. 2003. Selective inhibition of hepatitis B virus replication by RNA interference. Biochem Biophys Res Commun, 309: 482-484.
        doi: 10.1016/j.bbrc.2003.08.021

    118. Zender L, Kock R, Eckhard M, et al. 2002. Gene therapy by intrahepatic and intratumoral trafficking of p53-VP22 induces regression of liver tumors. Gastroen-terology, 123: 608-618.
        doi: 10.1053/gast.2002.34756

  • 加载中

Tables(2)

Article Metrics

Article views(4593) PDF downloads(11) Cited by()

Related
Proportional views

    Molecular Therapy and Prevention of Liver Diseases

    Abstract: Molecular analyses have become an integral part of biomedical research as well as clinical medicine. The definition of the genetic basis of many human diseases has led to a better understanding of their pathogenesis and has in addition offered new perspectives for their diagnosis, therapy and prevention. Genetically, human diseases can be classified as hereditary monogenic, acquired monogenic and polygenic diseases. Based on this classification, gene therapy is based on six concepts: (1) gene repair, (2) gene substitution, (3) cell therapy, (4) block of gene expression or function, (5) DNA vaccination and (6) gene augmentation. While major advances have been made in all areas of gene therapy during the last years, various delivery, targeting and safety issues need to be addressed before these strategies will enter clinical practice. Nevertheless, gene therapy will eventually become part of the management of patients with various liver diseases, complementing or replacing existing therapeutic and preventive strategies.

    • Molecular biology and recombinant DNA tech-nology increasingly contribute to the diagnosis, therapy and prevention of human diseases. Molecular methods allow the early and/ or specific detection of inherited, infectious and malignant liver diseases. In addition, such analyses increasingly lead to a better understanding of the pathogenesis of the various liver diseases which in turn had an impact on patient management, including the presymptomatic identifi-cation of patients at risk, the correct staging of the disease and the follow-up of patients undergoing therapy. Thus, molecular biology is increasingly becoming an integral part of basic as well as clinical hepatology. In the following we will briefly review current concepts and potential applications of gene therapy for the treatment or prevention of various liver diseases.

    • Genetically, human diseases can be classified into three major categories (2, 27, 36, 74): (1) Hereditary monogenic diseases that are caused by a single gene defect and inherited by the classical Mendelian rules. There are more than 4, 000 monogenic diseases described. For an increasing number of these diseases the genetic basis is being identified. (2) Acquired monogenic diseases are infections as well as malignancies that are caused by the mutation or epigenetic modification of a single gene. (3) polygenic diseases are associated with mutations of several genes that are acquired and frequently accumulated during life-time. Several common human diseases belong to this category, such as most malignancies.

      Gene therapy is defined as the introduction of genetic material into human cells with a therapeutic or preventive benefit. In a broader definition, cell or organ transplantation are included. In the following we will discuss the basic concepts of gene therapy (2, 27, 36, 74, 77, 78, 88) as well as some therapeutic and preventive applications for liver diseases.

    • Based on the genetic classification of diseases detailed above, the principle of gene therapy involves 6 concepts (Table 1): gene repair, gene substitution and cell therapy for hereditary monogenic diseases, block of gene expression and DNA vaccination for acquired monogenic diseases and gene augmentation and DNA vaccination for polygenic diseases. For clinical applications, gene therapy is explored with the aim to either provide novel therapeutic strategies for diseases for which there is no treatment available or to replace and in some cases complement xisting treatment modalities, thereby increasing therapeutic efficacy and/ or reduce adverse events.

      Table 1.  Concepts of Gene Therapy

    • An increasing number liver diseases has been molecularly defined as a defect of a single gene (Table 2). In this context, one therapeutic concept is the in vitro or in vivo repair of the defective gene. Indeed, in the Gunn rat model of the Crigler Najjar syndrome type Ⅰ Kren et al. (50, 59, 65, 92) were able to partially correct the genetic defect underlying the UDP-glucuronosyl transferase deficiency by the intravenous injection of a cyclic normal/ wild-type chimeric oligonucleotide. While these findings have not been independently confirmed or extended to other hereditary monogenic (liver) diseases, the data suggest that it is in principle possible to repair a gene defect in vivo. Further, it has been shown that cellular RNA species can be modified by trans-splicing group Ⅰ ribozymes. Such ribozymes may in principle allow to treat a variety of inherited diseases at the RNA level (50, 65, 92).

      Table 2.  Hereditary Monogenic Liver Diseases (selection)

    • The targeted substitution of a defective cellular gene by the normal/ wild-type homologue with pro-duction of the physiological gene product is another approach to correct a hereditary or acquired mono-genic gene defect. Indeed, in an animal model of here-ditary tyrosinemia type 1 (HT1), a liver disease caused by a deficiency of fumarylacetoacetate hydrolase (FAH), multiple injections of a retroviral vector carrying the FAH gene resulted in a gene transfer efficiency of > 90% of hepatocytes and the restoration of a normal liver function (33, 82). In patients, examples for gene substitution are the partial cor-rection of severe hemophilia A by the ex vivo transduction of autologous skin fibroblasts with the normal/wild-type factor Ⅷ gene, followed by la-paroscopic implantation of the genetically modified fibroblasts into the omentum majus (89) or of hemo-philia B by adenovirus-associated vector (AAV)-based gene transfer (55).

      In rare situations in which a hepatocelluar car-cinoma (HCC) is caused by the mutation of a tumor suppressor gene, e.g., the p53 gene, the substitution of the mutated by the normal/wild-type gene in vitro can reduce the number of tumor cell colonies and restore cisplatin sensitivity (115, 118).

    • Allogeneic or ex vivo genetically modified auto-logous hepatocyte transplantation is a promising strategy to treat hereditary monogenic liver diseases. In patients with familial hypercholesterolemia (FH) that is caused by various mutations in the low density lipoprotein (LDL) receptor gene (11), apart from orthotopic liver transplantation (7, 45), liver-directed gene therapy has been performed in a pilot study in five patients (34, 35). Autologous liver cells, prepared from a surgical biopsy, were transduced ex vivo with a recombinant retrovirus expressing the normal LDL receptor. These ex vivo genetically modified hepa-tocytes were transplanted by portal infusion and resulted in significant and prolonged reductions in LDL cholesterol in 3/ 5 patients for at least four months, demonstrating the feasibility of engrafting a limited number of ex vivo transduced hepatocytes. Also, allogeneic hepatocyte transplantation has been successfully used in patients to partially correct Crigler-Najjar syndrome type Ⅰ (26) and glycogen storage disease type Ⅰ (79).

    • For diseases caused by the expression of an acquired gene or the overexpression of an endogenous gene, blocking gene expression can be an effective therapeutic approach. Several strategies can be em-ployed: interference with the transcription of genes by binding of transcription factors to nucleic acids introduced into or synthesized in the cells (decoy strategy) (42, 43), by binding of single-stranded nucleic acids to double-stranded DNA, forming a triple helix structure (42, 43), hybridization of RNA molecules possessing endonuclease activity (ribozymes) to RNA, resulting in its sequence-specific cleavage (40, 99), RNA interference (RNAi) by small inhibiting RNA (siRNA) or microRNA (miRNA) (23, 56, 98, 111) block of translation by antisense oligonucleotides (14, 42, 43, 100) and the intracellular synthesis of peptides or proteins, interfering with their normal counterpart, termed dominant negative (DN) mutant strategy (44). These different strategies have been applied to a number of malignant and infectious diseases. In particular ribozymes, siRNAs, antisense oligonucleotides and DN mutants have been experi-mentally explored to treat hepatitis B virus (HBV) and hepatitis C virus (HCV) infections.

      Ribozymes. Ribozymes ('ribonucleic acid enzymes') were originally discovered as naturally occurring RNA molecules that catalyze the sequence-specific cleavage of RNA and RNA splicing reactions (40, 99). This catalytic activity is the major attraction of the ribozyme concept since one ribozyme can cleave many target RNAs. Ribozymes that cleave RNA are being developed as inhibitors of gene expression and viral replication. Several studies have clearly demon-strated that hammerhead ribozymes can specifically cleave HBV RNA (6, 105) or HCV RNA (66, 90) in vitro. in vivo, however, an efficient ribozyme-mediated cleavage of HBV RNA could not be demonstrated to date. For HCV infection, the elimi-nation of HCV RNA in infected hepatocytes by ribozymes has also been reported (66, 110).

      Small interfering RNA. RNAi is a recently discovered basic intracellular mechanism (23, 56, 98, 111) that has been explored also for the inhibition of HBV and HCV infection. For HBV, inhibition of viral gene expression and replication has been shown in vitro (58, 97, 117) and in different mouse models in vivo (29, 32, 57, 71, 102). For HCV, inhibition of viral gene expression and replication has been shown in vitro in the replicon system (54, 87, 113). While effective in blocking viral gene expression and replication, in vivo oversaturation of celluar miRNA/ short hairpin RNA (shRNA) pathways can result in lethal hepatotoxicity (32). For future RNAi-based strategies in animals or humans, these findings indicate that the control of intracellular shRNA expression levels through optimizing shRNA dose and sequence will be key to reduce the risk of over-saturating endogenous small RNA pathways.

      Antisense oligonucleotides. Antisense nucleic acids are designed to specifically bind to RNA or mRNA, resulting in the formation of RNA-DNA (antisense oligodeoxynucleotides) or RNA-RNA hybrids (anti-sense oligoribonucleotides) with an arrest of RNA replication, reverse transcription or mRNA translation (14, 42, 43, 100, 107). Antisense effects can be potentiated by degradation of RNA in RNA-DNA hybrids by cellular RNases H. While conceptually simple, it is clear now that not all desired as well as undesired effects are caused by the target sequence specific antisense action of the oligonucleotides or the cellular enzymes mentioned above (10, 22).

      The antisense strategy has been successfully applied in vitro to HBV infection (9, 30, 80, 114) and to HCV infection (1, 39, 75, 96, 103, 108). In addition, studies in nude mice (116), in the duck hepatitis B virus (DHBV) (81) and the woodchuck hepatitis virus (WHV) model of HBV infection (5) demonstrated the in vivo applicability of this approach. While no toxic effects have been observed in these experiments, the contribution of non-antisense effects to the inhibition of viral replication or gene expression has not been systematically assessed in most studies. Independent of the antisense or non-antisense mechanism of the biological effects, an in vitro screening procedure for the identification of functionally active oligonucleotides (10, 101) should greatly facilitate the design of oligonucleotide based antiviral therapies.

      Interfering peptides or proteins. The intracellular synthesis of interfering peptides or proteins, including single chain or whole non-secreted antibodies, is aimed at the specific interference with the assembly or function of viral structural or non-structural proteins and represents a type of intracellular immunization (3). This approach has been shown for block mammalian and avian hepadnavirus gene expression and repli-cation in vitro. For example, the fusion of different polypeptides of various lengths to the carboxy-terminus of the viral core protein yields DN mutants (21, 93, 94, 106). These DN mutants are species-specific and suppress viral replication by at least 90 % at an effector to target ratio of 1:10. Moreover, the non-secretory form of the hepatitis B e antigen (HBeAg) was shown to effectively inhibit viral replication and may indeed act as a natural regulator of HBV propagation (14, 42, 43, 100). The potential advantage of DN mutants over ribozymes or antisense oligonucleotides is their relative independence from viral sequence variations, minimizing the risk of selecting or accumulating 'therapy escape' mutants.

    • A novel approach is DNA vaccination resulting in the manipulation of the immune system by intro-duction of expression vectors into muscle cells or dendritic cells and long lasting cellular and humoral immune responses. The direct gene transfer into muscle (8) represents an exciting new development and elegant application of gene therapy (72, 83). The therapeutic DNA vaccine acts by the intracellular plasmid-derived synthesis of a viral protein which enters the cell's MHC class Ⅰ pathway (72). Only proteins that originate within the cell can be processed by MHC class Ⅰ molecules that carry fragments of the protein to the cell surface. There they stimulate CD8+ cytotoxic T cells, resulting in cell-mediated immunity. In principle, this strategy is applicable to the treatment of acquired genetic diseases, associated with the expression of disease-specific antigens serving as targets for CD8+ cytotoxic T cells.

      Therapeutic DNA vaccination has been experi-mentally explored for HBV (20, 62, 70, 95, 104) as well as HCV infection (64, 69) and holds great promise as an effective molecular therapy for these viral diseases. In this context, the coexpression of HBsAg and interleukin-2 was shown to greatly increase humoral as well as cellular immune response (16).

      Further, DNA-based tumor vaccination against HCC may be possible, for example, by intramuscular introduction of a plasmid expressing HCC-specific antigens or antigens that are highly overexpressed in HCC cells, such as AF-20 antigen, insulin receptor substrate-1 (109) alpha-fetoprotein (31), aspartyl asparaginyl hydroxylase, mutated p53 protein and others. Potential limitations of this strategy include the regulation of the immune response as well as the low level expression of the targeted antigen in non-malignant cells (28), rendering them susceptible to immune mediated elimination as well.

    • Polygenic diseases are among the most prevalent clinical problems. In this situation, gene augmentation is aimed at the local expression of a therapeutic gene product that is physiologically not expressed or expressed at therapeutically insufficient levels. This strategy is explored among others for the treatment of hepatocellular carcinoma (HCC). Gene augmentation is aimed at the local expression of a therapeutic gene product that is physiologically not expressed or expressed at therapeutically insufficient levels.

      Suicide gene therapy. An interesting strategy to treat HCCs is genetic prodrug activation therapy via the introduction of a 'suicide gene' into malignant cells followed by the administration of the prodrug. This concept has been experimentally explored in HCC cells in vitro and in vivo, e.g., for the HSV-tk gene (47, 52, 53, 86, 112) the gene encoding cytosine deaminase (CD) that converts the prodrug 5-fluorocytosine to 5-fluorouracil which inhibits RNA and DNA synthesis during the S-phase of the cell cycle (51), the gene encoding purine nucleoside phosphorylase that converts purine analogs into freely diffusible toxic metabolites (61, 76) as well as the gene encoding cytochrome p450 4B1 (76). A significant bystander effect of cell killing caused by suicide gene expression could be demonstrated in vitro and in vivo, based on cell-cell contact rather than release of cytotoxic substances from the transduced cells (63). At the same time, the bystander effect may also affect non-malignant dividing cells in the target tissue, poten-tially limiting the application of this strategy.

      Immune therapy. In the process of malignant transformation new antigenic surface proteins can be expressed (tumor antigens) or oncofetal proteins can be re-expressed, e.g., alpha fetoprotein (AFP).

      AFP-specific immune therapy has been explored in mice and humans. Vaccination with an AFP-expres-sing DNA construct resulted in tumor rejection and prolonged survival in a mouse model (31). Also in patients AFP-specific T cells could be detected (12, 13). Since AFP is not only expressed by tumor cells but also by regenerating liver cells and in liver cirrhosis immunization against AFP carries the risk of autoimmune hepatitis, as has been experimentally shown in mice (28).

      Immune therapy with antigen presenting cells (APC) is another strategy that has been explored using dendritic cells (DC) exposed to tmor lysates, peptides or ex vivo tranduced with tumor antigen expressing DNA constructs. While this strategy is cenceptually very interesting, to date there are no data available that demonstrate it clinical efficacy (49).

      Cytokine gene therapy has been explored using tumor necrosis factor (TNF)-alpha, GM-CSF, inter-feron-apha or -gamma, interleukin (IL)-2, -4, -6, -7, -12 and -18, B7-1 as well as CD40 ligand. Complete regression of a HCC was demonstrated in vivo by TNF-alpha (15), IL-2 (46), IL-12 (4) and an activa-table interferon regulatory factor-1 in mice (60). Gene transfer was achieved in vivo by delivering retroviral (15) or adenoviral vectors (46) systemically, directly into the tumor or into the peritoneal cavity. A pilot study in patients with gastrointestinal tumors ex-ploring the intratumoral injection of an adenoviral IL-12 expression construct showed only marginal efficacy, however (91).

      Antiangiogenic gene therapy. This concept has been experimentally explored in a HCC mouse model using the angiostatin gene. Angiostatin gene transfer resulted in reduced tumor volume and vascular density (48).

      Oncolytic viruses. Such a new and elegant approach uses p53 mutations for selective, adenovirus-mediated lysis of tumor cells. Thus, an adenovirus mutant was engineered that replicates selectively in p53-deficient human tumor cells (37, 41, 68). Other examples are the adenoviral introduction of Smac antagonizes the inhibitor of apoptosis proteins in HCC tumor cells and enhances tumor cell death (85) and tumor-specific replication-restricted adenoviral vectors (38). Further, the intravascular administration of a replication-competent genetically engineered herpes simplex virus (HSV)-1 resulted in oncolysis of a diffuse HCC (84). More efficient HSV-1-based vectors have been developed (67).

    • DNA-based prophylactic vaccination against HBV infection, for example, is possible by intramuscular introduction of a plasmid expressing hepatitis B surface antigen (HBsAg). HBsAg is taken up by cells via phagocytosis or endocytosis, processed through the major histocompatibility complex (MHC) class Ⅰ¡ system and primarily stimulates an antibody response through CD4+ helper T cells with the production of anti-HBs (17-19, 72, 73, 83). While the DNA-based vaccination against HBV infection induces anti-HBs antibodies and prevents HBV infection, DNA-based vaccination against HCV infection of chimpanzees has been shown not to prevent infection but to result in tghe rsolution of acute HCV infection through an effective vaccine-induced cellular imune response (24, 25).

    • Molecular analyses have become an integral part of biomedical research as well as clinical medicine. The definition of the genetic basis of many human diseases has led to a better understanding of their pathogenesis and has in addition offered new perspectives for their diagnosis, therapy and prevention. Genetically, human diseases can be classified as hereditary monogenic, acquired monogenic and polygenic diseases. Based on this classification, gene therapy is based on four concepts: gene repair, gene substitution, cell or organ transplanation, block of gene expression or function, gene augmentation and DNA vaccination. While the recent developments in gene therapy for liver diseases are promising, various delivery, targeting and safety issues need to be addressed before these strategies will enter clinical practice. Nevertheless, gene therapy will become part of the management of patients with liver diseases, complementing existing diagnostic, thera-peutic and preventive strategies.

    Table (2) Reference (118) Relative (20)

    目录

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return