Skip to main navigation menu Skip to main content Skip to site footer

Narrative Review

Vol. 2 No. 2 (1996)

Advances in Cancer Gene Therapy

DOI
https://doi.org/10.26443/mjm.v2i2.702
Submitted
November 8, 2020
Published
2020-12-01

Abstract

N/A

References

  1. Blaese RM, Anderson WF, Culver K. The ADA human gene therapy clinical protocol. Human Gene Therapy 1: 327-62; 1990.
  2. Bordignon C, Notarangelo LD, Nobili N, et al. Gene therapy in peripheral blood lymphocytes and bone marrow for ADA- immunodeficient patients. Science 270: 470-5; 1995.
  3. Blaese RM, Culver KW, Miller AD, et al. T lymphocyte-directed gene therapy for ADA-SCID: Initial trial results after 4 years. Science 270: 475-80; 1995.
  4. James ND, Sikora K. Tumor-associated antigens. In: Lachmann PJ, Peters SK, Rosen FS, and Walport MJ, editors. Clinical Aspects of Immunology. Oxford: Blackwell Scientific Publications, 1993.
  5. Lurquin C, Van Pel A, Mariame B. Structure for the gene coding for tumor antigen P91A: a peptide encoded by the mutated exon is recognized with Ld by cytolytic T cells. Cell 58:293-303; 1989.
  6. Van Der BP., Traversari C, Chomez P, et al. A gene encoding an antigen recognized by cytotoxic T lymphocytes on a human melanoma. Science 254: 1643-8; 1991.
  7. Forni G, Varesio L, Giovarelli M, et al. Dynamic state of spontaneous immune reactivity towards a mammary adenocarcinoma. In: Spreafico F, Arnon R, editors. Tumor-Associated Antigens and Their Specific Immune Response.London: Academic Press, 1979.
  8. Pierro FD, Cavallo F, Pericle F, et al. Strategies for cytokine utilization in tumor therapy. Medical Oncology and Tumor Pharmacotherapy 10: 53-9; 1993.
  9. Zatloukal K, Schmidt W, Cotten M, et al. Somatic gene therapy for cancer: the utility of transfer infection in generating 'tumor vaccines'. Gene 135: 199-207; 1993.
  10. Zwiebel JA, Su N, MacPherson A, et al. The gene therapy of cancer: transgenic immunotherapy. Seminars in Hematology 30: 119-29; 1993.
  11. Rosenberg SA. The immunotherapy of solid cancers based on cloning the genes encoding tumor-rejection antigens. Annual Review of Medicine 47: 481-91; 1996.
  12. Graham RA, Burchell JM, Taylor-Papadimitriou J. The polymorphic epithelial mucin: Potential as an immunogen for a cancer vaccine. Cancer Immunology, Immunotherapy 42: 71-80; 1996.
  13. Culver KW. Clinical applications of gene therapy for cancer. Clinical Chemistry 40: 510-2.; 1994
  14. Pardoll D. Immunotherapy with cytokine gene-transduced tumor cells: the next wave in gene therapy for cancer. Current Opinion in Oncology 4: 1124-9; 1992.
  15. Borysiewicz LK, Fiander A, Nimako M, et al. A recombinant vaccinia virus encoding human papillomavirus types 16 and 18, E6 and E7 proteins as immunotherapy for cervical cancer. Lancet 347: 1523- 7; 1996.
  16. Schlom J, Kantor J, Abrams S, et al. Strategies for the development of recombinant vaccines for the immunotherapy of breast cancer. Breast Cancer Research and Treatment 38: 27-39; 1996.
  17. Rosenberg SA, White DE. Vitiligo in patients with melanoma: Normal tissue antigens can be targets for cancer immunotherapy. Journal of Immunotherapy 19: 81-4; 1996.
  18. Roth J, Dittmer D, Rea D, et al. p53 as a target for cancer vaccines: Recombinant canarypox virus vectors expressing p53 protect mice against lethal tumor cell challenge. Proceedings of the National Academy of Sciences of the United Steates of America 93: 4781-6; 1996.
  19. Reiser H, Schneeberger EE. Expression and function of B7-1 and B7-2 in hapten-induced contact sensitivity. European Journal of Immunology 26: 880-5; 1996.
  20. Gause WC, Urban JF, Linsley P, et al. Role of B7 signaling in the differentiation of naive CD4+ T cells to effector interleukin-4-producing T helper cells. Immunologic Research 14: 176-88; 1995.
  21. Ghiotto-Ragueneau M, Battifora M, Truneh A, et al. Comparison of CD28-B7.1 and B7.2 functional interaction in resting human T cells: Phosphatidylinositol 3-kinase association to CD28 and cytokine production. European Journal of Immunology 26: 34-41; 1996.
  22. Natesan M, Razi-Wolf Z, Reiser H. Costimulation of IL-4 production by murine B7-1 and B7-2 molecules. Journal of Immunology 156: 2783-91; 1996.
  23. Zhang YQ, Van Neerven RJJ, Kasran A, et al. Differential requirements for co-stimulatory signals from B7 family members by resting versus recently activated memory T cells towards soluble recall antigens. International Immunology 8: 37-44; 1996.
  24. Baskar S, Clements VK, Glimcher LH, et al. Rejection of MHC class II-transfected tumor cells requires induction of tumor-encoded B7-1 and/or B7-2 costimulatory molecules. Journal of Immunology 156:3821-7; 1996.
  25. Chen L, Ashe S, Brady WA. Costimulation of antitumor immunity by the B7 counter receptor for the T lymphocyte molecules CD28 and CTLA-4. Cell 71: 1093-102; 1992.
  26. Honma S, Tsukada S, Honda S. Biological-clinical significance of selective loss of HLA-class-I allelic product expression in squamous-cell carcinoma of the uterine cervix. International Journal of Cancer (United States) 57: 650-5; 1994.
  27. Haddada H, Ragot T, Cordier L, et al. Adenoviral interleukin-2 gene transfer into P815 tumor cells abrogates tumorigenicity and induces antitumoral immunity in mice. Human Gene Therapy 4: 703-11; 1993.
  28. Rosenberg SA, Anderson WF, Blaese MR, et al. Immunization of cancer patients using autologous cancer cells modified by insertion of the gene for interleukin-2. Human Gene Therapy 3: 75-90; 1992.
  29. Chaux P, Moutet M, Faivre J, et al. Inflammatory cells infiltrating human colorectal carcinomas express HLA class II but not B7-1 and B7-2 costimulatory molecules of the T-cell activation. Laboratory Investigation 74: 975-83; 1996.
  30. Ellis JR, Keating PJ, Baird J, et al. The association of an hpv16 oncogene variant with HLA-B7 has implications for vaccine design in cervical cancer. Nature Medicine 1: 464-70; 1995.
  31. La Motte RN, Rubin MA, Barr E, et al. Therapeutic effectiveness of the immunity elicited by P815 tumor cells engineered to express the B7-2 costimulatory molecule. Cancer Immunology, Immunotherapy 42: 161- 9; 1996.
  32. Dunussi-Joannopoulos K, Weinstein HJ, Nickerson PW, et al. Irradiated B7-1 transduced primary acute myelogenous leukemia (AML) cells can be used as therapeutic vaccines in murine AML. Blood 87: 2938-46; 1996.
  33. Fujii H, Inobe M, Kimura F, et al. Vaccination of tumor cells transfected with the B7-1 (CD80) gene induces the anti-metastatic effect and tumor immunity in mice. International Journal of Cancer 66: 219-24; 1996.
  34. Wang YC, Zhu LH, McHugh R, et al. Induction of autologous tumor-specific cytotoxic T-lymphocyte activity against a human renal carcinoma cell line by B7-1 (CD80) costimulation. Journal of Immunotherapy 19: 1-8; 1996.
  35. MinasiaN LM, Gansbacher B. Second International Conference on Gene Therapy of Cancer. Annual of Oncology 5: 313-6; 1994.
  36. Townsend SE, Allison JP. Tumor rejection after direct costimulation of CD8+ T cell by B7-transfected melanoma cells. Science 259: 369-71; 1993.
  37. Conry RM, LoBuglio AF, Curiel DT. Phase Ia trial of a polynucleotide anti-tumor immunization to human carcinoembryonic antigen in patients with metastatic colorectal cancer. Human Gene Therapy 7: 755- 72; 1996.
  38. Clinical Protocols. Cancer Gene Therapy 3: 265-78; 1996.
  39. Nabel GJ, Nabel EG, Yang Z, et al. Molecular genetic interventions for cancer. Cold Spring Harbor Symposia on Quantitative Biology 59: 699-707; 1994.
  40. Hersh EM, Akporiaye E, Harris D, et al. Phase I study of immunotherapy of malignant melanoma by direct gene transfer. Human Gene Therapy 5: 1371-84; 1994.
  41. Nabel GJ, Yang ZY, Nabel EG, et al. Direct gene transfer for treatment of human cancer. Ann N Y Acad Sci 772: 227-31; 1995.
  42. Nabel GJ, Nabel EG, Yang ZY, et al. Direct gene transfer with DNA-liposome complexes in melanoma: expression, biologic activity, and lack of toxicity in humans. Proceedings of the National Academy of Science of the USA 90: 11307-11; 1993.
  43. Immunotherapy of malignancy by in vivo gene transfer into tumors. Human Gene Therapy 3: 399-410; 1992.
  44. Nabel EG, Yang Z, Muller D, et al. Safety and toxicity of catheter gene delivery to the pulmonary vasculature in a patient with metastatic melanoma. Human Gene Therapy 1994;5:1089-94.
  45. Response to the points to consider for immunotherapy of malignancy by in vivo gene transfer into tumors. Human Gene Therapy 3: 705-11; 1992.
  46. Parmiani G, Colombo MP. Somatic gene therapy of human melanoma: preclinical studies and early clinical trials Somatic gene therapy of human melanoma: preclinical studies and early clinical trials. Melanoma Research 5: 295-301; 1995.
  47. Nabel GJ, Chang AE, Nabel EG, et al. Immunotherapy for cancer by direct gene transfer into tumors. Human Gene Therapy 5: 57-77; 1994.
  48. Berns AJ, Clift S, Cohen LK, et al. Phase I study of non-replicating autologous tumor cell injections using cells prepared with or without GM-CSF gene transduction in patients with metastatic renal cell carcinoma. Human Gene Therapy 6: 347-68; 1995.
  49. Poplin EA, Alberts DS, Rinehart JJ, et al. GM-CSF, carboplatin, doxorubicin: a phase I study. Cancer Chemother Pharmacol 33: 340-6; 1994.
  50. Chang AE, Sondak VK, Bishop DK, et al. Adoptive immunotherapy of cancer with activated lymph node cells primed in vivo with autologous tumor cells transduced with the GM-CSF gene. Human Gene Therapy 7: 773-92; 1996.
  51. Wong L, Taylor CW, Radwanski E, et al. Comparison of 4- and 24-hour intravenous infusion schedules for granulocyte-macrophage colony-stimulating factor. Journal of Immunotherapy with Emphasis on Tumor Immunology 18: 57-65; 1995.
  52. Gansbacher B, Houghton A, Livingston P, et al. A pilot study of immunization with HLA-A2 matched allogenic melanoma cells that secrete interleukin-2 in patients with metastatic melanoma. Human Gene Therapy 3: 677-90; 1992.
  53. Gansbacher B, Motzer R, Houghton A, et al. A pilot study of immunization with interleukin-2 secreting allogenic HIA-A2 matched renal cell carcinoma cells in patients with advanced renal cell carcinoma. Human Gene Therapy 3: 691-703; 1992.
  54. Economou JS, Figlin RA, Jacobs E, et al. The treatment of patients with metastatic melanoma and renal cell cancer using in vitro expanded and genetically-engineered (neomycin phosphotransferase) bulk, CD8(+) and /or CD4(+) tumor infiltrating lymphocytes and bulk, CD8(+) and /or CD4(+) peripheral blood leukocytes in combination with recombinant interleukin-2 alone or with recombinant interleukin-2 and recombinant alpha interferon. Human Gene Therapy 3: 411-30; 1992.
  55. Osanto S, Brouwenstyn N, Vaessen N, et al. Immunization with interleukin-2 transfected melanoma cells. a phase I-II study in patients with metastatic melanoma. Human Gene Therapy 4: 323-30; 1993.
  56. Stingl G, Brocker EB, Mertelsmann R, et al. Phase I study to the immunotherapy of metastatic malignant melanoma by a cancer vaccine consisting of autologous cancer cells transfected with the human IL-2 gene. Human Gene Therapy 7: 551-63; 1996.
  57. Sobol RE, Royston I, Fakhrai H, et al. Injection of colon carcinoma patients with autologous irradiated tumor cells and fibroblasts genetically modified to secrete interleukin-2 (IL-2): a phase I study. Human Gene Therapy 6: 195-204; 1995.
  58. Human gene therapy clinical trials in Europe. Human Gene Therapy 7: 1258-9; 1996.
  59. Suminami Y, Elder EM, Lotze MT, et al. In situ interleukin-4 gene expression in cancer patients treated with genetically modified tumor vaccine. Journal of Immunotherapy with Emphasis on Tumor Immunology 17: 238-48; 1995.
  60. Lotze MT, Rubin JT, Carty S, et al. Gene therapy of cancer: a pilot study of IL-4-gene-modified fibroblasts admixed with autologous tumor to elicit an immune response. Human Gene Therapy 5: 41-55; 1994.
  61. Cascinelli N, Foa R, Parmiani G, et al. Active immunization of metastatic melanoma patients with interleukin-4 transduced, allogenic melanoma cells. a phase I-II study. Human Gene Therapy 5: 1059-64; 1994.
  62. Mackiewicz A, Gorny A, Laciak M, et al. Gene therapy of human melanoma. immunization of patients with autologous tumor cells admixed with allogenic melanoma cells secreting interleukin 6 and soluble interleukin 6 receptor. Human Gene Therapy 6: 805-11; 1995.
  63. Schmidt-Wolf IG, Huhn D, Neubauer A, et al. Interleukin-7 gene transfer in patients with metastatic colon carcinoma, renal cell carcinoma, melanoma, or with lymphoma. Human Gene Therapy 5: 1161-8; 1994.
  64. Tahara H, Lotze MT. Antitumor effects of interleukin-12 (IL-12): applications for the immunotherapy and gene therapy of cancer. Gene Therapy 2: 96-106; 1995.
  65. Tahara H, Lotze MT, Robbins PD, et al. IL-12 gene therapy using direct injection of tumors with genetically engineered autologous fibroblasts. Human Gene Therapy 6: 1607-24; 1995.
  66. Meyskens FL, Jr., Kopecky KJ, Taylor CW, et al. Randomized trial of adjuvant human interferon gamma versus observation in high-risk cutaneous melanoma: a southwest oncology group study. Journal of the National Cancer Institute 87: 1710-3; 1995.
  67. Seigler HF, Darrow TL, Abdel-Wahab Z, et al. A phase I trial of human gamma interferon transduced autologous tumor cells in patients with disseminated malignant melanoma. Human Gene Therapy 5: 761-77; 1994.
  68. Immunization of cancer patients using autologous cancer cells modified by insertion of the gene for tumor necrosis factor. Human Gene Therapy 3: 57-73; 1992.
  69. Rosenberg SA, Anderson WF, Blaese M, et al. The development of gene therapy for the treatment of cancer. Annals of Surgery 218: 455-63; 1993.
  70. Bordignon C, Bonini C, Verzeletti S, et al. Transfer of the HSV-tk gene into donor peripheral blood lymphocytes for in vivo modulation of donor anti-tumor immunity after allogenic bone marrow transplantation. Human Gene Therapy 6: 813-9; 1995.
  71. Oldfield EH, Ram Z, Chiang Y, et al. Intrathecal gene therapy for the treatment of leptomeningeal carcinomatosis. gti 0108. a phase I/II study. Human Gene Therapy 6: 55-85; 1995.
  72. Kun LE, Gajjar A, Muhlbauer M, et al. Stereotactic injection of herpes simplex thymidine kinase vector producer cells (pa317-g1tk1svna.7) and intravenous ganciclovir for the treatment of progressive or recurrent primary supratentorial pediatric malignant brain tumors. Human Gene Therapy 6: 1231-55; 1995.
  73. Klatzmann D, Philippon J, Valery CA, et al. Gene therapy for glioblastoma in adult patients: Safety and efficacy evaluation of an in situ injection of recombinant retroviruses producing cells carrying the thymidine kinase gene of the herpes simplex type 1 virus, to be followed with the administration of ganciclovir. Human Gene Therapy 7: 109-26; 1996.
  74. Klatzmann D, Herson S, Cherin P, et al. Gene therapy for metastatic malignant melanoma: Evaluation of tolerance to intratumoral injection of cells producing recombinant retroviruses carrying the herpes simplex virus type 1 thymidine kinase gene, to be followed by ganciclovir administration. Human Gene Therapy 7: 255-67; 1996.
  75. Raffel C, Culver K, Kohn D, et al. Gene therapy for the treatment of recurrent pediatric malignant astrocytomas with in vivo tumor transduction with the herpes simplex thymidine kinase gene/ganciclovir system. Human Gene Therapy 5: 863-90; 1994.
  76. Culver KW, Van Gilder J, Link CJ, et al. Gene therapy for the treatment of malignant brain tumors with in vivo tumor transduction with the herpes simplex thymidine kinase gene/ganciclovir system. Human Gene Therapy 5: 343-79; 1994.
  77. Oldfield EH, Ram Z, Culver KW, et al. Gene therapy for the treatment of brain tumors using intra-tumoral transduction with the thymidine kinase gene and intravenous ganciclovir. Human Gene Therapy 4: 39-69; 1993.
  78. Whartenby KA, Abraham GN, Calabresi PA, et al. Gene-modified cells for the treatment of cancer. Pharmacology and Therapeutics 66: 175-90; 1995.
  79. Link CJ, Jr., Moorman D, Seregina T, et al. A phase I trial of in vivo gene therapy with the herpes simplex thymidine kinase/ganciclovir system for the treatment of refractory or recurrent ovarian cancer. Human Gene Therapy 7: 1161-79; 1996.
  80. Eck SL, Alavi JB, Alavi A, et al. Treatment of advanced CNS malignancies with the recombinant adenovirus H5.010RSVTK: A Phase I trial. Human Gene Therapy 7: 1465-82; 1996.
  81. Hesdorffer C, Antman K, Bank A, et al. Human mdr gene transfer in patients with advanced cancer. Human Gene Therapy 5: 1151-60; 1994.
  82. Deisseroth AB, Kavanagh J, Champlin R. Use of safety-modified retroviruses to introduce chemotherapy resistance sequences into normal hematopoietic cells for chemoprotection during the therapy of ovarian cancer: a pilot trial. Human Gene Therapy 5: 1507-22; 1994.
  83. Deisseroth AB, Holmes F, Hortobagyi G, et al. Use of safety-modified retroviruses to introduce chemotherapy resistance sequences into normal hematopoietic cells for chemoprotection during the therapy of breast cancer: A pilot trial. Human Gene Therapy 7: 401-16; 1996.
  84. Holt JT, Arteaga CB, Robertson D, et al. Gene therapy for the treatment of metastatic breast cancer by in vivo transduction with breast-targeted retroviral vector expressing antisense c-fos RNA. Human Gene Therapy 7: 1367-80; 1996.
  85. Johnson TR, Trojan J, Anthony DD, et al. Gene therapy of rat brain glioblastoma by an episome-based transcriptional cassette expressing antisense igf-i cDNA. Indian Journal of Biochemistry and Biophysics 31: 1-13; 1994.
  86. Bishop MR, Iversen PL, Bayever E, et al. Phase I trial of an antisense oligonucleotide ol(1)p53 in hematologic malignancies. J Clinical Oncology 14: 1320-6; 1996.
  87. Roth JA. Gene replacement strategies for cancer. Israel Journal of Medical Sciences 32: 89-94; 1996.
  88. Roth JA. Modification of tumor suppressor gene expression and induction of apoptosis in non-small cell cancer (NSCLC) with an adenovirus vector expressing wildtype p53 and Cisplatin. Human Gene Therapy 7: 1013-30; 1996.
  89. Roth JA. Modification of tumor suppressor gene expression in non-small cell lung cancer (NSCLC) with a retroviral vector expressing wildtype (normal) p53. Human Gene Therapy 7: 861-74; 1996.
  90. Sobol RE, Scanlon KJ. Cancer Gene Therapy-Clinical Perspective 1995. Cancer Gene Therapy 3: 3; 1996.
  91. Williams IR, Ort RJ, Daley D, et al. Constitutive expression of B7-1 (CD80) on mouse keratinocytes does not prevent development of chemically induced skin papillomas and carcinomas. Journal of Immunology 156: 3382-8; 1996.
  92. Li YW, Hellström KE, Newby SA, et al. Costimulation by CD48 and B7-1 induces immunity against poorly immunogenic tumors. Journal of Experimental Medicine 183: 639-44; 1996.
  93. Creery WD, Diaz-Mitoma F, Filion L, et al. Differential modulation of B7-1 and B7-2 isoform expression on human monocytes by cytokines which influence the development of T helper cell phenotype. European Journal of Immunology 26: 1273-7; 1996.
  94. Ozawa H, Aiba S, Nakagawa S, et al. Interferon-gamma and interleukin-10 inhibit antigen presentation by Langerhans cells for T helper type 1 cells by suppressing their CD80 (B7-1) expression. European Journal of Immunology 26: 648-52; 1996.
  95. Ozawa H, Nakagawa S, Tagami H, et al. Interleukin-1b and granulocyte macrophage colony-stimulating factor mediate langerhans cell maturation differently. Journal of Investigative Dermatology 106: 441-5; 1996.
  96. Rao JB, Chamberlain RS, Bronte V, et al. IL-12 is an effective adjuvant to recombinant vaccinia virus- based tumor vaccines - Enhancement by simultaneous B7-1 expression. Journal of Immunology 156: 3357- 65; 1996.
  97. Foa R, Cignetti A, Riera L, et al. Cytokine gene therapy in oncology. Folia Biologiea 40: 37-48; 1994.
  98. Hodge JW, Abrams S, Schlom J, et al. Induction of antitumor immunity by recombinant vaccinia viruses expressing B7-1 or B7-2 costimulatory molecules. Cancer Research 54: 5552-5; 1994.
  99. Arient F, Sule-Suso J, Melani C, et al. Interleukin-2 gene-transduced human melanoma cells efficiently stimulate MHC-Unrestricted and MHC-Restricted autologous lymphocytes. Human Gene Therapy 5: 1139- 50; 1994.
  100. Dranoff G, Jaffee E, Lazenby A, et al. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony- stimulating factor stimulates potent, specific, and long- lasting anti- tumor immunity. Proceedings of the Nattional Academy of Sciences of the USA 90: 3539-43; 1993.
  101. McGary CT and Welch DR. Highly metastatic 13762NF rat mammary adenocarcinoma clones secrete IL-3 or GM-CSF like activity. Proceedings for 86th American Association for Cancer Research 36: 76Abs456; 1995.
  102. Roncaroli F and Cavalieri M. Receptor for GM-CSF in axillary lymph-nodal metastasis of human breast carcinomas. Proceedings for 86th American Association for Cancer Research 36: 80Abs478; 1995.
  103. Ruco L. Tumor-infiltrating leukocytes: Histology, immunohistochemistry and in situ hybridization. In: Mantovani A, R.G.T2 - Tumor- Associated Leukocytes: Pathophysiology and Therapeutic Applications, editor. Austin: Landes Company, 1996.
  104. Hwu P, Rosenberg SA. The use of gene-modified tumor- infiltrating lymphocytes for cancer therapy. Annals of the New York Academy of Sciences 716: 188-99; 1994.
  105. Rodenberg SA, Yannelli JR, Yang JC, et al. Treatment of patient with metastatic melanoma with autologous tumor- infiltrating lymphocytes and interleukin 2. Journal of the National Cancer Institute 86: 1159-66; 1994.
  106. Nakamura Y, Wakimoto H, Abe J, et al. Adoptive immunotherapy with murine tumor-specific T lymphocytes engineered to secrete interleukin 2. Cancer Research 54: 5757-60; 1994.
  107. Rodenberg A, Anderson WF, Blaese M, et al. The development of gene therapy for the treatment of cancer. Annals of Surgery 218: 455-64; 1993.
  108. Primus FJ, Finch MD, Wetzel SA, et al. Monoclonal antibody gene transfer, implications for tumor- specific cell-mediated cytotoxicity. Ann N Y Acad Sci 716: 154-66; 1994.
  109. Moolten FL. An Alternative to the magic bullet paradigm for specific cancer therapy. Medical Hypotheses 24: 43-51; 1987.
  110. Moolten FL, Wells JM, Heyman RA, et al. Lymphoma regression induced by ganciclovir in mice bearing a herpes thymidine kinase transgene. Human Gene Therapy 1: 125-34; 1990.
  111. Huber BE, Richards CA, Krenitsky TA. Retroviral-mediated gene therapy for the treatment of hepatocellular carcinoma: an innovative approach for cancer therapy. Proceedings of the National Academy of Sciences in the United States of America. 88: 8039-43; 1991.
  112. Huber BE, Richards CA, Austin EA. Virus-directed enzyme/prodrug therapy (VDEPT). Annals of the New York Academy of Sciences 716: 104-14; 1994.
  113. Austin E, Huber B. A first step in the development of gene therapy for colorectal carcinoma: cloning, sequencing, and expression of escherichia coli cytosine deaminase. Molecular Pharmacology 43: 380-7; 1993.
  114. Culver KW, Ram Z, Wallbridge S, et al. In vivo gene transfer with retroviral vector-producer cells for treatment of experimental brain tumor. Science 256: 1550-2; 1992.
  115. Smith MJ, Rousculp MD, Goldsmith KT, et al. Surfactant protein a-directed toxin gene kills lung cancer cells in vitro. Human Gene Therapy 5: 29-35; 1994.
  116. Sikora K, Harris J, Hurst H, et al. Therapeutic strategies using c-erbb-2 promoter-controlled drug activation. Annals of the New York Academy of Sciences 716: 115-24; 1994.
  117. Chen S, Shine HD, Goodman JC, et al. Gene therapy for brain tumors: regression of experimental gliomas by adenovirus-mediated gene transfer in vivo. Proceedings of the National Academy of Science of the USA 91: 3054-7; 1994.
  118. Kumagai T, Tanio Y, Osaki T, et al. Eradication of myc-overexpressing small cell lung cancer cells transfected with herpes simplex virus thymidine kinase gene containing myc-max response elements. Cancer Research 56: 354-8; 1996.
  119. Bonnekoh B, Greenhalgh DA, Bundman DS, et al. Inhibition of melanoma growth by adenoviral- mediated hsv thymidine kinase gene transfer in vivo. Journal of Investigative Dermatology 104: 313-7; 1995
  120. Sacco MG, Mangiarini L, Villa A, et al. Local regression of breast tumors following intramammary ganciclovir administration in double transgenic mice expressing neu oncogene and herpes simplex virus thymidine kinase. Gene Therapy 2: 493-7; 1995.
  121. Hwang HC, Smythe WR, Elshami AA, et al. Gene therapy using adenovirus carrying the herpes simplex-thymidine kinase gene to treat in vivo models of human malignant mesothelioma and lung cancer. American Journal of Respiratory Cell and Molecular Biology 13: 7-16; 1995.
  122. Yoshida K, Kawami H, Yamaguchi Y, et al. Retrovirally transmitted gene therapy for gastric carcinoma using herpes simplex virus thymidine kinase gene. Cancer 75: 1467-71; 1995.
  123. O'Malley BW, Jr., Chen SH, Schwartz MR, et al. Adenovirus-mediated gene therapy for human head and neck squamous cell cancer in a nude mouse model. Cancer Research 55: 1080-5; 1995.
  124. Smythe WR, Hwang HC, Elshami AA, et al. Treatment of experimental human mesothelioma using adenovirus transfer of the herpes simplex thymidine kinase gene. Annals of Surgery 222: 78-86; 1995.
  125. Vrionis FD, Wu JK, Qi PM, et al. Tumor cells expressing the herpes simplex virus thymidine kinase gene in the treatment of Walker 256 meningeal neoplasia in rats. Journal of Neurosurgery 84: 250-7; 1996.
  126. Tong XW, Block A, Chen SH, et al. In vivo gene therapy of ovarian cancer by adenovirus-mediated thymidine kinase gene transduction and ganciclovir administration. Gynecologic Oncology 61: 175-9; 1996.
  127. Colak A, Goodman JC, Chen SH, et al. Adenovirus-mediated gene therapy in an experimental model of breast cancer metastatic to the brain. Human Gene Therapy 6: 1317-22; 1995.
  128. Colak A, Goodman JC, Chen SH, et al. Adenovirus-mediated gene therapy for experimental spinal cord tumors: tumoricidal efficacy and functional outcome. Brain Research 691: 76-82; 1995.
  129. Freeman SM, Abboud CN, Whartenby KA, et al. The "bystander effect": tumor regression when a fraction of the tumor mass is genetically modified. Cancer Research 53: 5274-83; 1993.
  130. Chen CY, Chang YN, Ryan P, et al. Effect of herpes simplex virus thymidine kinase expression levels on ganciclovir-mediated cytotoxicity and the "bystander effect". Human Gene Therapy 6: 1467-76; 1995.
  131. Kaneko Y, Tsukamoto A. Gene therapy of hepatoma: bystander effects and non-apoptotic cell death induced by thymidine kinase and ganciclovir. Cancer Letters 96: 105-10; 1995.
  132. Marini FC, 3rd, Nelson JA, Lapeyre JN. Assessment of bystander effect potency produced by intratumoral implantation of hsvtk-expressing cells using surrogate marker secretion to monitor tumor growth kinetics. Gene Therapy 2: 655-9; 1995.
  133. Fick J, Barker FG, 2nd, Dazin P, et al. The extent of heterocellular communication mediated by gap junctions is predictive of bystander tumor cytotoxicity in vitro. Proceedings of the National Academy of Science of the USA 92: 11071-5; 1995.
  134. Kuriyama S, Nakatani T, Masui K, et al. Bystander effect caused by suicide gene expression indicates the feasibility of gene therapy for hepatocellular carcinoma Bystander effect caused by suicide gene expression indicates the feasibility of gene therapy for hepatocellular carcinoma. Hepatology 22: 1838-46; 1995.
  135. Elshami AA, Kucharczuk JC, Sterman DH, et al. The role of immunosuppression in the efficacy of cancer gene therapy using adenovirus transfer of the herpes simplex thymidine kinase gene. Annals of Surgery 222: 298-10; 1995.
  136. Culver KW, Moorman DW, Muldoon RR, et al. Toxicity and immunologic effects of in vivo retrovirus- mediated gene transfer of the herpes simplex-thymidine kinase gene into solid tumors. Cold Spring Harbor Symposium on Quantitative Biology 59: 685-90; 1994.
  137. Trinh QT, Austin EA, Murray DM, et al. Enzyme/prodrug gene therapy: comparison of cytosine deaminase/5-fluorocytosine versus thymidine kinase/ganciclovir enzyme/prodrug systems in a human colorectal carcinoma cell line. Cancer Research 55: 4808-12; 1995.
  138. Bi WL, Parysek LM, Warnick R, et al. In vitro evidence that metabolic cooperation is responsible for the bystander effect observed with HSV tk retroviral gene therapy. Human Gene Therapy 4: 725-31; 1993.
  139. Barba D, Hardin J, Sadelain M, et al. Development of anti-tumor immunity following thymidine kinase- mediated killing of experimental brain tumors. Proceedings of the National Academy of Sciences of the USA 91:4348-52; 1994.
  140. Eastham JA, Hall SJ, Sehgal I, et al. In vivo gene therapy with p53 or p21 adenovirus for prostate cancer. Cancer Research 55: 5151-5; 1995.
  141. Cai DW, Mukhopadhyay T, Roth JA. Suppression of lung cancer cell growth by ribozyme-mediated modification of p53 pre-mRNA. Cancer Gene Therapy 2: 199-205; 1995.
  142. Milligan JF, Jomes RJ, Froehler BC, et al. Development of antisense therapeutics. Annals of the New York Academy of Sciences 716: 228-41; 1994.
  143. Scanlon KJ. Therapeutic application of anti-oncogene ribozymes in cancer. Proceedings for 86th American Association for Cancer Research 36: 653; 1995.
  144. Normanno N, Bianco C, Damiano V, et al. Growth inhibition of human colon carcinoma cells by anti- EGF-related growth factors antisense oligonucliotides. Proceedings for 86th American Association for Cancer Research 36: 431Abs2571; 1995.
  145. Gewirtz AM. Oligodeoxynucleotides as therapeutic agents for human leukemia. Proceedings for 86th American Association for Cancer Research 36: 654; 1995.
  146. Ohta Y, Tone T, Shitara T, et al. H-ras ribozyme-mediated alteration of the human melanoma phenotype. Annals of the New York Academy of Sciences 716: 242-56; 1994.
  147. Hynes NE, Beerli RR, Graus-Porta D. Single-chain antibody-mediated intracellular retention of ErbB-2 reverts transformation and impairs NDF and EGF signaling. Proceedings for 86th American Association for Cancer Research 36: 424Abs 2529; 1995.
  148. Friedman T. Gene therapy of cancer through restoration of tumor-suppressor function? Cancer 70 (6 suppl):1810-7; 1990.
  149. Roemer K, Friedmann T. Mechanisms of action of the p53 tumor suppressor and prospects for cancer gene therapy by reconstitution of p53 function. Annals of the New York Academy of Sciences 716: 265-82; 1994.
  150. Hamada K, Zhang W, Alemany R, et al. Growth inhibition of human cervical cancer cells by the recombinant adenovirus p53. Proceedings for 86th American Association for Cancer Research 36: 437Abs2609; 1995.
  151. Cirielli C, Pili R, Gloe TR, et al. Adenovirus-mediated gene transfer of wild-type p53 induces melanoma cell apoptosis in vitro and tumor growth inhibition in vivo. Proceedings for 86th American Association for Cancer Research 36: 421Abs2508; 1995.
  152. Fujiwara T, Cai DW, Georges RN, et al. Terapeutic effect of a retroviral wild- type p53 expression vector in an orthotopic lung cancer model. Journal of the National Cancer Institute 86: 1458-62; 1994.
  153. Carbone DP, Minna JD. in vivo gene therapy of human lung cancer using wild-type p53 delivered by retrovirus. Journal of the National Cancer Institute 86: 1437-8; 1994.
  154. Arteaga CL, Holt JT. Tissue-targeted antisense c-fos retroviral vector inhibits established breast cancer xenografts in nude mice. Cancer Research 56: 1098-103; 1996.
  155. Clayman GL, el-Naggar AK, Roth JA, et al. In vivo molecular therapy with p53 adenovirus for microscopic residual head and neck squamous carcinoma. Cancer Research 55:1-6; 1995.
  156. Kanjilal S, Strom SS, Clayman GL, et al. p53 mutations in nonmelanoma skin cancer of the head and neck: molecular evidence for field cancerization. Cancer Research 55:3604-9; 1995.
  157. Carson DA, Lois A. Cancer progression and p53. Lancet 346: 1009-11; 1995.
  158. Mujoo K, Maneval DC, Anderson SC, et al. Adenoviral-mediated p53 tumor suppressor gene therapy of human ovarian carcinoma Adenoviral-mediated p53 tumor suppressor gene therapy of human ovarian carcinoma. Oncogene 12: 1617-23; 1996.
  159. Yung WKA, Shi YX, Zhang WW, et al. Growth suppression of human glioma cells by restoration of wild-type p53 gene utilizing an adenovirus vector. Proceedings for 86th American Association for Cancer Research 36: 423Abs2524; 1995.
  160. Cirielli C, Riccioni T, Yang C, et al. Adenovirus-mediated gene transfer of wild-type p53 results in melanoma cell apoptosis in vitro and in vivo. International Journal of Cancer 63: 673-9; 1995.
  161. Zheng PS, Iwasaka T, Ouchida M, et al. Growth suppression of a cervical cancer cell line (tmcc-1) by the human wild-type p53 gene. Gynecologic Oncology 60: 245-50; 1996.
  162. Seth P, Brinkmann U, Schwartz GN, et al. Adenovirus-mediated gene transfer to human breast tumor cells: An approach for cancer gene therapy and bone marrow purging. Cancer Research 56: 1346-51; 1996.
  163. Liu TJ, el-Naggar AK, McDonnell TJ, et al. Apoptosis induction mediated by wild-type p53 adenoviral gene transfer in squamous cell carcinoma of the head and neck. Cancer Research 55: 3117-22; 1995.
  164. Gomez-Manzano C, Fueyo J, Kyritsis AP, et al. Adenovirus-mediated transfer of the p53 gene produces rapid and generalized death of human glioma cells via apoptosis. Cancer Research 56: 694-9; 1996.
  165. Schackert G, Frank S, Schackert HK. Gene therapy: Approaches for the treatment of malignant gliomas. Onkologie 19:16-22; 1996.
  166. Gjerset RA, Turla ST, Sobol RE, et al. Use of wild-type p53 to achieve complete treatment sensitization of tumor cells expressing endogenous mutant p53. Molecular Carcinogenesis 14: 275-85; 1995.
  167. Clayman GL, Liu TJ, Overholt SM, et al. Gene therapy for head and neck cancer. comparing the tumor suppressor gene p53 and a cell cycle regulator waf1/cip1 (p21). Archives of Otolaryngology - Head and Neck Surgery 122: 489-93; 1996.
  168. Runnebaum IB, Kreienberg R. p53 trans-dominantly suppresses tumor formation of human breast cancer cells mediated by retroviral bulk infection without marker gene selection: an expeditious in vitro protocol with implications towards gene therapy. Hybridoma 14: 153-7; 1995.
  169. Lesoon-Wood LA, Kim WH, Kleinman HK, et al. Systemic gene therapy with p53 reduces growth and metastases of a malignant human breast cancer in nude mice. Human Gene Therapy 6: 395-405; 1995.
  170. Zhang WW, Alemany R, Wang J, et al. Safety evaluation of ad5cmv-p53 in vitro and in vivo. Human Gene Therapy: 155-64; 1995.
  171. Da Costa LT, Jen J, He TC, et al. Converting cancer genes into killer genes. Proceedings of the National Academy of Science of the USA. 93:4192-6; 1996
  172. Wills KN, Huang WM, Harris MP, et al. Gene therapy for hepatocellular carcinoma: chemosensitivity conferred by adenovirus-mediated transfer of the HSV-1 thymidine kinase gene. Cancer Gene Therapy 1995; 2: 191-7; 1995.
  173. Tanaka T, Kanai F, Okabe S, et al. Adenovirus-mediated prodrug gene therapy for carcinoembryonic antigen-producing human gastric carcinoma cells in vitro. Cancer Research 56: 1341-5; 1996.
  174. Kaneko S, Hallenbeck P, Kotani T, et al. Adenovirus-mediated gene therapy of hepatocellular carcinoma using cancer-specific gene expression. Cancer Research 55: 5283-7; 1995.
  175. Vile R, Miller N, Chernajovsky Y, et al. A comparison of the properties of different retroviral vectors containing the murine tyrosinase promoter to achieve transcriptionally targeted expression of the HSVtk or IL-2 genes. Gene Therapy 1: 307-16; 1994.
  176. Ido A, Nakata K, Kato Y, et al. 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 1995;55:3105-9.

Downloads

Download data is not yet available.

Similar Articles

You may also start an advanced similarity search for this article.