14
Views
1
CrossRef citations to date
0
Altmetric
Review

Advances in cancer gene therapy

, &
Pages 711-735 | Published online: 25 Feb 2005

Bibliography

  • WEINBERG RA: How cancer arises. ScL Am. (1996) 275(3):62–70.
  • ANDERSON WF: Human gene therapy. Nature (1998) 392(Suppl) 6679):25–30.
  • ••This review evaluates the problems and the potentialsolutions of gene therapy as a new field of medicine. The problem lies in the development of safe and efficient gene-delivery systems.
  • ROTH JA, CRISTIANO RJ: Gene therapy for cancer: what have we done and where are we going. J. Natl. Cancer Inst. (1997) 89(1):21–39.
  • GIOVANNANGELI C, HELENE C: Progress in develop-ments of triplex-based strategies. Antis ense Nucleic Acid Drug Dev. (1997) 7(4):413–421.
  • VASQUEZ KM, WILSON JH: Triplex-directed modifica-tion of genes and gene activity. Trends Biochem. ScL (1998) 23(0:4–9.
  • EBBINGHAUS SW, FORTINBERRY H, GAMPER HBJ:Inhibition of transcription elongation in the HER-2/neu coding sequence by triplex-directed covalent modification of the template strand. Biochem-istry (1999) 38(2):619–628.
  • •These studies demonstrated the successful application of triplex-forming oligonucleotide-alkylator conjugates to inhibit transcription elongation in the HER-2h7eu gene, and show that covalent modification of the DNA strand used as the transcription template is necessary to prevent RNA polymerase elongation.
  • SHEVELEV A, BURFEIND P, SCHULZE E et al. Potential triple helix-mediated inhibition of IGF-I gene expres-sion significantly reduces tumorigenicity of glioblas-toma in an animal model. Cancer Gene Ther. (1997) 4(2):105–112.
  • RININSLAND F, JOHNSON TR, CHERNICKY CL et al.: Suppression of insulin-like growth factor Type I receptor by a triple- helix strategy inhibits IGF-I transcription and tumorigenic potential of rat C6 glioblastoma cells. Proc. Natl. Acad. Sci. USA (1997) 94(105854–5859.
  • ••These results suggested that a triple-helix strategy can beused to inhibit transcription elongation of the IGF-IR gene, and emphasise the efficacy of triplex-mediated gene inhibi-tion in an animal model.
  • GEWIRTZ AM, SOKOL DL, RATAJCZAK MZ: Nucleic acid therapeutics: state of the art and future prospects. Blood (1998) 92 (3):712–736.
  • ORR RM, MONIA BP: Antisense therapy for cancer. Curr. Res. Mol. Ther. (1998) 1(2):102–108.
  • SOMMER W, HEILIG M: Antisense oligonucleotides are clinically tested. They inhibit the expression of disease-related genes. Lakartidningen (1999) 96(4)348–354.
  • NARAYANAN R, AKHTAR S: Antisense therapy. Curr. Opin. Oncol. (1996) 8(6):509–515.
  • •This paper provided a strong rationale for identification and validation of disease targets for antisense therapy. Analogues modified from normal phosphodiester oligode-oxynucleotides have entered clinical trials of diseases including AIDS, cancer, and inflammation.
  • WATSON PH, PON RT, SHIU RP: Inhibition of c-myc expression by phosphorothioate antisense oligonu-cleotide identifies a critical role for c-myc in the growth of human breast cancer. Cancer Res. (1991) 51(15)3996–4000.
  • SALEH M, STACKER SA, WILKS AF: Inhibition of growth of C6 glioma cells in vivo by expression of antisense vascular endothelial growth factor sequence. Cancer Res. (1996) 56(2):393–401.
  • CHENG SY, HUANG HJ, NAGANE M eta].: Suppression of glioblastoma angiogenicity and tumorigenicity by inhibition of endogenous expression of vascular endothelial growth factor. Proc. Natl. Acad. Sci. USA (1996) 93(16):8502–8507.
  • ALEMANY R, RUAN S, KATAOKA M et al Growth inhibi-tory effect of anti-K-ras adenovirus on lung cancer cells. Cancer Gene Ther. (1996) 3(5):296–301.
  • LAFARGE-FRAYSSINET C, DUC HT, FRAYSSINET C et al:Antisense insulin-like growth factor I transferred into a rat hepatoma cell line inhibits tumorigenesis by modulating major histocompatibility complex I cell surface expression. Cancer Gene Ther. (1997) 4(5):276–285.
  • HAMADA K, SAKAUE M, ALEMANY R et al: Adenovirus-mediated transfer of HPV 16 E6/E7 antisense RNA to human cervical cancer cells. Gynecol. Oncol (1996) 63(2)219–227.
  • BURFEIND P, CHERNICKY CL, RININSLAND F et al.:Antisense RNA to the Type I insulin-like growth factor receptor suppresses tumor growth and prevents invasion by rat prostate cancer cells in vivo. Proc. Natl. Acad. Sci. USA (1996) 93(14):7263–7268.
  • GEORGES RN, MUKHOPADHYAY T, ZHANG Y et al. Prevention of orthotopic human lung cancer growth by intratracheal instillation of a retroviral antisense K-ras construct. Cancer Res. (1993) 53(8):1743–1746.
  • KASHLES O, YARDEN Y, FISCHER R et al.: A dominant negative mutation suppresses the function of normal epidermal growth factor receptors by heterodimeriza-tion. Mol. Cell Biol. (1991) 11(3):1454–1463.
  • MILLAUER B, LONGHI MP, PLATE KH et al. Dominant- negative inhibition of Flk-1 suppresses the growth of many tumor types in vivo. Cancer Res. (1996) 56(7) 1615–1620
  • MILLAUER B, SHAWVER LK, PLATE KH et al. Glioblas- toma growth inhibited in vivo by a dominant-negative Flk-1 mutant. Nature (1994) 367(6463):576–579.
  • DUNN SE, EHRLICH M, SHARP NJ et al. A dominant negative mutant of the insulin-like growth factor-I receptor inhibits the adhesion, invasion, and metastasis of breast cancer. Cancer Res. (1998) 58(15) 3353–3361
  • PELEGRIN M, MARIN M, NOEL D, PIECHACZYK M: Geneti- cally engineered antibodies in gene transfer and gene therapy. Human Gene Ther. (1998) 9:2165–2175.
  • ••Four of the major applications of antibody gene engineeringin the field of gene therapy are reviewed here. These are (1) the redefinition of viral vector tropism of infection for better transduction of cells of therapeutical interest, (2) the grafting of new cell recognition activities to effector cells of the immune system to kill cancer and pathogen-infected cells, (3) the inhibition of cellular and viral functions through intracellular expression of antibody-derived molecules, and (4) the systemic delivery of therapeutic monoclonal antibodies by non-B cells in living organisms.
  • JONES SD, MARASCO WA. Antibodies for targeted gene therapy: extracellular gene targeting and intracellular expression. Adv. Drug Deliv. Rev. (1998) 31:153–170.
  • HUSTON JS, MCCARTNEY J, TAI MS et al.: Medical applications of single-chain antibodies. Intern. Rev. Immunol. (1993) 10(2-3):195–217.
  • CHEN SY, BAGLEY J, MARASCO WA. Intracellular antibodies as a new class of therapeutic molecules for gene therapy. Human Gene Ther. (1994) 5(5):595–601.
  • MARASCO WA: Intrabodies: turning the humoral immune system outside in for intracellular immuniza-tion. Gene Ther. (1997) 4(0:11–15.
  • ••In this article, the studies from laboratories that have usedintrabodies as molecular reagents for cancer therapy and for the control of infectious diseases are review and future directions of this technology is discussed.
  • DESHANE J, CABRERA G, GRIM JE et al.: Targeted eradica- tion of ovarian cancer mediated by intracellular expression of anti-erbB-2 single-chain antibody. Gynecol. Oncol. (1995) 59(1):8–14.
  • DESHANE J, GRIM J, LOECHEL S et al: Intracellular antibody against erbB-2 mediates targeted tumor cell eradication by apoptosis. Cancer Gene Ther. (1996) 3(2)89–98.
  • DESHANE J, LOECHEL F, CONRY RM et al. Intracellular single-chain antibody directed against erbB2 down-regulates cell surface erbB2 and exhibits a selective anti- proliferative effect in erbB2 overexpressing cancer cell lines. Gene Ther. (1994) 1(5) 332–337
  • DESHANE J, SIEGAL GP, ALVAREZ RD et al.: Targeted tumor killing via an intracellular antibody against erbB-2. j Clin. Invest. (1995) 96(6):2980–2989.
  • •This paper highlights the specific killing of erbB-2-overe-xpressing tumour cells using a recombinant adenovirus encoding an anti-erbB-2 single chain antibody. This strategy of gene therapy for ovarian carcinoma offers the potential to achieve highly specific, targeted killing of human tumour cells and thus establishes the rationale to undertake human clinical trials on this basis.
  • DESHANE J, SIEGAL GP, WANG M et al. Transductional efficacy and safety of an intraperitoneally delivered adenovirus encoding an anti-erbB-2 intracellular single-chain antibody for ovarian cancer gene therapy. Gynecol. Oncol (1997) 64(3)378–385.
  • GRIM J, DESHANE J, FENG M et al.: erbB-2 knockout employing an intracellular single-chain antibody (sFv) accomplishes specific toxicity in erbB-2-expressing lung cancer cells. Am. J. Respir. Cell Mol. (1996) 15(3):348–354.
  • WRIGHT M, GRIM J, DESHANE J et al. An intracellular anti-erbB-2 single-chain antibody is specifically cytotoxic to human breast carcinoma cells overex-pressing erbB-2. Gene Ther. (1997) 4(4):317–322.
  • NEUBAUER A, THIEDE C, HUHN D et al.: P53 and induction of apoptosis as a target for anticancer therapy. Leukemia (1996) 10\(Suppl. 3):S2–S4.
  • XU HJ, ZHOU Y, SEIGNE J et al. Enhanced tumor suppressor gene therapy via replication-deficient adenovirus vectors expressing an N-terminal truncated retinoblastoma protein. Cancer Res. (1996) 56(10) 2245–2249
  • FAVROT M, COLL JL, LOUIS N, NEGOESCU A: Cell death and cancer: replacement of apoptotic genes and inacti-vation of death suppressor genes in therapy. Gene Ther. (1998) 5(6):728–739.
  • ••This review provides a critical evaluation of the increasinguse of gene therapy in the treatment of malignancies to induce active cell death (ACD, apoptosis). This approach is consistent with the notion that cancer is an anomalous accumulation of cells largely resulting from diminished cell death. The review details the main genes potentially useful for therapy. Among these, p53 has received the majority of the investigators' attention and provided encouraging results.
  • NIELSEN LL, MANEVAL DC: p53 tumor suppressor gene therapy for cancer. Cancer Gene Ther. (1998) 5(1) :52–63.
  • •This is a comprehensive review of the various clinical targets suggested for p53 gene therapy is presented together with challenges and prospects for future clinical investigation.
  • EVAN G, LITTLE WOOD T: A matter of life and cell death. Science (1998) 281(5380:1317–1322.
  • •This paper presents the relationship between mutations in somatic cells affecting critical genes that regulate cell prolif-eration and survival causing fatal cancers in multicellular organisms. Growing understanding of these innate mechanisms is suggesting novel therapeutic strategies for cancer.
  • 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 (1995) 14 (2) :153–157.
  • BOUVET M, BOLD RJ, LEE J et al.: Adenovirus-mediated wild-type p53 tumor suppressor gene therapy induces apoptosis and suppresses growth of human pancre-atic cancer. Ann. Surg. Oncol. (1998) 5(8):681–688.
  • PUTZER BM, BRAMSON JL, ADDISON CL et al.: Combina-tion therapy with interleukin-2 and wild-type p53 expressed by adenoviral vectors potentiates tumor regression in a murine model of breast cancer. Human Gene Ther. (1998) 9(5):707–718.
  • PIROLLO KF, HAO Z, RAIT A et al.: p53 mediated sensiti-zation of squamous cell carcinoma of the head and neck to radiotherapy. Oncogene (1 9 9 7) 14(14):1735–1746.
  • NIELSEN LL, GURNANI M, SYED J et al.: Recombinant El-deleted adenovirus-mediated gene therapy for cancer: efficacy studies with p53 tumor suppressor gene and liver histology in tumor xenograft models. Human Gene Ther. (1998) 9(5):681–694.
  • HAMADA K, ALEMANY R, ZHANG WW et al.: Adenovirus-mediated transfer of a wild-type p53 gene and induction of apoptosis in cervical cancer. Cancer Res. (1996) 56(13):3047–3054.
  • FUJIWARA T, CAI DW, GEORGES RN et al.: Therapeutic effect of a retroviral wild-type p53 expression vector in an orthotopic lung cancer model. J. Natl. Cancer Inst. (1994) 86(19):1458–1462.
  • SCHULER M, ROCHLITZ C, HOROWITZ JA et al.: A Phase I study of adenovirus-mediated wild-type p53 gene transfer in patients with advanced non-small cell lung cancer. Human Gene Ther. (1998) 9(14):2075–2082.
  • ••This paper reports a Phase I dose escalation study of a singleintratumoural injection of a replication-defective adenoviral expression vector encoding wild-type p53, carried out in patients with incurable non-small cell lung cancer. Wild-type p53 gene therapy by intratumoural injection of a replication-defective adenoviral expression vector is safe, feasible, and biologically effective in patients with advanced non-small cell lung cancer.
  • ROTH JA, SWISHER SG, MERRITT JA et al.: Gene therapy for non-small cell lung cancer: a preliminary report of a Phase I trial of adenoviral p53 gene replacement. Semin. Oncol (1998) 25(3 Suppl. 8):33–37.
  • ••This paper presents the results of a Phase I study; admini-stration of an adenoviral p53 vector (Adp53) to 21 patients with advanced non-small cell lung cancer produced little toxicity. Up to six intratumoural injections at monthly intervals were well-tolerated. Expression of the p53 transgene was evident, along with potentially useful clinical responses. Time to disease progression in the indicator lesion treated with Adp53 appears to be enhanced by higher doses of vector, concomitant cisplatin therapy, and evidence of apoptosis on tumour biopsy specimens. Phase II trials should now be undertaken to determine the response rate to Adp53.
  • HOLT JT, THOMPSON ME, SZABO C et al.: Growth retardation and tumour inhibition by BRCAL Nature Genet. (1996) 12(3):298–302.
  • ••This paper showed that retroviral transfer of the wild-typeBRCA 1 gene inhibits growth in vitro of all breast and ovarian cancer cell lines tested, but not colon or lung cancer cells or fibroblasts.
  • JENSEN RA, THOMPSON ME, JETTON TL et al.: BRCA1 issecreted and exhibits properties of a granin. Nature Genet. (1996) 12(3):303–308.
  • DEIGNER HP: Modulating apoptosis: current applica-tions and prospects for future drug development. Curr. Med. Chem. (1999) 6(5):399–414.
  • •Agents modulating apoptosis are of extraordinary promise for the treatment of several states of disease including cancer, AIDS, neurodegenerative and ischemic diseases. In this review, a brief summary of cellular pathways relevant to programmed cell death is given and potential therapeutic targets therein are emphasized.
  • ASHKENAZI A, DIXIT VM: Death receptors: signaling and modulation. Science (1998) 281 (5381) :1305–1308.
  • GREEN DR: Apoptotic pathways: the roads to ruin. Cell (1998) 94 (0:695–698.
  • GREEN DR, REED JC: Mitochondria and apoptosis. Science (1998) 281(5380:1309–1312.
  • THORNBERRY NA, LAZEBNIK Y: Caspases: enemies within. Science (1998) 281(5380:1312–1316.
  • SINKOVICS J, HORVATH J. Apoptosis by genetic engineering. Leukemia (1994) 1 (Suppl. 8):S98–102.
  • FAVROT M, COLL JL, LOUIS N et al.: Cell death and cancer: replacement of apoptotic genes and inactiva-tion of death suppressor genes in therapy. Gene Ther. (1998) 5(0:728–739.
  • EALOVEGA MW, MCGINNIS PK, SUMANTRAN VN et al:bcl-xs gene therapy induces apoptosis of human mammary tumors in nude mice. Cancer Res. (1996) 56(9) :1965–1969.
  • CLARKE MF, APEL IJ, BENEDICT MA et al.: A recombinant bcl-xs adenovirus selectively induces apoptosis in cancer cells but not in normal bone marrow cells. Proc. Natl. Acad. ScL USA (1995) 92:11024–11028.
  • ••This paper demonstrates that a recombinant bcl-xsadenovirus specifically and efficiently kills carcinoma cells arising from multiple organs including breast, colon, stomach, and neuroblasts, but not normal hematopoietic progenitor cells. These results suggest that bc1-x, adenovirus vector may prove useful in killing cancer cells contami-nating the bone marrow of patients undergoing autologous bone marrow transplantation.
  • BARGOU RC, WAGENER C, BOMMERT K et al.: Overex- pression of the death-promoting gene bax-alpha which is downregulated in breast cancer restores sensitivity to different apoptotic stimuli and reduces tumor growth in SCID mice. J. Clin. Invest. (1996) 97(10:2651–2659.
  • •In this paper the authors compare the expression pattern of members of the Bc1–2 family in normal and breast cancer tissue samples. Only the bax-alpha, which promotes apoptosis, is expressed in high amounts in normal breast epithelium, whereas only weak or no expresssion could be deteted in cancer samples.
  • SUMANTRAN VN, EALOVEGA MW, NUNEZ G et al: Overexpression of Bel, sensitizes MCF-7 cells to chemotherapy-induced apoptosis. Cancer Res. (1995) 55(12)2507–2510.
  • WELLER M, MALIPIERO U, RENSING-EHL A, BARR PJ, FONTANA A: Fas/apo-1 gene transfer for human malignant glioma. Cancer Res. (1995) 55 (13) 2936–2944.
  • MACKEY TJ, BORKOWSKI A, AMIN P et al.: Bc1-2/bax ratio as a predictive marker for therapeutic response to radiotherapy in patients with prostate cancer. Urology (1998) 52(6):1085–1090.
  • HARIMA Y, HARIMA K, SHIKATA N et al.: Box and Bc1-2 expressions predict response to radiotherapy in human cervical cancer. J. Cancer Res. Clin. Oncol. (1998) 124(0:503–510.
  • SCHUBERT EL, HANSEN MF, STRONG LC: The retinoblas-toma gene and its significance. Ann. Med. (1994) 26 (3) :177–184.
  • TERAO R, HONDA K, HATANO E et al.: Suppression of proliferative cholangitis in a rat model with direct adenovirus-mediated retinoblastoma gene transfer to the biliary tract. Hepatology (1998) 28 (3):605–612.
  • DEMERS GW, HARRIS MP, WEN SF et al.: A recombinant adenoviral vector expressing full-length human retinoblastoma susceptibility gene inhibits human tumor cell growth. Cancer Gene Ther. (1998) 5(0207–214.
  • XU HJ, XU K, ZHOU Y eta].: Enhanced tumor cell growth suppression by an N-terminal truncated retinoblas-toma protein. Proc. NMI Acad. Sci. USA (1994) 91(20:9837–9841.
  • •This paper presents the preclinical studies in the treatment of human non-small cell lung cancer and bladder carcinoma cells with a recombinant adenovirus vector expressing the N-terminal truncated retinoblastoma protein. This vector completely suppressed the tumourigenicity of the treated tumour cells in nude mice.
  • MOOLTEN FL. Drug sensitivity (suicide') genes for selective cancer chemotherapy. Cancer Gene Ther. (1995) 1 (4):279–287.
  • DEONARAIN MP, SPOONER RA, EPENETOS AA: Genetic delivery of enzymes for cancer therapy. Gene Ther. (1995) 2(4):235–244.
  • FREEMAN SM, WHARTENBY KA, FREEMAN JL et al.: In situ use of suicide genes for cancer therapy. Sem. Oncol. (1996) 23(1):31–45.
  • •This paper reviews the emerging gene therapy approach in modificating tumour cells with a suicide gene such as HSV-tk and GCV therapy. Death of tumour cells modified with the HSV-tk gene leads to killing of unmodified in situ tumour cells in a phenomenon termed the 'bystander effect'. The basis both for this effect and other gene therapy trials underway for the treatment of cancer are discussed.
  • RIGG A, SIKORA K: Genetic prodrug activation therapy.Mol. Med. Today (1997) 3(8):359–366.
  • NICULESCU-DUVAZ I, SPOONER R, MARAIS R et al:Gene-directed enzyme prodrug therapy. Bioconjug. Chem. (1998) 9(0:4–22.
  • MARTIN LA, LEMOINE NR: Direct cell killing by suicide genes. Cancer Meta. Rev. (1996) 15(3):301–316.
  • MOOLTEN FL, WELLS JM: Curability of tumors bearing herpes thymidine kinase genes transferred by retroviral vectors. J. Natl. Cancer Inst. (1990) 82(4)297–300.
  • BRAND K, ARNOLD W, BARTELS T et al.: Liver-associated toxicity of the HSV-tk/GCV approach and adenoviral vectors. Cancer Gene Ther. (1997) 4 (1) :9–16.
  • ••This paper demonstrated that the Ad-HSV-tk/GCV approachis toxic for the resting liver, and that additional safety features such as tumour-restricted transgene expression should be included in adenoviral vectors expressing the HSV-tk gene.
  • VAN DER EB MM, CRAMER SJ, VERGOUWE Y et al.: Severehepatic dysfunction after adenovirus-mediated transfer of the herpes simplex virus thymidine kinase gene and ganciclovir administration. Gene Ther. (1998) 5(4):451–458.
  • STERMAN DH, TREAT J, LITZKY LA et al.: Adenovirus-mediated herpes simplex virus thymidine kinase/ganciclovir gene therapy in patients with localized malignancy: results of a Phase I clinical trial in malignant mesothelioma. Human Gene Ther. (1998) 9(7):1083–1092.
  • ••This study demonstrates that intrapleural administration ofan adenoviral vector containing the HSV-tk gene is well tolerated and results in detectable gene transfer when delivered at high doses. Further development of therapeutic trials for treatment of localised malignancy using this vector is thus warranted.
  • ALVAREZ RD, CURIEL DT: A Phase I study of recombi- nant adenovirus vector-mediated intraperitoneal delivery of herpes simplex virus thymidine kinase (HSV-TK) gene and intravenous ganciclovir for previously treated ovarian and extraovarian cancer patients. Human Gene Ther. (1997) 8(5):597–613.
  • ••The results of a Phase I clinical trial in patients with extrao-varian cancer treated with a recombinant adenovirus vector expressing the HSV-tk gene are presented.
  • HIRSCHOWITZ EA, OHWADA A, PASCAL WR eta].:In vivo adenovirus-mediated gene transfer of the Escherichia coli cytosine deaminase gene to human colon carcinoma-derived tumors induces chemosensitivity to 5-fluorocytosine. Human Gene Ther. (1995) 6(8):1055–1063.
  • PEDERSON LC, VICKERS SM, BUCHSBAUM DJ et al. Combined cytosine deaminase expression, 5-fluorocytosine exposure, and radiotherapy increases cytotoxicity to cholangiocarcinoma cells. J. Gastrointest. Surg. (1998) 2(3):283–291.
  • PATTERSON AV, ZHANG H, MOGHADDAM A et al. Increased sensitivity to the prodrug 5'-deoxy-5- -fluorouridineandmodulationof5-fluoro-2'-deoxyuridine sensitivity in MCF-7 cells transfected with thymidine phosphorylase. Br. J. Cancer (1995) 72 (3) 669–675
  • SORSCHER EJ, PENG S, BEBOK Z et al. Tumor cell bystander killing in colonic carcinoma utilizing the Escherichia coli DeoD gene to generate toxic purines. Gene Ther. (1994) 1 (4) 233–238
  • LOCKETT LJ, MOLLOY PL, RUSSELL PJ et al.: Relative efficiency of tumor cell killing in vitro by two enzyme-prodrug systems delivered by identical adenovirus vectors. Clin. Cancer Res. (1997) 3 (11) :2075–2080.
  • DOUGLAS JT, CURIEL DT: Targeted gene therapy. Tumour Targeting (1995) 1:67–84.
  • •This review discusses the major considerations when using gene therapy strategies to target adenovirus vectors.
  • BILBAO G, GOMEZ-NAVARRO J, CONTRERAS JL et al.: Advances in adenoviral vectors for cancer gene therapy. Exp. Opin. Ther. Patents (1997) 7 (12) :1427–1446.
  • •An overview of the major advances in the use of recombi-nant adenovirus vectors for cancer gene therapy.
  • CHU TH, DORNBURG R: Toward highly efficient cell-type-specific gene transfer with retroviral vectors displaying single-chain antibodies. J. Virol. (1997) 71(1)720–725.
  • KASAHARA N, DOZY AM, KAN YW. Tissue-specific targeting of retroviral vectors through ligand-receptor interactions. Science (1994) 266(5189):1373–1376.
  • HUNTER E, SWANSTROM R: Retrovirus envelope glycoproteins. Curr. Top. Microbiol. Immunol. (1990) 157:187–253.
  • ANDERSON WF. Human gene therapy. Science (1992) 256(5058) :808–813.
  • MCCABE, ER: Clinical application of gene therapy: emerging opportunities and current limitations. Biochem. Med. Metab. Biol. (1993) 50(3):241–253.
  • DMITRIEV I, KRASNYKH V, MILLER CR et al. An adenovirus vector with genetically modified fibers demonstrates expanded tropism via utilization of a coxsackievirus and adenovirus receptor-independent cell entry mechanism. J. Virol. (1998) 72(12):9706–9713.
  • KRASNYKH VN, DMITRIEV I, MIKHEEVA G et al.: Charac- terization of an adenoviral vector containing a heterologous peptide epitope in the HI-loop of the fiber knob. J. Virol. (1998) 72(3):1844–1852.
  • •This paper shows the modification of the tropism of the adenovirus via the HI loop of the fibre knob.
  • WICKHAM TJ, SEGAL DM, ROELVINK PW et al.: Targeted adenovirus gene transfer to endothelial and smooth muscle cells by using bispecific antibodies. J. Virol. (1996) 70(10):6831–6838.
  • ••In this paper the results demonstrated that bispecificantibodies can be successfully used to target adenovirus vector to a specific cellular receptor and thereby increase the efficiency of gene transfer.
  • BERGELSON JM, CUNNINGHAM JA, DROGUETT G et al.:Isolation of a common receptor for coxsackie B viruses and adenoviruses 2 and 5. Science (1997) 275:1320–1323.
  • TOMKO RP, XU R, PHILIPSON L: HCAR and MCAR: thehuman and mouse cellular receptors for subgroup C adenoviruses and group B coxsackieviruses. Proc. Natl. Acad. ScL USA (1997) 94(7):3352–3356.
  • HENRY LJ, XIA D, WILKE ME et al: Characterization ofthe knob domain of the adenovirus Type 5 fiber protein expressed in Escherichia coif J. Virol. (1994) 68(8)5239–5246.
  • LOUIS N, FENDER P, BARGE A etal: Cell-binding domain of adenovirus serotype 2 fiber. J. Virol. (1994) 68(6):4104–4106.
  • YOSHIDA Y, SADATA A, ZHANG W et al.: Generation of fiber-mutant recombinant adenoviruses for gene therapy of malignant glioma. Human Gene Ther. (1998) 9:2503–2515.
  • MICHAEL SI, HONG JS, CURIEL DT et al. Addition of a short peptide ligand to the adenovirus fiber protein. Gene Ther. (1995) 2(9):660–668.
  • WICKHAM TJ, ROELVINK PW, BROUGH DE et al: Adenovirus targeted to heparan-containing receptors increases its gene delivery efficiency to multiple cell types. Nature Biotech. (1996) 14(11):1570–1573.
  • WICKHAM TJ, TZENG E, SHEARS LL et al: Increased in vitro and in vivo gene transfer by adenovirus vectors containing chimeric fiber proteins. J. Vim]. (1997) 71(10:8221–8229.
  • DOUGLAS JT, ROGERS BE, ROSENFELD ME et al: Targeted gene delivery by tropism-modified adenoviral vectors. Nature Biotech. (1996) 14:1574–1578.
  • GOLDMAN CK, ROGERS BE, DOUGLAS JT et al.: Targeted gene delivery to Kaposi's sarcoma cells via the fibroblast growth factor receptor. Cancer Res. (1997) 57(8):1447–1451.
  • MILLER CR, BUCHSBAUM DJ, REYNOLDS PN et al.: Differ-ential susceptibility of primary and established human glioma cells to adenovirus infection: Targeting via the epidermal growth factor receptor achieves fiber receptor-independent gene transfer. Cancer Res. (1998) 58:5738–5748.
  • RANCOURT C, ROGERS BE, SOSNOWSKI BA et al.: Basic fibroblast growth factor enhancement of adenovirus-mediated delivery of the herpes simplex virus thymidine kinase gene results in augmented therapeutic benefit in a murine model of ovarian cancer. Clin. Cancer Res. (1998) 4:2455–2461.
  • REYNOLDS PN, MILLER CR, GOLDMAN CK et al. Targeting adenoviral infection with basic fibroblast growth factor enhances gene delivery to vascular endothelial and smooth muscle cells. Tumour Targeting (1998) 3(3):156–168.
  • ROGERS BE, DOUGLAS JT, AHLEM C et al.: Use of a novel cross-linking method to modify adenoviral tropism. Gene Therapy (1997) 4(12):1387–1392.
  • ROGERS BE, DOUGLAS JT, SOSNOWSKI BA et al.: Enhanced in vivo gene delivery to human ovarian cancer xenografts utilizing a tropsim-modified adenovirus vector. Tumour Targeting (1998) 3:25–31.
  • GOLDMAN CK, ROGERS BE, DOUGLAS JT et al: Targeted gene delivery to Kaposi's sarcoma cells via the fibroblast growth factor receptor. Cancer Res. (1997) 57 (8):1447–1451.
  • MILLER CR, BUCHSBAUM DJ, REYNOLDS PN et al. Differ- ential susceptibility of primary and established human glioma cells to adenovirus infection: targeting via the epidermal growth factor receptor achieves fiber receptor-independent gene transfer. Cancer Res. (1998) 58(24):5738–5748.
  • RANCOURT C, ROGERS BE, SOSNOWSKI BA et al. Basic fibroblast growth factor enhancement of adenovirus-mediated delivery of the herpes simplex virus thymidine kinase gene results in augmented therapeutic benefit in a murine model of ovarian cancer. Chn. Cancer Res. (1998) 4(10):2455–2461.
  • ROGERS BE, DOUGLAS JT, AHLEM C et al.: Use of a novel cross-linking method to modify adenovirus tropism. Gene Ther. (1997) 4(12):1387–1392.
  • WICKHAM TJ, SEGAL DM, ROELVINK PW et al.: Targeted adenovirus gene transfer to endothelial and smooth muscle cells by using bispecific antibodies. J. Virol. (1997) 71(10):6831–6838.
  • WICKHAM TJ, LEE GM, TITUS JA et al.: Targeted adenovirus-mediated gene delivery to T cells via CD3. Virol. (1997) 71 (10):7663–7669.
  • HARRIS JD, GUTIERREZ AA, HURST HC, SIKORA K, LEMOINE NR: Gene therapy for cancer using tumor-specific prodrug activation. Gene Ther. (1994) 1(3):170–175.
  • MANOME Y, ABE M, HAGEN MF et al. Enhancer sequences of the DF3 gene regulate expression of the herpes simplex virus thymidine kinase gene and confer sensitivity of human breast cancer cells to ganciclovir. Cancer Res. (1994) 54(20)5408–5413.
  • CHEN L, CHEN D, MANOME Y et al.: Breast cancer selective gene expression and therapy mediated by recombinant adenoviruses containing the DF3/MUC1 promoter. J. Clin. Invest. (1995) 96(6):2775–2782.
  • KANAI F, LAN KH, SHIRATORI Y et al.: In vivo gene therapy for alpha-fetoprotein-producing hepatocel-lular carcinoma by adenovirus-mediated transfer of cytosine deaminase gene. Cancer Res. (1997) 57 (3):461–465.
  • 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 Ther. (1995) 2(3) 191–197
  • SHIRATORI KF, YOSHIDA Y, WAKIMOTO H, HAMADA H, et al Gene therapy for alpha-fetoprotein-producing human hepatoma cells by adenovirus-mediated transfer of the herpes simplexvirus thymidine kinase gene. Hepatology (1996) 23(6):1359–68.
  • BRAND K, LOSER P, ARNOLD W et al. Tumor cell-specific transgene expression prevents liver toxicity of the adeno-HSVtk/GCV approach. Gene Ther. (1998) 5(10):1363–1371.
  • SHIRAKAWA T, KO SC, GARDNER TA et al.: In vivo suppression of osteosarcoma pulmonary metastasis with intravenous osteocalcin promoter-based toxic gene therapy. Cancer Gene Ther. (1998) 5(5):274–280.
  • BROWN JM, GIACCIA AJ: The unique physiology of solid tumors: opportunities (and problems) for cancer therapy. Cancer Res. (1998) 58:1408–1416.
  • WALTON T, WANG JL, RIBAS A et al.: Endothelium-specific expression of an E-selectin promoter recombinant adenoviral vector. Anticancer Res. (1998) 18(3A):1357–1360.
  • LEMMON ML, VAN ZIJL P, FOX ME et al: Anaerobic bacteria as a gene delivery system that is controlled by the tumour microenvironment. Gene Ther. (1997) 4:791–796.
  • YOSHIDA Y, HAMADA H: Adenovirus-mediated inducible gene expression through tetracycline-controllable transactivator with nuclear localization signal. Biochem. Biophys. Res. Commun. (1997) 230(2)426–430.
  • MASSIE B, COUTURE F, LAMOUREUX L et al: Inducible overexpression of a toxic protein by an adenovirus vector with a tetracycline-regulatable expression cassette. J. Vim]. (1998) 72(3):2289–2296.
  • DELORT JP, CAPECCHI MR: TAXI/UAS: a molecular switch to control expression of genes in vivo. Human Gene Ther. (1996) 7(7):809–820.
  • WANG Y, O'MALLEY BWJ, TSAI SY et al: A regulatory system for use in gene transfer. Proc. Nati Acad. ScL USA (1994) 91(17):8180–8184.
  • MANOME Y, KUNIEDA T, WEN PY et al: Transgene expression in malignant glioma using a replication-defective adenoviral vector containing the Egr-1 promoter: activation by ionizing radiation or uptake of radioactive iododeoxyuridine. Human Gene Ther. (1998) 9(10):1409–1417.
  • CULVER KW, RAM Z, WALLBRIDGE S et al.: In vivo gene transfer with retroviral vector-producer cells for treatment of experimental brain tumors. Science (1992) 256(5063)1550–1552.
  • FREEMAN SM, ABBOUD CN, WHARTENBY KA et al: The bystander effect: tumor regression when a fraction of the tumor mass is genetically modified. Cancer Res. (1993) 53:5274–5274.
  • BI WL, PARYSEK LM, WARNICK R, STAMBROOK PJ: In vitro evidence that metabolic cooperation is respon-sible for the bystander effect observed with HSV-tk retroviral gene therapy. Human Gene Ther. (1993) 4 (6):725–731.
  • TOURAINE RL, VAHANIAN N, RAMSEY WJ et al.: Enhance-ment of the herpes simplex virus thymidine kinase/ganciclovir bystander effect and its antitumor efficacy in vivo by pharmacologic manipulation of gap junctions. Human Gene Ther. (1998) 9(16):2385–2391.
  • GAGANDEEP S, BREW R, GREEN B et al: Prodrug-activated gene therapy: involvement of an immuno-logical component in the 'bystander effect'. Cancer Gene Ther. (1996) 3(2):83–88.
  • COLOMBO BM, BENEDETTI S, OTTOLENGHI S et al: The 'bystander effect': association of U-87 cell death with ganciclovir-mediated apoptosis of nearby cells and lack of effect in athymic mice. Human Gene Ther. (1995) 6:763–772.
  • SMYTHE WR, HWANG HC, AMIN KM et al.: Use of recombinant adenovirus to transfer the herpes simplex virus thymidine kinase (HSVtk) gene to thoracic neoplasms: an effective in vitro drug sensiti-zation system. Cancer Res. (1994) 54 (8) :2055–2059.
  • 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. Am. J. Respir. Cell Mol. Biol. (1995) 13(1):7–16.
  • BONNEKOH B, GREENHALGH DA, BUNDMAN DS et al: Inhibition of melanoma growth by adenoviral-mediated hsv thymidine kinase gene transfer in vivo. J. Invest. Dermatol. (1995) 104(3): 313–317.
  • 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 Res. (1995) 55 (5):1080–1085.
  • O'MALLEY BW, COPE KA, CHEN SH et al.: Combination gene therapy for oral cancer in a murine model. Cancer Res. (1996) 56(8):1737–1741.
  • SACCO MG, MANGIARINI L, VILLA A et al.: Local regres-sion of breast tumors following intramammary ganciclovir administration in double transgenic mice expressing neu oncogene and herpes simplex virus thymidine kinase. Gene Ther. (1995) 2(7):493–497.
  • RAM Z, WALBRIDGE S, SHAWKER T, CULVER KW, BLAESE RM, OLDFIELD EH: The effect of thymidine kinase transduction and ganciclovir therapy on tumor vasculature and growth of 91 gliomas in rats. J. Neurosurg. (1994) 81(2):256–260.
  • IZQUIERDO M, CORTES M, DE FELIPE P et al.: Long-term rat survival after malignant brain tumor regression by retroviral gene therapy. Gene Ther. (1995) 2(1):66–69.
  • IZQUIERDO M, MARTIN V, DE FELIPE P et al. Human malignant brain tumor response to herpes simplex thymidine kinase (HSVtk)/ganciclovir gene therapy. Gene Ther. (1996) 3(6):491–495.
  • 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 Ther. (1996) 7(3):401–416.
  • RAFFERTY JA, HICKSON I, CHINNASAMY N et al: Chemoprotection of normal tissues by transfer of drug resistance genes. Cancer Meta. Rev. (1996) 15:365–383.
  • SORRENTINO BP, BRANDT SJ, BODINE D et al.: Selection of drug-resistant bone marrow cells in vivo after retroviral transfer of human MDR1. Science (1992) 257(5066)99–103.
  • KOC ON, ALLAY JA, LEE K et al.: Transfer of drug resistance genes into hematopoietic progenitors to improve chemotherapy tolerance. Sem. Oncol. (1996) 23(1)46–65.
  • ALLAY JA, KOC ON, DAVIS BM etal.: Retroviral-mediated gene transduction of human alkyltransferase comple-mentary DNA confers nitrosourea resistance to human hematopoietic progenitors. Clin. Cancer Res. (1996) 2(8):1353–1359.
  • DOROSHOW JH, METZ MZ, MATSUMOTO L et al.: Transduction of NIH 3T3 cells with a retrovirus carrying both human MDR1 and glutathione S-transferase pi produces broad-range multidrug resistance. Cancer Res. (1995) 55 (18) 4073–4078.
  • BIENZLE D, ABRAMS-OGG AC, KRUTH SA et al: Gene transfer into hematopoietic stem cells: long-term maintenance of in vitro activated progenitors without marrow ablation. Proc. Natl. Acad. ScL USA (1994) 91(0:350–354.
  • DORIGO O, TURLA ST, LEBEDEVA S et al.: Sensitization of rat glioblastoma multiforme to cisplatin in vivo following restoration of wild-type p53 function. J. Neurosurg. (1998) 88(3):535–540.
  • BARNES MN, DESHANE JS, SIEGAL GP et al.: Novel gene therapy strategy to accomplish growth factor modula-tion induces enhanced tumor cell chemosensitivity. Clin. Cancer Res. (1996) 2(7):1089–1095.
  • PICHE A, GRIM J, RANCOURT C et al: Modulation of Bc1-2 protein levels by an intracellular anti-Bc1-2 single-chain antibody increases drug-induced cytotoxicity in the breast cancer cell line MCF-7. Cancer Res. (1998) 58(102134–2140.
  • PEDERSON LC, BUCHSBAUM DJ, VICKERS SM et al: Molecular chemotherapy combined with radiation therapy enhances killing of cholangiocarcinoma cells in vitro and in vivo. Cancer Res. (1997) 57(19)4325–4332.
  • ROSENBERG SA. Principals of cancer management: biologic therapy. In: Principal and Practice of Oncology (Edition 5). DeVita, Helman S, Rosenberg SA (Eds.), Lippincott-Raven, Philadelphia, USA (1997)349–373.
  • BURNETT FM: The concept of immunological surveil-lance. Prog. Exp. Tumour Res. (1970) 13:1–27.
  • PARDOLL DM: Cancer vaccines. Nature Med. (1998) 4(5 Suppl.):525–531.
  • OCHSENBEIN AF, KLENERMAN P, KARRER U et al. Immune surveillance against a solid tumor fails because of immunological ignorance. Proc. Natl. Acad. ScL USA (1999) 96(5):2233–2238.
  • WOJTOWICZ-PRAGA S: Reversal of tumor-induced immunosuppression: a new approach to cancer therapy. J. Immunother. (1997) 20(3):165–177.
  • STAVELEY-O'CARROLL K, SOTOMAYOR E, MONTGOMERY J et al.: Induction of antigen-specific T cell anergy: An early event in the course of tumor progression. Proc. Natl. Acad. Sci. USA (1998) 95 (3):1178–1183.
  • ROSENBERG SA, AEBERSOLD P, CORNETTA K et al: Gene transfer into humans-immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduc-tion. New Engl. J. Med. (1990) 323(9):570–578.
  • HWU P, ROSENBERG SA: The genetic modification of TIL for cancer therapy: preclinical and clinical studies. Gene Therapy for Neoplastic Diseases. Washington, DC, USA (1993).
  • ESHHAR Z: Tumor-specific T-bodies: towards clinical application. Cancer Immunol. Immunother. (1997) 45(3-4):131–136.
  • MASTRANGELO MJ, SATO T, LATTIME EC et al.: Cellular vaccine therapies for cancer. Cancer Treat. Res. (1998) 94:35–50.
  • HELLSTROM I, HELLSTROM KE: Tumor vaccines - a reality at last? J. Immunother. (1998) 21(2):119–126.
  • BHATTACHARYA-CHATTERJEE M, FOON KA: Anti-idiotype antibody vaccine therapies of cancer. Cancer Treat. Res. (1998) 94:51–58.
  • HERSH EM, STOPECK AT: Advanced in the biological therapy and gene therapy of malignant disease. Clin. Cancer Res. (1997) 3(12 Pt 2):2623–2629.
  • MOREL A, DE LA COSTE A, FERNANDEZ N et al.: Does preventive vaccination with engineered tumor cells work in cancer- prone transgenic mice? Cancer Gene Ther. (1998) 5(2):92–100.
  • CONRY RM, LOBUGLIO AF, CURIEL DT: Polynucleotide-mediated immunization therapy of cancer. Semin. Oncol. (1996) 23(0:135–147.
  • CONRY RM, WHITE SA, FULTZ PN et al.: Polynucleotide immunization of nonhuman primates against carcinoembryonic antigen. Clin. Cancer Res. (1998) 4(10:2903–2912.
  • WOLFF JA, LUDTKE JJ, ACSADI G et al.: Long-term expression of plasmid DNA and foreign gene expres-sion in mouse muscle. Human Mol. Genet. (1992) 1:363–369.
  • MCDONNELL WM, ASKARI FK: DNA vaccines. New Engl. J. Med. (1996) 334(1):42–45.
  • CONRY RM, WIDERA G, LOBUGLIO AF et al.: Selected strategies to augment polynucleotide immunization. Gene Ther. (1996) 3(0:67–74
  • UN KY, GUARNIERI FG, STAVELEY-O'CARROLL KF et al.: Treatment of established tumors with a novel vaccine that enhances major histocompatibility class II presentation of tumor antigen. Cancer Res. (1996) 56(0:21–26.
  • CONRY RM, LOBUGLIO AF, LOECHEL F et al.: A carcinoembryonic antigen polynucleotide vaccine for human clinical use. Cancer Gene Ther. (1995) 2(1)33–38.
  • HENGGE UR, PFUTZNER W, WILLIAMS M, GOOS M, VOGEL JC: Efficient expression of naked plasmid DNA in mucosal epithelium: prospective for the treatment of skin lesions. J. Invest. Dermatol. (1998) 111(4)605–608.
  • JOHANNING FW, CONRY RM, LOBUGLIO AF et al.: A Sindbis virus mRNA polynucleotide vector achieves prolonged and high level heterologous gene expres-sion in vivo. Nucleic Acids Res. (1995) 23 (9) :1495–1501.
  • MARTINON F, KRISHNAN S, LENZEN G et al.: Induction of virus-specific cytotoxic T lymphocytes in vivo by liposome- entrapped mRNA. Eur. J. Immunol. (1993) 23(7):1719–1722.
  • IIJIMA Y, OHNO K, IKEDA H et al: Cell-specific targeting of a thymidine kinase/ganciclovir gene therapy system using a recombinant Sindbis virus vector. Int. J. Cancer (1999) 80(0:110–118.
  • HERWEIJER H, LATENDRESSE JS, WILLIAMS P et al.: A plasmid-based self-amplifying Sindbis virus vector. Human Gene Ther. (1995) 6 (9) :1161–1167.
  • HAHN CS, HAHN YS, BRACIALE TJ et al.: Infectious Sindbis virus transient expression vectors for studying antigen processing and presentation. Proc. Natl. Acad. Sci. USA (1992) 89(7):2679–2683.
  • RESTIFO NP: Cancer therapy using a self-replicative RNA vaccine. FDA-NCI Workshop on Tumour Vaccines. Bethesda, MD, USA (1998).
  • BEELMAN CA, PARKER R: Degradation of mRNA in eukaryotes. Cell (1995) 81 (2) :179–183.
  • STRONG TV, HAMPTON TA, LOURO I et al.: Incorpora-tion of beta-globin untranslated regions into a Sindbis virus vector for augmentation of heterologous mRNA expression. Gene Ther. (1997) 4(6):624–627.
  • HEIDENREICH O, PIEKEN W, ECKSTEIN F: Chemically modified RNA: approaches and applications. FASEBJ. (1993) 7(0:90–96.
  • HART DN: Dendritic cells: unique leukocyte popula-tions which control the primary immune response. Blood (1997) 90(9):3245–3287.
  • HSU FJ, BENIKE C, FAGNONI F et al.: Vaccination of patients with B-cell lymphoma using autologous antigen- pulsed dendritic cells. Nature Med. (1996) 2(1):52–58.
  • MURPHY G, TJOA B, RAGDE H et al.: Phase I clinical trial: T-cell therapy for prostate cancer using autologous dendritic cells pulsed with HLA-A0201-specific peptides from prostate-specific membrane antigen. Prostate (1996) 29(6):371–380.
  • BAKKER AB, MARLAND G, DE BA et al. Generation of antimelanoma cytotoxic T lymphocytes from healthy donors after presentation of melanoma-associated antigen-derived epitopes by dendritic cells in vitro. Cancer Res. (1995) 55(22) 5330–5334
  • PORGADOR A, GILBOA E: Bone marrow-generated dendritic cells pulsed with a class I-restricted peptide are potent inducers of cytotoxic T lymphocytes. J. Exp. Med. (1995) 182(1):255–260.
  • NESTLE FO, ALIJAGIC S, GILLIET M et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nature Med. (1998) 4(3):328–332.
  • GILBOA E, NAIR SK, LYERLY HK: Immunotherapy of cancer with dendritic-cell-based vaccines. Cancer Immunol. Immunother. (1998) 46(2):82–87.
  • NAIR SK, BOCZKOWSKI D, MORSE M et al. Induction of primary carcinoembryonic antigen (CEA) -specific cytotoxic T lymphocytes in vitro using human dendritic cells transfected with RNA. Nature Biotechnol (1998) 16(4)364–369.
  • DION LD, GOLDSMITH KT, STRONG TV et al.: ElA RNA transcripts amplify adenovirus-mediated tumor reduction. Gene Ther. (1996) 3(11):1021–1025.
  • HART I, COLACO C: Immunotherapy. Fusion induces tumour rejection. Nature (1997) 388(6643):626–627.
  • GONG J, CHEN D, KASHIWABA M et al. Induction of antitumor activity by immunization with fusions of dendritic and carcinoma cells. Nature Med. (1997) 3(5) 558–561
  • ROWSE GJ, TEMPERO RM, VANLITH ML et al. Tolerance and immunity to MUC1 in a human MUC1 transgenic murine model. Cancer Res. (1998) 58(2):315–321.
  • GONG J, CHEN D, KASHIWABA M et al.: Reversal of tolerance to human MUC1 antigen in MUC1 transgenic mice immunized with fusions of dendritic and carcinoma cells. Proc. Natl. Acad. Sci. USA (1998) 95(10:6279–6283.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.