Summary
In the last 5 years significant progress has been made in our understanding of the molecular nature of anti-tumour T cell-mediated responses.
This review describes the involvement of the cellular immune system in the recognition and destruction of cancer cells. Four aspects are discussed: (i) the generalised immune activation induced by the systemic administration of cytokines, in particular, interleukin-2; (ii) the specific T cell-mediated reactions against tumour cells through the recognition of tumour-associated molecules, including the MAGE antigens, MART 1 (melanoma antigen recognised by T cells 1) and tyrosinase proteins described in melanomas, and minor histocompatibility antigens in the setting of allogenic bone marrow transplantation for leukaemia; (iii) the potentially significant but still hypothetical immune-mediated recognition of molecules either tumour-associated or transformation-related (including altered oncogenic proteins); and (iv) the role of co-stimulatory molecules in the induction of tumour-specific immunity.
The current and future therapeutic applications in cancer treatment and potential limitations to this approach are discussed.
Similar content being viewed by others
References
Montano X. Molecular therapeutic approaches. In: Neel B, Kumar R, editors. The molecular basis of human cancer. Mount Kisco, NY: Futura Publishing Co Inc., 1993: 449–75
Townsend ARM, Gotch FM, Davey J. Cytotoxic T cells recognize fragments of the influenza virus nucleoprotein. Cell 1985; 42: 457–67
Townsend ARM, Bastin J, Gould K, et al. Cytotoxic T lymphocytes recognize influenza hemagglutinin that lack a signal sequence. Nature 1986; 324: 575–7
Harding CV, Unanue ER. Quantitation of antigen-presenting cell MHC class II/peptide complexes necessary for T cell stimulation. Nature 1990; 346: 574–6
Gambacorti-Passerini C, Sondel PM. Problems, controversies, and perspectives on the clinical use of interleukin-2. Curr Opin Oncol 1990; 2: 1139–45
Gambacorti-Passerini C, Radrizzani M, Marolda R, et al. In vivo activation of lymphocytes in melanoma patients receiving escalating doses of recombinant interleukin 2. Int J Cancer 1988; 41: 700–6
Shiloni E, Lafreniere R, Mule JJ, et al. Effect of immunotherapy with allogenic lymphokine activated killer (LAK) cells and recombinant interleukin-2 (RIL-2) on established pulmonary and hepatic metastases in mice. Cancer Res 1986; 46: 5633–40
Rosenberg SA. The immunotherapy and gene therapy of cancer. J Clin Oncol 1992; 10: 180–99
Rosenberg SA, Lotze MT, Yang JC, et al. Experience with the use of high-dose interleukin-2 in the treatment of 652 cancer patients. Ann Surg 1989; 210: 474–85
Rosenstein M, Ettinghausen SE, Rosenberg SA. Extravasation of intravascular fluid mediated by the systemic administration of recombinant interleukin-2. J Immunol 1986; 137: 1735–42
Lotze MT, Matory YL, Rayner AA, et al. Clinical effects and toxicity of interleukin-2 in patients with cancer. Cancer 1986; 58: 2764–72
Ettinghausen SE, Moore JG, White DE, et al. Hematologic effects of immunotherapy with lymphokine activated killer cells and recombinant interleukin-2 in cancer patients. Blood 1987; 69: 1654–60
Vetto JT, Papa MZ, Lotze MT, et al. Reduction of toxicity of interleukin-2 and lymphokine activated killer cells in humans by the administration of corticosteroids. J Clin Oncol 1987; 5: 496–503
Belldegrun A, Webb DE, Austin HA, et al. Effect of interleukin-2 on renal function in patients receiving immunotherapy for advanced cancer. Ann Intern Med 1987; 106: 817–22
Denicoff KD, Rubinow DR, Papa MZ, et al. The neuropsychi-atric effects of the treatment with interleukin-2 and lympho-kine activated killer cells. Ann Intern Med 1987; 107: 293–300
Cotran RS, Pober JS, Gimbrone Jr MS, et al. Endothelial activation during interleukin-2 (IL-2) immunotherapy: a possible mechanism for the vascular leak syndrome. J Immunol 1988; 140: 1883–8
Gaspari AA, Lotze MT, Rosenberg SA, et al. Dermatologic changes associated with interleukin-2 administration. JAMA 1987; 258: 1624–9
Saris SC, Rosenberg SA, Friedman RB, et al. Penetration of recombinant interleukin-2 across the blood cerebrospinal fluid barrier. J Neurosurg 1988; 69: 29–34
Ognibene FP, Rosenberg SA, Lotze MT, et al. Interleukin-2 administration causes reversible hemodynamic changes and left ventricular dysfunction similar to those seen in septic shock. Chest 1988; 95: 750–4
Webb DE, Austin HA, Belldegrun A, et al. Metabolic and renal effects of interleukin-2 immunotherapy for metastatic cancer. Clin Nephrol 1988; 30: 141–5
Schwartzentruber D, Lotze MT, Rosenberg SA, et al. Clonic perforation: an unusual complication of therapy with high-dose interleukin-2. Cancer 1988; 62: 2350–3
Lee RE, Gaspari AA, Lotze MT, et al. Interleukin-2 and psoriasis. Arch Dermatol 1988; 124: 1811–5
Lee RE, Lotze MT, Skibber JM, et al. Cardiorespiratory effects of immunotherapy with interleukin-2. J Clin Oncol 1989; 7: 7–20
Belldegrun A, Webb DE, Austin HA, et al. Renal toxicity of interleukin-2 administration in patients with metastatic renal cell cancers. J Urol 1989; 141: 499–503
Donicoff KD, Durkin TM, Lotze MT, et al. The neuroendocrine effects of interleukin-2 treatment. J Clin Endocrinol Metab 1989; 69: 402–10
Kragel AH, Travis WD, Steis RG, et al. Myocarditis or acute myocardial infarction associated with interleukin therapy for cancer. Cancer 1990; 66: 1513–6
Bock SN, Lee RE, Fisher B, et al. A prospective randomized trial evaluating prophylactic antibodies to prevent triple-lumen catheter-related sepsis in patients treated with immunotherapy. J Clin Oncol 1990; 8: 161–9
Atkins MB, Mier JW, Parkinson DR, et al. Hypothyroidism after treatment with interleukin-2 and lymphokine-activated killer cells. N Engl J Med 1988; 318: 1557–63
Schwartzentruber DJ, White DE, Zweig MH, et al. Thyroid dysfunction associated with immunotherapy for patients with cancer. Cancer 1991; 68: 2384–90
Gambacorti-Passerini C, Supino R, Marinoni A, et al. Human lymphoma clones with stable resistance to lymphokine activated killer cells show decreased LFA-3 and ICAM-1 expression and resistance to TNF-α. J Immunol Res 1991; 3: 102–10
Sondel PM, Hank JA, Kohler PC, et al. Destruction of autolo-gous human lymphocytes by interleukin-2-activated cyto-toxic cells. J Immunol 1986; 137: 502–11
Gelmo BT, Palladino MA, Jaffe HS, et al. Circulating cytokines in patients with metastatic cancer treated with recombinant interleukin and lymphokine-activated killer cells. Cancer 1988; 48: 5864–7
Rubin JT, Elwood LJ, Rosenberg SA, et al. Immunohistochemical correlates of response to recombinant interleukin-2-based immunotherapy in man. Cancer Res 1989; 49: 7086–92
Blay JY, Favrot MC, Negrier S, et al. Correlation between clinical response to interleukin 2 therapy and sustained production of tumor necrosis factor. Cancer Res 1990; 50: 2371–4
Ellehorn JD, Hirsch R, Schreiber H, et al. In vivo administration of anti-CD3 prevents malignant progressor tumor growth. Science 1988; 242: 569–71
Hank J, Albertini M, Wesly O, et al. Clinical and immunological effects of treatment with murine anti-CD3 monoclonal antibody together with interleukin-2 in patients with cancer. Clin Cancer Res 1995; 1: 481–92
Hank JA, Robinson RR, Surfur J, et al. Augmentation of antibody dependent cell mediated cytotoxicity following in vivo therapy with recombinant interleukin 2. Cancer Res 1990; 50: 5234–9
Liao SK, Horton L, Flahart L, et al. ADCC of a mouse-human chimeric monoclonal antibody of the human IgG1 subclass with specificity for human adenocarcinomas [abstract]. Proc Am Assoc Cancer Res 1989; 30: 391
Nitta T, Sato K, Yagita H, et al. Preliminary trial of specific targeting therapy against malignant glioma. Lancet 1990; 1: 368–71
Lamers CH, Gratama JW, Warnaar SO, et al. Inhibition of bi-specific antibody (bsAb)-targeted cytolysis by human anti-mouse antibodies in ovarian carcinoma patients treated with bsAb-targeted activated T-lymphocytes. Int J Cancer 1995; 60: 450–7
Ball ED, Guyre PM, Mills L, et al. Initial trial of bispecific antibody-mediated immunotherapy of CD 15-bearing tumors: cytotoxicity of human tumor cells using a bispecific antibody comprised of anti-CD 15 (MoAb PM81) and anti-CD64/Fc gamma RI (MoAb 32). J Hematother 1992; 1: 85–94
Kroesen BJ, Buter J, Sleijfer DT, et al. Phase I study of intravenously applied bispecific antibody in renal cell cancer patients receiving subcutaneous interleukin 2. Br J Cancer 1994; 70: 652–61
Kroesen BJ, ter Haar A, Spakman H, et al. Local antitumor treatment in carcinoma patients with bispecific-monoclonal-antibody-redirected T cells. Cancer Immunol Immunother 1993; 37: 400–7
Shen GL, Li J-L, Vitetta ES. Bispecific anti-CD22/anti-CD3-ricin A chain immunotoxin is cytotoxic to Daudi lymphoma cells but not T cell in vitro and shows both A-chain-mediated and LAK-T-mediated killing. J Immunol 1994; 152: 2368–76
Fanger MW, Morganelli PM, Guyre PM. Use of bispecific antibodies in the therapy of tumors. Cancer Treat Res 1993; 68: 181–94
Rosenberg SA, Spiess PJ, Lafreniere R. A new approach to the adoptive immunotherapy of cancer with tumor infiltrating lymphocytes. Science 1986; 233: 1318–21
Muul LM, Spiess PJ, Director EP, et al. Identification of specific cytolytic immune responses against autologous tumors in humans bearing malignant melanoma. J Immunol 1987; 138: 989–95
Itoh K, Tilden AB, Balch CM. Interleukin-2 activation of cytotoxic T-lymphocytes infiltrating into human metastatic melanomas. Cancer Res 1986; 46: 3011–7
Topalian SL, Solomon D, Rosenberg SA. Tumor-specific cytolysis by lymphocytes infiltrating human melanomas. J Immunol 1989; 142: 3714–25
Barth Jr RJ, Bock SN, Mule JJ, et al. Unique murine tumor associated antigens identified by tumor infiltrating lymphocytes. J Immunol 1990; 144: 1531–7
Barth Jr RJ, Mule JJ, Spiess PJ, et al. Interferon gamma and tumor necrosis factor have a role in tumor regressions mediated by murine CD8+ tumor infiltrating lymphocytes. J Exp Med 1991; 173: 647–58
Barth Jr RJ, Mule JJ, Asher AL, et al. Identification of unique murine tumor associated antigens by tumor infiltrating lymphocytes using tumor specific secretion of interferon-gamma and tumor necrosis factor. J Immunol Methods 1991; 140: 269–79
Schwartzentruber DJ, Topalian SL, Mancini MJ, et al. Specific release of granulocyte-macrophage colony-stimulating factor, tumor necrosis factor-α, and IFN-γ by human tumor-infiltrating lymphocytes after autologous tumor stimulation. J Immunol 1991; 146: 153–64
Kawakami Y, Zakut R, Topalian SL, et al. Shared human melanoma antigens. Recognition by tumor-infiltrating lymphocytes in HLA-2.1-transfected melanomas. J Immunol 1992; 148: 638–43
Rosenberg SA, Packard BS, Aebersold PM, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunother-apy of patients with metastatic melanoma, special report. N Engl J Med 1988; 319: 1676–80
Tepper RI, Pattengale PK, Leder P. Murine interleukin-4 displays potent anti-tumor activity in vivo. Cell 1989; 57: 503–12
Hock H, Dorsch M, Kunzendorf U, et al. Vaccinations with tumor cells genetically engineered to produce different cyto-kines: affectivity not superior to a classical adjuvant. Cancer Res 1993; 53: 714–6
Real FX, Mattes AN, Houghton HF, et al. Class I (unique) tumor antigens of human melanoma. Identification of a 90,000 dal-ton cell surface glycoprotein by autologous antibody. J Exp Med 1984; 160: 1219–33
Lloyd KO. Humoral immune responses to tumor-associated carbohydrate antigens. Semin Cancer Biol 1991; 2: 421–31
Vijayasaradhi S, Bouchard B, Houghton AN. The melanoma antigen gp75 is the human homologue of the mouse b (brown) locus gene product. J Exp Med 1990; 171: 1375–80
Schlichtholz B, Legros Y, Gillet D, et al. The immune response to p 53 in breast cancer patients is directed against immuno-dominant epitopes unrelated to the mutational hot spot. Cancer Res 1992; 52: 6380–4
Lubin R, Schlichtholz B, Bengoufa D, et al. Analysis of p53 antibodies in patients with various cancers define B-cell epitopes of human p53: distribution on primary structure and exposure on protein surface. Cancer Res 1993; 53: 5872–6
Schlichtholz B, Tredaniel J, Lubin R, et al. Analyses of p53 antibodies in sera of patients with lung carcinoma define im-munodominant regions in the p53 protein. Br J Cancer 1994; 69: 809–16
Disis ML, Calenoff E, McLaughlin G, et al. Existent T-cell and antibody immunity to HER2/neu protein in patients with breast cancer. Cancer Res 1994; 54: 16–20
Nijman HW, Van der Burg SH, Vierboom MP, et al. P53, a potential target for tumor-directed T cells. Immunol Lett 1994; 40: 171–8
Disis ML, Smith JW, Murphy AE, et al. In vitro generation of human cytotoxic T-cells specific for peptides derived from the HER-2/neu protooncogene protein. Cancer Res 1994; 54: 1071–6
Barnd DL, Lan MS, Metzgar RS, et al. Specific tumor histo-compatibility complex-unrelated recognition of tumor associated mucins by human cytotoxic T cells. Proc Natl Acad Sci USA 1989; 86: 7159–63
Van der Bruggen P, Traversari C, Chomez P, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on human melanoma. Science 1991; 254: 1643–7
Gaugler B, Van den Eynde B, van der Bruggen P, et al. Human gene MAGE-3 codes for an antigen recognized on a human melanoma by autologous cytolytic T lymphocytes. J Exp Med 1994; 179: 921–30
Coulie PG, Brichard V, Van Pel A, et al. A new gene coding for a differentiation antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas. J Exp Med 1994; 180: 35–42
Kawakami Y, Eliyahu S, Delgado CH, et al. Cloning of the gene coding for a shared human melanoma antigen recognized by autologous T cells infiltrating into tumor. Proc Natl Acad Sci USA 1994; 91: 3515–9
Kawakami Y, Eliyahu S, Sakaguchi K, et al. Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2 restricted tumor infiltrating lymphocytes. J Exp Med 1994; 180: 347–52
Brichard V, Van Pel A, Wolfel T, et al. The tyrosinase gene codes for an antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas. J Exp Med 1993; 178: 489–95
Bakker ABH, Schreurs MWJ, de Boer AJ, et al. Melanocyte lineage-specific antigen gp 100 is recognized by melanoma-derived tumor-infiltrating lymphocytes. J Exp Med 1994; 179: 1005–9
Kawakami Y, Eliyahu S, Delgado CH, et al. Identification of a human melanoma antigen recognized by tumor-infiltrating lymphocytes associated with in vivo tumor rejection. Proc Natl Acad Sci USA 1994; 91: 6458–62
Gambacorti-Passerini C. Immunogenicity of fusion proteins: an example of tumor-specific/transformation-related antigens. Int J Clin Lab Res 1993; 23: 186–91
Goulmy E. Minor histocompatibility antigens in man and their role in transplantation. Transplant Rev 1988; 2: 29–53
Van der Harst D, Goulmy E, Falkenburg JHF, et al. Recognition of minor histocompatibility antigens on lymphocytic and my-eloid leukemic cells by cytotoxic T-cell clones. Blood 1994; 83: 1060–6
de Buerger M, Bakker A, van Rood JJ, et al. Tissue distribution of minor histocompatibility antigens. Ubiquitous versus restricted tissue distribution indicated heterogeneity amongst human cytotoxic T lymphocyte-defined non-MHC antigens. J Immunol 1992; 149: 1788–94
Jung S, Schluesener HJ. Human T lymphocytes recognize a peptide of a single point-mutated, oncogenic ras proteins. J Exp Med 1991; 173: 273–6
Meeker T, Lowder J, Cleary ML, et al. Emergence of idiotype variants during treatment of B-cell lymphoma with anti-idiotype antibodies. N Engl J Med 1985; 312: 1658–65
Cheeveu MA, Disis ML, Bernhard H, et al. Immunity to oncogenic proteins. Immunol Rev. In press
Cline MJ. The molecular basis of leukemia. N Engl J Med 1994; 330: 328–36
Barbacid M. Ras genes. Ann Rev Biochem 1987; 56: 779–827
Bos JL. Ras oncogenes in human cancer: a review. Cancer Res 1989; 49: 4682–9
Seeburg PH, Colby WW, Capon DJ, et al. Biological properties of human c-Ha-ras1 genes mutated at codon 12. Nature 1984; 312: 71–5
Der CJ, Finkel T, Cooper GM. Biological and biochemical properties of human rasH genes mutated at codon 61. Cell 1986; 44: 167–76
Healy F, Drouet C, Romero P, et al. Conversion of a self peptide sequence into a Kd-restricted neoantigen by Tyr substitution. J Exp Med 1991; 174: 1657–60
Peace DJ, Chen W, Nelson H, et al. T cell recognition of transforming proteins encoded by mutated ras proto-oncogenes. J Immunol 1991; 146: 2059–65
Fenton RG, Taub DD, Kwak LW, et al. Cytotoxic T-cell response and in vivo protection against tumor cells harboring activated ras proto-oncogenes. J Natl Cancer Inst 1993; 85: 1294–302
Skipper J, Stauss HS. Identification of two cytotoxic T lymphocyte-recognized epitopes in the ras protein. J Exp Med 1993; 177: 1493–8
Peace DJ, Smith JW, Chen W, et al. Lysis of ras oncogene-trans-formed cells by specific cytotoxic T lymphocytes elicited by primary in vitro immunization with mutated ras peptide. J Exp Med 1994; 179: 473–9
Gedde-Dahl T III, Fossum B, Eriksen JA, et al. T cell clones specific for p21 ras-derived peptides: characterization of their fine specificity and HLA restriction. Eur J Immunol 1993; 23: 754–60
Fossum B, Gedde-Dahl III T, Hansen T, et al. Overlapping epitopes encompassing a point mutation (12 Gly-Arg) in p21 ras can be recognized by HLA-DR, -DP and -DQ restricted T cells. Eur J Immunol 1993; 23: 2687–91
Gedde-Dahl III T, Spurkland A, Fossum B, et al. T cell epitopes encompassing the mutational hot spot position 61 of p21 ras. Promiscuity in ras peptide binding to HLA. Eur J Immunol 1994; 24: 410–4
Nishisho I, Nakamura Y, Miyoshi Y, et al. Mutations of chromosome 5p21 genes in FAP and colorectal cancer patients. Science 1991; 253: 665–9
Miyoshi Y, Nagase H, Ando H, et al. Somatic mutations of the APC gene of the colorectal tumors: mutation cluster region in the APC gene. Hum Mol Genet 1992; 1: 229–33
Townsend A, Ohlen C, Rogers M, et al. Source of unique tumor antigens [letter]. Nature 1994; 371: 662
Rabbitts TH. Chromosomal transloctions in human cancer. Nature 1994; 372: 143–9
de Klein A, van Kessel AG, Grosveld G, et al. A cellular onco-gene is translocated to the Philadelphia chromosome in chronic myelocytic leukemia. Nature 1982; 300: 765–7
Mitani K, Ogawa S, Tanaka T, et al. Generation of the AML1-EVI-1 fusion gene in the t(3; 21)(q26; q22) causes blastic crisis in chronic myelocytic leukemia. EMBO J 1994; 13: 504–10
Gillard EF, Solomon E. Acute promyelocytic leukemia and the t(15; 17) translocation. Semin Cancer Biol 1993; 4: 359–67
Chen Z, Brand NJ, Chen A, et al. Fusion between a novel kruppel-like zink finger gene and the retinoic acid receptor-alpha locus due to a variant t(l 1; 17) translocation associated with acute promyelocytic leukaemia. EMBO J 1993; 12: 1161–7
Kamps MP, Murre C, Sun XH, et al. A new homeobox gene contributes the DNA binding domain of the t(1; 19) translocation protein in pre-B ALL. Cell 1990; 60: 547–55
Nourse J, Mellentin JD, Galili N, et al. Chromosomal translocation t(l; 19) results in synthesis of a homeobox fusion mRNA that codes for a potential chimeric transcription factor. Cell 1990; 60: 535–45
Hunger SP, Ohyashiki K, Toyama K, et al. HLF, a novel hepatic bZIP protein, shows altered DNA-binding properties following fusion to E2A in t(17; 19) acute lymphoblastic leukemia. Genes Dev 1992; 6: 1608–20
Inaba T, Roberts WM, Shapiro LH, et al. Fusion of the leucine zipper gene HLF to the E2A gene in human acute B-lineage leukemia. Science 1992; 257: 531–4
Djabali M, Selleri L, Parry P, et al. A trithorax-like gene is interrupted by chromosome 11q23 translocations in acute leu-kemias. Nat Genet 1992; 2: 113–8
Gu Y, Nakamura T, Alder H, et al. The t(4; 11) chromosome translocation of human acute leukemias fuses the ALL-1 gene, related to drosophila trithorax, to the AF-4 gene. Cell 1992; 71: 701–8
Tkachuk DC, Kohler S, Cleary ML. Involvement of a homolog of drosophila trithorax by 11q23 chromosomal translocations in acute leukemias. Cell 1992; 71: 691–700
Yamamoto K, Seto M, Komatsu H, et al. Two distinct portions of LTG19/ENL at 19p13 are involved in t(11; 19) leukemia. Oncogene 1993; 8: 2617–25
Parsad R, Gu Y, Alder H, et al. Cloning of the ALL-1 fusion partner, the AF-6 gene, involved in acute myeloid leukemias with the t(6; 11) chromosome translocation. Cancer Res 1993; 53: 5624–8
Baer R. TALI, TAL2 and LYL1: a family of basic helix-loop-helix proteins implicated in T cell acute leukaemia. Semin Cancer Biol 1993; 4: 341–7
Ohki M. Molecular basis of the t(8; 21) translocation in acute myeloid leukaemia. Semin Cancer Biol 1993; 4: 369–75
von Lindern M, Fornerod M, van Baal S, et al. The translocation (6; 9), associated with a specific subtype of acute myeloid leukemia, results in the fusion of two genes, dek and can, and the expression of a chimeric, leukemia-specific dek-can mRNA. Mol Cell Biol 1992; 12: 1687–97
Ichikawa H, Shimizu K, Hayashi Y, et al. An RNA-binding protein gene, TLS/FUS, is fused to ERG in human myeloid leukemia with t(16; 21) chromosomal translocation. Cancer Res 1994; 54: 2865–8
Liu P, Tarle SA, Hajra A, et al. Fusion between transcription factor CBF beta/PEBP2 beta and a myosin heavy chain in acute myeloid leukemia. Science 1993; 261: 1041–4
Morris SW, Kirstein MN, Valentine MB, et al. Fusion of a kinase gene, ALK, to a nuclear protein gene, NPM, in non-Hodgkin’s lymphoma. Science 1994; 263: 1281–4
Baer R, Chen KC, Smith SD, et al. Fusion of an immunoglob-ulin variable gene and a T cell receptor constant gene in the chromosome 14 inversion associated with T cell tumors. Cell 1985; 43: 705–13
Denny CT, Yoshikai Y, Mak TW, et al. A chromosome 14 inversion in a T-cell lymphoma is caused by a site-specific recombination between immunoglobulin and T-cell receptor loci. Nature 1986; 320: 549–51
Nucifora G, Begy CR, Erickson P, et al. The 3; 21 translocation in myelodysplasia results in a fusion transcript between the AML1 gene and the gene for EAP, a highly conserved protein associated with the Epstein-Barr virus small RNA EBER 1. Proc Natl Acad Sci USA 1993; 90: 7784–8
Golub TR, Barker GF, Lovett M, et al. Fusion of PDGF receptor beta to a novel ets-like gene, tel, in chronic myelomonocytic leukemia with t(5; 12) chromosomal translocation. Cell 1994; 77: 307–16
Pierotti MA, Santoro M, Jenkins RB, et al. Characterization of an inversion on the long arm of chromosome 10 juxtaposing D10S170 and RET and creating the oncogenic sequence RET/PTC. Proc Natl Acad Sci USA 1992; 89: 1616–20
Greco A, Pierotti MA, Bongarzone I, et al. TRK-T1 is a novel oncogene formed by the fusion of TPR and TRK genes in human papillary thyroid carcinomas. Oncogene 1992; 7: 237–42
Delattre O, Zucman J, Plougastel B, et al. Gene fusion with an ETS DNA-binding domain caused by chromosome translo-cation in human tumours. Nature 1992; 359: 162–5
Sorensen PH, Lessnick SL, Lopez-Terrada D, et al. A second Ewing’s sarcoma translocation, t(21; 22), fuses the EWS gene to another ETS-family transcription factor, ERG. Nat Genet 1994; 6: 146–51
Zucman J, Delattre O, Desmaze C, et al. EWS and ATF-1 gene fusion induced by t(12; 22) translocation in malignant melanoma of soft parts. Nat Genet 1993; 4: 341–5
Crozat A, Aman P, Mandahl N, et al. Fusion of CHOP to a novel RNA-binding protein in human myxoid liposarcoma. Nature 1993; 363: 640–4
Rabbitts TH, Forster A, Larson R, et al. Fusion of the dominant negative transcription regulator CHOP with a novel gene FUS by translocation t(12; 16) in malignant liposarcoma. Nat Genet 1993; 4: 175–80
Shapiro DN, Sublett JE, Li B, et al. Fusion of PAX3 to a member of the forkhead family of transcription factors in human alveolar rhabdomyosarcoma. Cancer Res 1993; 53: 5108–12
Clark J, Rocques PJ, Crew AJ, et al. Identification of novel genes, SYT and SSX, involved in the t(X; 18)(p11.2; q11.2) translocation found in human synovial sarcoma. Nat Genet 1994; 7: 502–8
Chen W, Peace DJ, Rovira DK, et al. T-cell immunity to the joining region of p210BCR-ABL protein. Proc Natl Acad Sci USA 1992; 89: 1468–72
ten Bosch GJ, Joosten A, Melief CJM, et al. Cross reactivity of BCR-ABL peptide specific T cells with EBV transformed B cells [abstract]. Blood 1994; 84: 432
Gambacorti-Passerini C, Grignani F, Arienti F, et al. Human CD4 lymphocytes specifically recognize a peptide representing the fusion region of the hybrid protein pml/RARa present in acute promyelocytic leukemia cells. Blood 1993; 81: 1369–75
Lanzavecchia A. Identifying strategies for immune intervention. Science 1993; 260: 937–44
Germain RN. MHC-dependent antigen processing and peptide presentation: providing ligands for T lymphocyte activation. Cell 1994; 76: 287–99
Guinan EC, Gribben JG, Boussiotis VA, et al. Pivotal role of the B7: CD28 pathway in transplantation tolerance and tumor immunity. Blood 1994; 84: 3261–82
June CH, Bluestone JA, Nadler LM, et al. The B7 and CD28 receptor families. Immunol Today 1994; 15: 321–31
June CH, Ledbetter JA, Linsley PS, et al. Role of the CD28 receptor in T-cell activation. Immunol Today 1990; 11: 211–6
Linsley PS, Ledbetter JA. The role of the CD28 receptor during T cell responses to antigen. Annu Rev Immunol 1993; 11: 191–212
Schwartz RH. Costimulation of T lymphocytes: the role of CD28, CTLA-4, and B7/BB1 in interleukin-2 production and immunotherapy. Cell 1992; 71: 1065–8
Jenkins MK, Ashwell JD, Schwartz RH. Allogeneic non-T spleen cells restore the responsiveness of normal T cell clones stimulated with antigen and chemically modified antigen-presenting cells. J Immunol 1988; 140: 3324–30
Webb S, Morris C, Sprent J. Extrathymic tolerance of mature T cells: clonal elimination as a consequence of immunity. Cell 1990; 63: 1249–56
Chen L, Ashe S, Brady WA, et al. Costimulation of antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4. Cell 1992; 71: 1093–102
Townsend SE, Allison JP. Tumor rejection after direct costimulation of CD8+ T cells by B7-transfected melanoma cells. Science 1993; 259: 368–70
Dranoff G, Jaffee E, Lazenby A, et al. Vaccinated with irradiated tumuor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc Natl Acad Sci USA 1993; 90: 3539–43
Ramarathinam L, Castle M, Wu Y, et al. T cell costimulation by B7/BB1 induces CD8 T cell-dependent tumor rejection: an important role of B7/BB1 in the induction, recruitment, and effector function of antitumor T cells. J Exp Med 1994; 179: 1205–14
Chen L, McGowan P, Ashe S, et al. Tumor immunogenicity determines the effect of B7 costimulation on T cell-mediated tumor immunity. J Exp Med 1994; 179: 523–32
Matulonis UA, Dosiou C, Lamont C, et al. Role of B7-1 in mediating an immune response to myeloid leukemia cells. Blood 1995; 85: 2507–15
Trinchieri G, Wysocka M, D’Andrea A, et al. Natural killer cell stimulatory factor (NKSF) or interleukin-12 is a key regulator of immune response and inflammation. Prog Growth Factor Res 1992; 4: 355–68
Brunda MJ. Interleukin-12. J Leukoc Biol 1994; 55: 280–8
Nastala CL, Edington HD, McKinney TG, et al. Recombinant IL-12 administration induces tumor regression in association with IFN-gamma production. J Immunol 1994; 153: 1697–706
Author information
Authors and Affiliations
Rights and permissions
About this article
Cite this article
Dermime, S., Barrett, J. & Gambacorti-Passerini, C. The Role of the Immune System in Anti-Tumour Responses. Drugs & Aging 7, 266–277 (1995). https://doi.org/10.2165/00002512-199507040-00002
Published:
Issue Date:
DOI: https://doi.org/10.2165/00002512-199507040-00002