Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Modulating T-cell immunity to tumours: new strategies for monitoring T-cell responses

Key Points

  • The identification of many human tumour antigens as potential targets for immunotherapy has led to clinical trials to augment the tumour immune response through the use of vaccines and adoptively transferred T cells. Immunological monitoring in these studies will be crucial for understanding the principles that underlie successful immunotherapeutic strategies.

  • Recent advances in immunological monitoring now enable more-direct detection of individual antigen-specific T cells on the basis of structural or functional properties. These advanced assays provide a degree of specificity and sensitivity that was not previously possible with classical cytolytic or proliferation assays.

  • Structure-based assays — through the use of peptide–MHC (major histocompatibility complex) multimers that bind antigen-specific T-cell receptors (TCRs), or quantitative polymerase chain reaction (PCR) assays that detect clone-specific regions of the TCR — provide an estimation of the total number of T cells independent of functional status. Such tests are highly sensitive but can be difficult to implement in large-scale studies.

  • Function-based assays — such as ELISPOT analysis, which detects antigen-specific T cells on the basis of proximal cytokine production — provide a reliable measure of the reactive T-cell population and are amenable to high-throughput analysis. However, ELISPOT analysis and other function-based assays — intracellular cytokine analysis, quantitative real-time PCR analysis of cytokine expression — might underestimate or fail to detect naive, anergic or functionally unresponsive antigen-specific T cells.

  • Judicious use of these advanced assays to monitor the immune response in clinical trials can be used to address questions related to the magnitude, homing property, function and avidity of anti-tumour T cells that are required for effective therapy.

  • An important corollary to an evaluation of the T-cell response will be an understanding of the tumour response to immune manipulation, particularly an evaluation of potential mechanisms of immune escape.

  • Pre-clinical and clinical studies have shown that tumour cells might circumvent the immune response through defective antigen expression and presentation, inhibition of T-cell effector function and induction of anti-apoptotic mechanisms.

  • Occasional application of immunological assays to current clinical trials has not always shown a correlation between increased immune response and clinical response. It is anticipated that incorporation of these advanced assays into future trials of antigen-specific immunotherapy, accompanied by evaluation for mechanisms of tumour immune escape, will help to explain these discrepancies and elucidate requirements for more-effective therapy.

Abstract

Advances in immunological monitoring provide the means to analyse the cellular immune response with greater sensitivity and precision than ever before. Novel immunological tools can be used not only to quantify the antigen-specific response, but also to analyse the phenotype and function of individual effector cells. Application of these tools to dissect the antitumour responses will lead to a greater understanding of the principles that underlie successful immunotherapeutic strategies and potential mechanisms of tumour immune evasion.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Structure-based T-cell assays.
Figure 2: Function-based T-cell assays.
Figure 3: Mechanisms of immune evasion.

Similar content being viewed by others

References

  1. van den Eynde, B. J. & van der Bruggen, P. T cell defined tumor antigens. Curr. Opin. Immunol. 9, 684–693 (1997).

    CAS  PubMed  Google Scholar 

  2. Mitchell, M. S. Perspective on allogeneic melanoma lysates in active specific immunotherapy. Semin. Oncol. 25, 623–635 (1998).

    CAS  PubMed  Google Scholar 

  3. Hsueh, E. C. et al. Active specific immunotherapy with polyvalent melanoma cell vaccine for patients with in-transit melanoma metastases. Cancer 85, 2160–2169 (1999).

    CAS  PubMed  Google Scholar 

  4. Musselli, C., Livingston, P. O. & Ragupathi, G. Keyhole limpet hemocyanin conjugate vaccines against cancer: the Memorial Sloan–Kettering experience. J. Cancer Res. Clin. Oncol. 127 (Suppl. 2), R20–R26 (2001).

    CAS  PubMed  Google Scholar 

  5. Dranoff, G. 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. Proc. Natl Acad. Sci. USA 90, 3539–3543 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Sampson, J. H. et al. Subcutaneous vaccination with irradiated, cytokine-producing tumor cells stimulates CD8+ cell-mediated immunity against tumors located in the 'immunologically privileged' central nervous system. Proc. Natl Acad. Sci. USA 93, 10399–10404 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Simons, J. W. et al. Bioactivity of autologous irradiated renal cell carcinoma vaccines generated by ex vivo granulocyte–macrophage colony-stimulating factor gene transfer. Cancer Res. 57, 1537–1546 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Tuting, T. et al. Autologous human monocyte-derived dendritic cells genetically modified to express melanoma antigens elicit primary cytotoxic T cell responses in vitro: enhancement by cotransfection of genes encoding the TH1-biasing cytokines IL-12 and IFN-α. J. Immunol. 160, 1139–1147 (1998).

    CAS  PubMed  Google Scholar 

  9. Kwak, L. W. et al. Induction of immune responses in patients with B-cell lymphoma against the surface-immunoglobulin idiotype expressed by their tumors. N. Engl. J. Med. 327, 1209–1215 (1992).

    CAS  PubMed  Google Scholar 

  10. Boczkowski, D., Nair, S. K., Snyder, D. & Gilboa, E. Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J. Exp. Med. 184, 465–472 (1996).

    CAS  PubMed  Google Scholar 

  11. Larregina, A. T. et al. Direct transfection and activation of human cutaneous dendritic cells. Gene Ther. 8, 608–617 (2001).

    CAS  PubMed  Google Scholar 

  12. Reichardt, V. L. et al. Idiotype vaccination using dendritic cells after autologous peripheral blood stem cell transplantation for multiple myeloma: a feasibility study. Blood 93, 2411–2419 (1999).

    CAS  PubMed  Google Scholar 

  13. Reddy, S. A., Okada, C., Wong, C., Bahler, D. & Levy, R. T cell antigen receptor vaccines for active therapy of T cell malignancies. Ann. NY Acad. Sci. 941, 97–105 (2001).

    CAS  PubMed  Google Scholar 

  14. Nestle, F. O. et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nature Med. 4, 328–332 (1998).

    CAS  PubMed  Google Scholar 

  15. Fong, L. et al. Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy. Proc. Natl Acad. Sci. USA 98, 8809–8814 (2001).In a clinical trial using dendritic cells pulsed with an altered peptide ligand of CEA to vaccinate patients with colon cancer, the authors show, using tetramer analysis, that expansion of CEA-specific T cells in vivo correlated with clinical responses.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Thurner, B. et al. Vaccination with Mage-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma. J. Exp. Med. 190, 1669–1678 (1999).Regression of individual lesions were observed in 6 out of 11 patients with advanced melanoma receiving a MAGE3-pulsed dendritic-cell vaccine. Antigen-loss variants were identified in residual tumour of some patients.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Rosenberg, S. A., Spiess, P. & Lafreniere, R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science 233, 1318–1321 (1986).

    CAS  PubMed  Google Scholar 

  18. Riddell, S. R. et al. Phase I study of cellular adoptive immunotherapy using genetically modified CD8+ HIV-specific T cells for HIV seropositive patients undergoing allogeneic bone marrow transplant. The Fred Hutchinson Cancer Research Center and the University of Washington School of Medicine, Department of Medicine, Division of Oncology. Hum. Gene Ther. 3, 319–338 (1992).

    CAS  PubMed  Google Scholar 

  19. Yee, C. et al. Melanocyte destruction after antigen-specific immunotherapy of melanoma: direct evidence of T cell-mediated vitiligo. J. Exp. Med. 192, 1637–1644 (2000).In a patient receiving adoptive therapy with T-cell clones targeting MART1, transferred T cells were tracked with peptide–MHC tetramers and shown to migrate to antigen-positive sites of skin and tumour and to mediate an antigen-specific immune response.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Yee, C., Riddell, S. R. & Greenberg, P. D. Prospects for adoptive T cell therapy. Curr. Opin. Immunol. 9, 702–708 (1997).

    CAS  PubMed  Google Scholar 

  21. Yee, C., Savage, P. A., Lee, P. P., Davis, M. M. & Greenberg, P. D. Isolation of high avidity melanoma-reactive CTL from heterogeneous populations using peptide–MHC tetramers. J. Immunol. 162, 2227–2234 (1999).Peptide–MHC tetramers were used to define the TCR affinity of antigen-specific T cells (see also reference 87).

    CAS  PubMed  Google Scholar 

  22. Simon, R. M. et al. Clinical trial designs for the early clinical development of therapeutic cancer vaccines. J. Clin. Oncol. 19, 1848–1854 (2001).

    CAS  PubMed  Google Scholar 

  23. Disis, M. L. et al. Delayed-type hypersensitivity response is a predictor of peripheral blood T-cell immunity after HER-2/neu peptide immunization. Clin. Cancer Res. 6, 1347–1350 (2000).

    CAS  PubMed  Google Scholar 

  24. Habal, N. et al. CancerVax, an allogeneic tumor cell vaccine, induces specific humoral and cellular immune responses in advanced colon cancer. Ann. Surg. Oncol. 8, 389–401 (2001).

    CAS  PubMed  Google Scholar 

  25. Hsueh, E. C., Gupta, R. K., Qi, K. & Morton, D. L. Correlation of specific immune responses with survival in melanoma patients with distant metastases receiving polyvalent melanoma cell vaccine. J. Clin. Oncol. 16, 2913–2920 (1998).

    CAS  PubMed  Google Scholar 

  26. Lee, P. et al. Effects of interleukin-12 on the immune response to a multipeptide vaccine for resected metastatic melanoma. J. Clin. Oncol. 19, 3836–3847 (2001).

    CAS  PubMed  Google Scholar 

  27. Hsu, F. J. et al. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nature Med. 2, 52–58 (1996).

    CAS  PubMed  Google Scholar 

  28. Bendandi, M. et al. Complete molecular remissions induced by patient-specific vaccination plus granulocyte–monocyte colony-stimulating factor against lymphoma. Nature Med. 5, 1171–1177 (1999).

    CAS  PubMed  Google Scholar 

  29. Marchand, M. et al. Tumor regression responses in melanoma patients treated with a peptide encoded by gene MAGE3. Int. J. Cancer 63, 883–885 (1995).

    CAS  PubMed  Google Scholar 

  30. Marchand, M. et al. Tumor regressions observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE-3 and presented by HLA-A1. Int. J. Cancer 80, 219–230 (1999).

    CAS  PubMed  Google Scholar 

  31. Mukherji, B. et al. Induction of antigen-specific cytolytic T cells in situ in human melanoma by immunization with synthetic peptide-pulsed autologous antigen presenting cells. Proc. Natl Acad. Sci. USA 92, 8078–8082 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Cormier, J. N. et al. Enhancement of cellular immunity in melanoma patients immunized with a peptide from Mart1/Melan A. Cancer J. Sci. Am. 3, 37–44 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Rosenberg, S. A. et al. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nature Med. 4, 321–327 (1998).

    CAS  PubMed  Google Scholar 

  34. Jager, E. et al. Monitoring CD8 T cell responses to NY-ESO-1: correlation of humoral and cellular immune responses. Proc. Natl Acad. Sci. USA 97, 4760–4765 (2000).References 26–34 describe some of the clinical trials that involve augmentation of an antigen-specific T-cell response.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Altman, J. D. et al. Phenotypic analysis of antigen-specific T lymphocytes. Science 274, 94–96 (1996).

    CAS  PubMed  Google Scholar 

  36. Davis, M. M. et al. T cell receptor biochemistry, repertoire selection and general features of TCR and Ig structure. Ciba Found. Symp. 204, 94–100; discussion 100–104 (1997).

    CAS  PubMed  Google Scholar 

  37. Schneck, J. P. Monitoring antigen-specific T cells using MHC–Ig dimers. Immunol. Invest. 29, 163–169 (2000).

    CAS  PubMed  Google Scholar 

  38. Yee, C., Davis, M. M. & Lee, P. P. The Use of Peptide–MHC Tetramers in T-Cell Therapy of Melanoma (Academic Press, San Diego, 2000).

    Google Scholar 

  39. Lee, P. P. et al. Characterization of circulating T cells specific for tumor-associated antigens in melanoma patients. Nature Med. 5, 677–685 (1999).Multiparametric analysis of endogenous populations of circulating tumour-specific T cells revealed an activational defect in T cells that recognize melanoma-associated differentiation antigens.

    CAS  PubMed  Google Scholar 

  40. Callan, M. F. et al. Direct visualization of antigen-specific CD8+ T cells during the primary immune response to Epstein–Barr virus in vivo. J. Exp. Med. 187, 1395–1402 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Haanen, J. et al. In situ detection of virus- and tumor-specific T-cell immunity. Nature Med. 6, 1056–1060 (2000).

    CAS  PubMed  Google Scholar 

  42. Lee, K. H. et al. Functional dissociation between local and systemic immune response during anti-melanoma peptide vaccination. J. Immunol. 161, 4183–4194 (1998).

    CAS  PubMed  Google Scholar 

  43. Pongers-Willemse, M. J. et al. Real-time quantitative PCR for the detection of minimal residual disease in acute lymphoblastic leukemia using junctional region specific TaqMan probes. Leukemia 12, 2006–2014 (1998).

    CAS  PubMed  Google Scholar 

  44. Coulie, P. G. et al. A monoclonal cytolytic T-lymphocyte response observed in a melanoma patient vaccinated with a tumor-specific antigenic peptide encoded by gene MAGE-3. Proc. Natl Acad. Sci. USA 98, 10290–10295 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Riddell, S. R. et al. T-cell mediated rejection of gene-modified HIV-specific cytotoxic T lymphocytes in HIV-infected patients. Nature Med. 2, 216–223 (1996).

    CAS  PubMed  Google Scholar 

  46. Thomis, D. C. et al. A FAS-based suicide switch in human T cells for the treatment of graft-versus-host disease. Blood 97, 1249–1257 (2001).

    CAS  PubMed  Google Scholar 

  47. Brodie, S. J. et al. In vivo migration and function of transferred HIV-1-specific cytotoxic T cells. Nature Med. 5, 34–41 (1999).In situ localization of gene-marked T-cell clones in situ following adoptive T-cell therapy.

    CAS  PubMed  Google Scholar 

  48. Lamana, M. L., Segovia, J. C., Guenechea, G. & Bueren, J. A. Systematic analysis of clinically applicable conditions leading to a high efficiency of transduction and transgene expression in human T cells. J. Gene Med. 3, 32–41 (2001).

    CAS  PubMed  Google Scholar 

  49. Kammula, U. S. et al. Functional analysis of antigen-specific T lymphocytes by serial measurement of gene expression in peripheral blood mononuclear cells and tumor specimens. J. Immunol. 163, 6867–6875 (1999).Application of real-time PCR analysis of cytokine expression to evaluate antigen-specific T-cell response to vaccination.

    CAS  PubMed  Google Scholar 

  50. Mocellin, S., Ohnmacht, G. A., Wang, E. & Marincola, F. M. Kinetics of cytokine expression in melanoma metastases classifies immune responsiveness. Int. J. Cancer 93, 236–242 (2001).

    CAS  PubMed  Google Scholar 

  51. Pala, P., Hussell, T. & Openshaw, P. J. M. Flow cytometric measurement of intracellular cytokines. J. Immunol. Methods 243, 107–124 (2000).

    CAS  PubMed  Google Scholar 

  52. Murali-Krishna, K. et al. Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection. Immunity 8, 177–187 (1998).

    CAS  PubMed  Google Scholar 

  53. Becker, C. et al. Adoptive tumor therapy with T lymphocytes enriched through an IFN-γ capture assay. Nature Med. 7, 1159–1162 (2001).

    CAS  PubMed  Google Scholar 

  54. Herr, W., Schneider, J., Lohse, A. W., Meyer zum Buschenfelde, K. H. & Wolfel, T. Detection and quantification of blood-derived CD8+ T lymphocytes secreting tumor necrosis factor-α in response to HLA-A2.1-binding melanoma and viral peptide antigens. J. Immunol. Methods 191, 131–142 (1996).

    CAS  PubMed  Google Scholar 

  55. Gnjatic, S. et al. Strategy for monitoring T cell responses to NY-ESO-1 in patients with any HLA class I allele. Proc. Natl Acad. Sci. USA 97, 10917–10922 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Herr, W. et al. The use of computer-assisted video image analysis for the quantification of CD8+ T lymphocytes producing tumor necrosis factor-α spots in response to peptide antigens. J. Immunol. Methods 203, 141–152 (1997).

    CAS  PubMed  Google Scholar 

  57. Zajac, A. J. et al. Viral immune evasion due to persistence of activated T cells without effector function. J. Exp. Med. 188, 2205–2213 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Taswell, C. Limiting dilution assays for the determination of immunocompetent cell frequencies. III. Validity tests for the single-hit Poisson model. J. Immunol. Methods 72, 29–40 (1984).

    CAS  PubMed  Google Scholar 

  59. Jager, E. et al. Induction of primary NY-ESO-1 immunity: CD8+ T lymphocyte and antibody responses in peptide-vaccinated patients with NY-ESO-1+ cancers. Proc. Natl Acad. Sci. USA 97, 12198–12203 (2000).Stabilization and regression of individual metastases in some patients with melanoma, receiving a peptide vaccine that targets NY-ESO-1, provide evidence of clinical correlation with ELISPOT analyses.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Molldrem, J. J. et al. Evidence that specific T lymphocytes may participate in the elimination of chronic myelogenous leukemia. Nature Med. 6, 1018–1023 (2000).

    CAS  PubMed  Google Scholar 

  61. Lee, K. H. et al. Increased vaccine-specific T cell frequency after peptide-based vaccination correlates with increased susceptibility to in vitro stimulation but does not lead to tumor regression. J. Immunol. 163, 6292–6300 (1999).

    CAS  PubMed  Google Scholar 

  62. Fong, L. et al. Dendritic cell-based xenoantigen vaccination for prostate cancer immunotherapy. J. Immunol. 167, 7150–7156 (2001).

    CAS  PubMed  Google Scholar 

  63. Cheever, M. A., Thompson, D. B., Klarnet, J. P. & Greenberg, P. D. Antigen-driven long term-cultured T cells proliferate in vivo, distribute widely, mediate specific tumor therapy, and persist long-term as functional memory T cells. J. Exp. Med. 163, 1100–1112 (1986).

    CAS  PubMed  Google Scholar 

  64. Ohlen, C., Kalos, M., Hong, D. J., Shur, A. C. & Greenberg, P. D. Expression of a tolerizing tumor antigen in peripheral tissue does not preclude recovery of high-affinity CD8+ T cells or CTL immunotherapy of tumors expressing the antigen. J. Immunol. 166, 2863–2870 (2001).

    CAS  PubMed  Google Scholar 

  65. Ochsenbein, A. F. et al. Immune surveillance against a solid tumor fails because of immunological ignorance. Proc. Natl Acad. Sci. USA 96, 2233–2238 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Mihm, M. C. Jr, Clemente, C. G. & Cascinelli, N. Tumor infiltrating lymphocytes in lymph node melanoma metastases: a histopathologic prognostic indicator and an expression of local immune response. Lab. Invest. 74, 43–47 (1996).

    PubMed  Google Scholar 

  67. Underwood, J. C. Lymphoreticular infiltration in human tumours: prognostic and biological implications: a review. Br. J. Cancer 30, 538–548 (1974).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Watt, A. G. & House, A. K. Colonic carcinoma: a quantitative assessment of lymphocyte infiltration at the periphery of colonic tumors related to prognosis. Cancer 41, 279–282 (1978).

    CAS  PubMed  Google Scholar 

  69. van Nagell, J. R. Jr, Donaldson, E. S., Wood, E. G. & Parker, J. C. Jr. The significance of vascular invasion and lymphocytic infiltration in invasive cervical cancer. Cancer 41, 228–234 (1978).

    PubMed  Google Scholar 

  70. Hanson, H. L. et al. Eradication of established tumors by CD8+ T cell adoptive immunotherapy. Immunity 13, 265–276 (2000).

    CAS  PubMed  Google Scholar 

  71. Speiser, D. E. & Ohashi, P. S. Activation of cytotoxic T cells by solid tumours? Cell. Mol. Life Sci. 54, 263–271 (1998).

    CAS  PubMed  Google Scholar 

  72. Ochsenbein, A. F. et al. Roles of tumour localization, second signals and cross priming in cytotoxic T-cell induction. Nature 411, 1058–1064 (2001).

    CAS  PubMed  Google Scholar 

  73. Hardy, J. et al. Bioluminescence imaging of lymphocyte trafficking in vivo. Exp. Hematol. 29, 1353–1360 (2001).

    CAS  PubMed  Google Scholar 

  74. Mizoguchi, H. et al. Alterations in signal transduction molecules in T lymphocytes from tumor-bearing mice. Science 258, 1795–1798 (1992).

    CAS  PubMed  Google Scholar 

  75. Correa, M. R. et al. Sequential development of structural and functional alterations in T cells from tumor-bearing mice. J. Immunol. 158, 5292–5296 (1997).

    CAS  PubMed  Google Scholar 

  76. Moser, J. M., Gibbs, J., Jensen, P. E. & Lukacher, A. E. CD94-NKG2A receptors regulate antiviral CD8+ T cell responses. Nature Immunol. 3, 189–195 (2002).

    CAS  Google Scholar 

  77. Deeths, M. J., Kedl, R. M. & Mescher, M. F. CD8+ T cells become nonresponsive (anergic) following activation in the presence of costimulation. J. Immunol. 163, 102–110 (1999).

    CAS  PubMed  Google Scholar 

  78. Staveley-O'Carroll, K. et al. Induction of antigen-specific T cell anergy: an early event in the course of tumor progression. Proc. Natl Acad. Sci. USA 95, 1178–1183 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Torre-Amione, G. et al. A highly immunogenic tumor transfected with a murine transforming growth factor type-β1 cDNA escapes immune surveillance. Proc. Natl Acad. Sci. USA 87, 1486–1490 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Novak, E. J., Liu, A. W., Nepom, G. T. & Kwok, W. W. MHC class II tetramers identify peptide-specific human CD4+ T cells proliferating in response to influenza A antigen. J. Clin. Invest. 104, R63–R67 (1999).Authors develop peptide–MHC class II tetramers that recognize human antigen-specific CD4+ T cells.

  81. Toes, R. E., Offringa, R., Blom, R. J., Melief, C. J. & Kast, W. M. Peptide vaccination can lead to enhanced tumor growth through specific T-cell tolerance induction. Proc. Natl Acad. Sci. USA 93, 7855–7860 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Alexander-Miller, M. A., Leggatt, G. R. & Berzofsky, J. A. Selective expansion of high- or low-avidity cytotoxic T lymphocytes and efficacy for adoptive immunotherapy. Proc. Natl Acad. Sci. USA 93, 4102–4107 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Gervois, N., Guilloux, Y., Diez, E. & Jotereau, F. Suboptimal activation of melanoma infiltrating lymphocytes (TIL) due to low avidity of TCR/MHC–tumor peptide interactions. J. Exp. Med. 183, 2403–2407 (1996).

    CAS  PubMed  Google Scholar 

  84. Crawford, F., Kozono, H., White, J., Marrack, P. & Kappler, J. Detection of antigen-specific T cells with multivalent soluble class II MHC covalent peptide complexes. Immunity 8, 675–682 (1998).

    CAS  PubMed  Google Scholar 

  85. Savage, P. A., Boniface, J. J. & Davis, M. M. A kinetic basis for T cell receptor repertoire selection during an immune response. Immunity 10, 485–492 (1999).

    CAS  PubMed  Google Scholar 

  86. Noppen, C., Spagnoli, G. C. & Schaefer, C. Isolation of multiple mRNAs from a few eukaryotic cells: a fast method to obtain templates for RT-PCR. Biotechniques 21, 394–396 (1996).

    CAS  PubMed  Google Scholar 

  87. Dutoit, V. et al. Functional avidity of tumor antigen-specific CTL recognition directly correlates with the stability of MHC/peptide multimer binding to TCR. J. Immunol. 168, 1167–1171 (2002).Peptide–MHC tetramers were used to define the TCR affinity of antigen-specific T cells (see also reference 21).

    CAS  PubMed  Google Scholar 

  88. Kessels, H. W., Wolkers, M. C., van den Boom, M. D., van der Valk, M. A. & Schumacher, T. N. Immunotherapy through TCR gene transfer. Nature Immunol. 2, 957–961 (2001).Designing T cells with a given antigen specificity by the transfer of genes that encode the TCR.

    CAS  Google Scholar 

  89. Urban, J. L., Holland, J. M., Kripke, M. L. & Schreider, H. Immunoselection of tumor cell variants by mice suppressed with ultraviolet radiation. J. Exp. Med. 156, 1025–1041 (1982).

    CAS  PubMed  Google Scholar 

  90. Saleh, F. H., Crotty, K. A., Hersey, P. & Menzies, S. W. Primary melanoma tumour regression associated with an immune response to the tumour-associated antigen Melan-A/ MART-1. Int. J. Cancer 94, 551–557 (2001).

    CAS  PubMed  Google Scholar 

  91. Shankaran, V. et al. IFN-γ and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 410, 1107–1111 (2001).Endogenous immunoselection of tumour cells can lead to the appearance of antigen-loss tumour variants that escape immune detection in the immunocompetent host.

    CAS  PubMed  Google Scholar 

  92. Panelli, M. C. et al. Expansion of tumor–T cell pairs from fine needle aspirates of melanoma metastases. J. Immunol. 164, 495–504 (2000).

    CAS  PubMed  Google Scholar 

  93. Vitale, M. et al. HLA class I antigen and transporter associated with antigen processing (TAP1 and TAP2) down-regulation in high-grade primary breast carcinoma lesions. Cancer Res. 58, 737–742 (1998).

    CAS  PubMed  Google Scholar 

  94. Ferrone, S. & Marincola, F. M. Loss of HLA class I antigens by melanoma cells: molecular mechanisms, functional significance and clinical relevance. Immunol. Today 16, 487–494 (1995).

    CAS  PubMed  Google Scholar 

  95. Hicklin, D. J., Kageshita, T. & Ferrone, S. Development and characterization of rabbit antisera to human MHC-linked transporters associated with antigen processing. Tissue Antigens 48, 38–46 (1996).

    CAS  PubMed  Google Scholar 

  96. Wang, Z., Margulies, L., Hicklin, D. J. & Ferrone, S. Molecular and functional phenotypes of melanoma cells with abnormalities in HLA class I antigen expression. Tissue Antigens 47, 382–390 (1996).

    CAS  PubMed  Google Scholar 

  97. Yang, G., Hellstrom, K. E., Mizuno, M. T. & Chen, L. In vitro priming of tumor-reactive cytolytic T lymphocytes by combining IL-10 with B7–CD28 costimulation. J. Immunol. 155, 3897–3903 (1995).

    CAS  PubMed  Google Scholar 

  98. Beck, C., Schreiber, H. & Rowley, D. Role of TGF-β in immune-evasion of cancer. Microsc. Res. Tech. 52, 387–395 (2001).

    CAS  PubMed  Google Scholar 

  99. Barth, R. J. Jr, Camp, B. J., Martuscello, T. A., Dain, B. J. & Memoli, V.A. The cytokine microenvironment of human colon carcinoma. Lymphocyte expression of tumor necrosis factor-α and interleukin-4 predicts improved survival. Cancer 78, 1168–1178 (1996).

    PubMed  Google Scholar 

  100. Zea, A. H. et al. Alterations in T cell receptor and signal transduction molecules in melanoma patients. Clin. Cancer Res. 1, 1327–1335 (1995).

    CAS  PubMed  Google Scholar 

  101. Maccalli, C. et al. Differential loss of T cell signaling molecules in metastatic melanoma patients' T lymphocyte subsets expressing distinct TCR variable regions. J. Immunol. 163, 6912–6923 (1999).

    CAS  PubMed  Google Scholar 

  102. Bladergroen, B. A. et al. Expression of the granzyme B inhibitor, protease inhibitor 9, by tumor cells in patients with non-Hodgkin and Hodgkin lymphoma: a novel protective mechanism for tumor cells to circumvent the immune system? Blood 99, 232–237 (2002).

    CAS  PubMed  Google Scholar 

  103. Medema, J. P., de Jong, J., van Hall, T., Melief, C. J. & Offringa, R. Immune escape of tumors in vivo by expression of cellular FLICE-inhibitory protein. J. Exp. Med. 190, 1033–1038 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Ambrosini, G., Adida, C. & Altieri, D.C. A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nature Med. 3, 917–921 (1997).

    CAS  PubMed  Google Scholar 

  105. Nakashima, M., Sonoda, K. & Watanabe, T. Inhibition of cell growth and induction of apoptotic cell death by the human tumor-associated antigen RCAS1. Nature Med. 5, 938–942 (1999).

    CAS  PubMed  Google Scholar 

  106. Mellado, M., de Ana, A. M., Moreno, M. C., Martinez, C. & Rodriguez-Frade, J. M. A potential immune escape mechanism by melanoma cells through the activation of chemokine-induced T cell death. Curr. Biol. 11, 691–696 (2001).

    CAS  PubMed  Google Scholar 

  107. Riker, A. I. et al. Development and characterization of melanoma cell lines established by fine-needle aspiration biopsy: advances in the monitoring of patients with metastatic melanoma. Cancer Detect. Prev. 23, 387–396 (1999).References 93–107 describe mechanisms of tumour immune escape through defects in antigen expression or antigen presentation (references 93–96 ), the presence of an immune-suppressive tumour microenvironment (references 97–101 ) and tumour upregulation of immunoprotective mechanisms (references 101–107).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work is supported, in part, by the Cancer Research Institute, National Institutes of Health/National Cancer Institute and the Damon Runyon Cancer Research Foundation. We apologize to authors whose work was not included due to space constraints.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Cassian Yee.

Related links

Related links

DATABASES

Cancer.gov

chronic myelogenous leukaemia

colorectal cancer

melanoma

ovarian cancer

uterine cancer

LocusLink

β2-microglobulin

CD4

CD8

CD45

CD69

CDR3

FAS

FASL

FLICE

FLIP

GM-CSF

granzyme B

IFN-α

IFN-γ

IL-10

IL-12

interleukin-2

MAGE3

MART1

NGFR

NY-ESO-1

PI9

RCAS1

survivin

TNF-α

transforming growth factor-β

Glossary

T-CELL-DEFINED TUMOUR ANTIGENS

Antigens expressed by tumour cells that are recognized by T cells. The types of tumour antigen, and their expression among various tumours and normal tissues are listed in Table 1.

T-CELL RECEPTOR

(TCR). The antigen receptor on CD4+ and CD8+ T cells that recognizes peptides bound to major histocompatibility complex (MHC) molecules on the surface of antigen-presenting cells. The most common form of TCR is composed of a heterodimer of α- and β-chains. The α- and β-chains of the TCR are composed of an immunoglobulin (Ig)-like variable domain and an Ig-like constant domain.

LYMPHOKINE-ACTIVATED KILLER CELLS

(LAKs). These immune cells are generated by incubating peripheral-blood lymphocytes with high doses of IL-2 in vitro. LAK cells are comprised of heterogeneous populations of T cells, natural killer cells and other LAK cells, and mediate non-MHC (major histocompatibility complex)-restricted killing.

TUMOUR-INFILTRATING LYMPHOCYTES

(TILs). These immune cells are cultivated from tumour aspirates with the presumption that lymphocytes that infiltrate the tumour are more likely to be tumour specific. TILs are expanded with high doses of IL-2 in vitro and are comprised of heterogeneous populations of T cells and natural killer cells. Adoptive therapy with TILs or LAKs requires co-administration of high-dose IL-2, which can be accompanied by significant morbidity.

IL-2

Interleukin-2. A growth factor for T cells and natural killer (NK) cells. Antigen-specific T cells express high affinity IL-2 receptors after antigen stimulation. NK cells and LAKs express intermediate-affinity IL-2 receptors and require higher doses of IL-2 to promote growth.

CELLULAR IMMUNITY

Immune response mediated by T lymphocytes.

CD4+ AND CD8+ T CELLS

T cells that express the CD4 surface antigen (CD4+ T cells) recognize cognate antigen in the context of the major histocompatibility complex (MHC) class II complex on antigen-presenting cells, and provide helper function to CD8+ T cells through the release of cytokines and the activation of professional antigen-presenting cells. CD8+ T cells recognize cognate antigen in the context of the MHC class I complex, and mediate direct cell killing through the release of lytic proteins.

DTH

Delayed-type hypersensitivity reaction. A skin test that assays for cell-mediated immune reactions. The test reagent (antigen) is injected just under the skin surface, and the resulting immune reaction caused by T-cell-dependent macrophage activation and inflammation causes a visible induration over the injection site that is measured in 'mm of induration'. The reaction is usually representative of a CD4+ T-cell response to antigen.

HUMORAL IMMUNITY

Immune response mediated by antibodies that are produced by B lymphoctyes.

PEPTIDE–MHC COMPLEX

Antigens are processed and presented on the surface of the cell as peptide fragments that are bound within the cleft of molecules encoded by the major histocompatibility complex (MHC) genes. Peptides bound to class I MHCs are recognized by CD8+ T cells. Peptides bound to class II MHCs are recognized by CD4+ T cells. The peptide sequence that is recognized by T cells in the context of the MHC is also known as an epitope.

RESTRICTING ALLELE

The major histocompatibility complex (MHC) allele that presents a given epitope is known as the restricting allele for that epitope. T cells will recognize the epitope only when presented by its restricting allele.

PHENOTYPIC ANTIBODIES

Antibodies to surface or intracellular proteins that provide additional information regarding the developmental, activational or functional state. For example, activated T cells upregulate expression of the early activation marker CD69 on their cell surface.

CDR3

(Complementarity-determining region 3). A highly variable region of the T-cell receptor (in the variable domain) that mediates T-cell recognition of the peptide–MHC complex. T-cell clones express unique CDR3 regions.

PERIPHERAL-BLOOD MONONUCLEAR CELLS

Nucleated cells of the peripheral blood, which include T and B lymphocytes, monocytes and natural killer cells.

RIBOPROBES

Short ribonucleotide fragments tagged with a radioactive conjugate that are used to identify specific mRNA.

T-CELL INFILTRATE

T cells that accumulate at the site of the tumour. An enriched population of tumour-specific T cells have been identified in T-cell infiltrates.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Yee, C., Greenberg, P. Modulating T-cell immunity to tumours: new strategies for monitoring T-cell responses. Nat Rev Cancer 2, 409–419 (2002). https://doi.org/10.1038/nrc820

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc820

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing