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:

Adoptive-cell-transfer therapy for the treatment of patients with cancer

Key Points

  • Adoptive-cell-transfer (ACT) therapy for patients with cancer relies on the ex vivo generation of highly active, tumour-specific lymphocytes, and their administration in large numbers to the autologous host.

  • Preclinical models have identified characteristics of lymphocyte cultures that are required for successful ACT therapy. The most important characteristic is the presence of high affinity, tumour-antigen-specific CD8+ T cells. There is generally a direct correlation between treatment efficacy and the number of transferred, tumour-specific cells.

  • Preclinical models have also identified ways to manipulate the host immune environment that increase ACT therapeutic efficacy. These include immunosuppression before cell administration and concurrent interleukin 2 administration with the transferred T cells.

  • ACT therapy directed at viral antigens has been effective for elimination of Epstein–Barr virus (EBV)-induced post-transplant lymphoproliferative disease. EBV-specific lymphocyte cultures suitable for ACT therapy were generated by repetitive in vitro stimulation using EBV-transformed lymphoblastoid lines.

  • Lymphocyte cultures that were selected for reactivity against melanoma antigens, including melanocyte-differentiation antigens, mediated cancer regression in some patients with metastatic melanoma. Melanoma-reactive cultures that were suitable for ACT therapy were generated from tumour-infiltrating lymphocytes that were rapidly expanded with anti-CD3 antibody.

  • The generation of tumour-antigen-specific lymphocyte cultures is evolving rapidly, spurred on by the identification of tumour antigens and the T-cell receptors that recognize them.

  • Further improvements to ACT therapy will depend on a deeper understanding of basic immunological processes, including the role of CD4+ T cells in the antitumour inflammatory response, the ability of lymphocytes to persist in vivo and travel to tumours, and the mechanisms of ACT augmentation by previous host immunosuppression.

Abstract

Adoptive immunotherapy — the isolation of antigen-specific cells, their ex vivo expansion and activation, and subsequent autologous administration — is a promising approach to inducing antitumour immune responses. The molecular identification of tumour antigens and the ability to monitor the persistence and transport of transferred cells has provided new insights into the mechanisms of tumour immunotherapy. Recent studies have shown the effectiveness of cell-transfer therapies for the treatment of patients with selected metastatic cancers. These studies provide a blueprint for the wider application of adoptive-cell-transfer therapy, and emphasize the requirement for in vivo persistence of the cells for therapeutic efficacy.

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: Mechanisms that limit immune responses.
Figure 2: Tumour regression by adoptive-cell-transfer therapy Activated T cells can mediate the regression of a large excess of metastatic melanoma.

Similar content being viewed by others

References

  1. Shastri, N., Schwab, S. & Serwold, T. Producing nature's gene-chips: the generation of peptides for display by MHC class I molecules. Annu. Rev. Immunol. 20, 463–493 (2002).

    CAS  PubMed  Google Scholar 

  2. Renkvist, N., Castelli, C., Robbins, P. F. & Parmiani, G. A listing of human tumor antigens recognized by T cells. Cancer Immunol. Immunother. 50, 3–15 (2001).

    CAS  PubMed  Google Scholar 

  3. Robbins, P. F. et al. A mutated beta-catenin gene encodes a melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J. Exp. Med. 183, 1185–1192 (1996).

    CAS  PubMed  Google Scholar 

  4. Jager, E. et al. Simultaneous humoral and cellular immune response against cancer-testis antigen NY-ESO-1: definition of human histocompatibility leukocyte antigen (HLA)-A2-binding peptide epitopes. J. Exp. Med. 187, 265–270 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kawakami, Y. 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. 180, 347–352 (1994).

    CAS  PubMed  Google Scholar 

  6. Kawakami, Y. et al. Recognition of multiple epitopes in the human melanoma antigen gp100 by tumor-infiltrating T lymphocytes associated with in vivo tumor regression. J. Immunol. 154, 3961–3968 (1995).

    CAS  PubMed  Google Scholar 

  7. Guidotti, L. G. & Chisari, F. V. Noncytolytic control of viral infections by the innate and adaptive immune response. Annu. Rev. Immunol. 19, 65–91 (2001).

    CAS  PubMed  Google Scholar 

  8. Harty, J. T., Tvinnereim, A. R. & White, D. W. CD8+ T cell effector mechanisms in resistance to infection. Annu. Rev. Immunol. 18, 275–308 (2000).

    CAS  PubMed  Google Scholar 

  9. 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 

  10. Weber, J. et al. Granulocyte-macrophage-colony-stimulating factor added to a multipeptide vaccine for resected Stage II melanoma. Cancer 97, 186–200 (2003).

    CAS  PubMed  Google Scholar 

  11. Wick, M. et al. Antigenic cancer cells grow progressively in immune hosts without evidence for T cell exhaustion or systemic anergy. J. Exp. Med. 186, 229–238 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Prevost-Blondel, A. et al. Tumor-infiltrating lymphocytes exhibiting high ex vivo cytolytic activity fail to prevent murine melanoma tumor growth in vivo. J. Immunol. 161, 2187–2194 (1998).

    CAS  PubMed  Google Scholar 

  13. 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 

  14. Matzinger, P. The danger model: a renewed sense of self. Science 296, 301–305 (2002).

    CAS  PubMed  Google Scholar 

  15. Shevach, E. M. CD4+ CD25+ suppressor T cells: more questions than answers. Nature Rev. Immunol. 2, 389–400 (2002).

    CAS  Google Scholar 

  16. Shimizu, J., Yamazaki, S. & Sakaguchi, S. Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. J. Immunol. 163, 5211–5218 (1999).

    CAS  PubMed  Google Scholar 

  17. Khong, H. T. & Restifo, N. P. Natural selection of tumor variants in the generation of 'tumor escape' phenotypes. Nature Immunol. 3, 999–1005 (2002).

    CAS  Google Scholar 

  18. Dudley, M. E. et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 298, 850–854 (2002). This report describes adoptive cell transfer with TILs following non-myeloablative chemotherapy for treatment of patients with melanoma, and represents the first successful use of this combination therapy for the treatment of solid malignancy in patients.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Rooney, C. M. et al. Infusion of cytotoxic T cells for the prevention and treatment of Epstein–Barr virus-induced lymphoma in allogeneic transplant recipients. Blood 92, 1549–1555 (1998). This paper describes the generation and clinical use of lymphocyte cultures for ACT therapy of patients with metastatic cancer.

    CAS  PubMed  Google Scholar 

  20. Overwijk, W. W. et al. gp100/pmel 17 is a murine tumor rejection antigen: induction of 'self'-reactive, tumoricidal T cells using high-affinity, altered peptide ligand. J. Exp. Med. 188, 277–286 (1998). Demonstration in an animal model that immune manipulation can result in efficacious tumour treatments mediated through unmutated melanocyte-differentiation antigens.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Sakai, K., Chang, A. E. & Shu, S. Y. Phenotype analyses and cellular mechanisms of the pre-effector T-lymphocyte response to a progressive syngeneic murine sarcoma. Cancer Res. 50, 4371–4376 (1990).

    CAS  PubMed  Google Scholar 

  22. Surman, D. R., Dudley, M. E., Overwijk, W. W. & Restifo, N. P. Cutting edge: CD4+ T cell control of CD8+ T cell reactivity to a model tumor antigen. J. Immunol. 164, 562–565 (2000).

    CAS  PubMed  Google Scholar 

  23. Chang, A. E. et al. Clinical observations on adoptive immunotherapy with vaccine-primed T- lymphocytes secondarily sensitized to tumor in vitro. Cancer Res. 53, 1043–1050 (1993).

    CAS  PubMed  Google Scholar 

  24. Peng, L., Shu, S. & Krauss, J. C. Treatment of subcutaneous tumor with adoptively transferred T cells. Cell Immunol. 178, 24–32 (1997).

    CAS  PubMed  Google Scholar 

  25. Zeh, H. J. 3rd, Perry–Lalley, D., Dudley, M. E., Rosenberg, S. A., & Yang, J. C. High avidity CTLs for two self-antigens demonstrate superior in vitro and in vivo antitumor efficacy. J. Immunol. 162, 989–994 (1999).

    CAS  PubMed  Google Scholar 

  26. Aruga, A., Shu, S. & Chang, A. E. Tumor-specific granulocyte/macrophage colony-stimulating factor and interferon gamma secretion is associated with in vivo therapeutic efficacy of activated tumor-draining lymph node cells. Cancer Immunol. Immunother. 41, 317–324 (1995).

    CAS  PubMed  Google Scholar 

  27. Barth, R. J. Jr, Mule, J. J., Spiess, P. J. & Rosenberg, S. A. Interferon gamma and tumor necrosis factor have a role in tumor regressions mediated by murine CD8+ tumor-infiltrating lymphocytes. J. Exp. Med. 173, 647–658 (1991).

    CAS  PubMed  Google Scholar 

  28. Cameron, R. B., Spiess, P. J. & Rosenberg, S. A. Synergistic antitumor activity of tumor-infiltrating lymphocytes, interleukin 2, and local tumor irradiation. Studies on the mechanism of action. J. Exp. Med. 171, 249–263 (1990).

    CAS  PubMed  Google Scholar 

  29. Cheever, M. A., Greenberg, P. D., Fefer, A. & Gillis, S. Augmentation of the anti-tumor therapeutic efficacy of long-term cultured T lymphocytes by in vivo administration of purified interleukin 2. J. Exp. Med. 155, 968–980 (1982).

    CAS  PubMed  Google Scholar 

  30. Chou, T., Bertera, S., Chang, A. E. & Shu, S. Adoptive immunotherapy of microscopic and advanced visceral metastases with in vitro sensitized lymphoid cells from mice bearing progressive tumors. J. Immunol. 141, 1775–1781 (1988).

    CAS  PubMed  Google Scholar 

  31. 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). This seminal paper describes the use of TILs, together with cyclophosphamide conditioning, to treat advanced tumours in mice.

    CAS  PubMed  Google Scholar 

  32. Bristol, J. A., Schlom, J. & Abrams, S. I. Persistence, immune specificity, and functional ability of murine mutant ras epitope-specific CD4(+) and CD8(+) T lymphocytes following in vivo adoptive transfer. Cell Immunol. 194, 78–89 (1999).

    CAS  PubMed  Google Scholar 

  33. Overwijk, W. W. et al. Tumor regression and autoimmunity following reversal of a functionally tolerant state of self-reactive CD8+ cells. J. Exp. Med. (in the press).

  34. Awwad, M. & North, R. J. Cyclophosphamide-induced immunologically mediated regression of a cyclophosphamide-resistant murine tumor: a consequence of eliminating precursor L3T4+ suppressor T-cells. Cancer Res. 49, 1649–1654 (1989).

    CAS  PubMed  Google Scholar 

  35. Berenson, J. R., Einstein, A. B. Jr & Fefer, A. Syngeneic adoptive immunotherapy and chemoimmunotherapy of a Friend leukemia: requirement for T cells. J. Immunol. 115, 234–238 (1975).

    CAS  PubMed  Google Scholar 

  36. Cheever, M. A., Greenberg, P. D. & Fefer, A. Specificity of adoptive chemoimmunotherapy of established syngeneic tumors. J. Immunol. 125, 711–714 (1980).

    CAS  PubMed  Google Scholar 

  37. North, R. J. Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on elimination of tumor-induced suppressor T cells. J. Exp. Med. 155, 1063–1074 (1982).

    CAS  PubMed  Google Scholar 

  38. Comoli, P. et al. Infusion of autologous Epstein–Barr virus (EBV)-specific cytotoxic T cells for prevention of EBV-related lymphoproliferative disorder in solid organ transplant recipients with evidence of active virus replication. Blood 99, 2592–2598 (2002).

    CAS  PubMed  Google Scholar 

  39. Haque, T. et al. Treatment of Epstein–Barr-virus-positive post-transplantation lymphoproliferative disease with partly HLA-matched allogeneic cytotoxic T cells. Lancet 360, 436–442 (2002).

    PubMed  Google Scholar 

  40. O'Reilly, R. J. et al. Biology and adoptive cell therapy of Epstein–Barr virus-associated lymphoproliferative disorders in recipients of marrow allografts. Immunol. Rev. 157, 195–216 (1997).

    CAS  PubMed  Google Scholar 

  41. Lucas, K. G., Small, T. N., Heller, G., Dupont, B. & O'Reilly, R. J. The development of cellular immunity to Epstein–Barr virus after allogeneic bone marrow transplantation. Blood 87, 2594–2603 (1996).

    CAS  PubMed  Google Scholar 

  42. Grimm, E. A., Mazumder, A., Zhang, H. Z. & Rosenberg, S. A. Lymphokine-activated killer cell phenomenon. Lysis of natural killer-resistant fresh solid tumor cells by interleukin 2- activated autologous human peripheral blood lymphocytes. J. Exp. Med. 155, 1823–1841 (1982).

    CAS  PubMed  Google Scholar 

  43. Mule, J. J., Shu, S., Schwarz, S. L. & Rosenberg, S. A. Adoptive immunotherapy of established pulmonary metastases with LAK cells and recombinant interleukin-2. Science 225, 1487–1489 (1984).

    CAS  PubMed  Google Scholar 

  44. Rosenberg, S. A. et al. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N. Engl. J. Med. 313, 1485–1492 (1985).

    CAS  PubMed  Google Scholar 

  45. Lum, L. G., LeFever, A. V., Treisman, J. S., Garlie, N. K. & Hanson J. P. Jr. Immune modulation in cancer patients after adoptive transfer of anti-CD3/anti-CD28-costimulated T cells-phase I clinical trial. J. Immunother. 24, 408–419 (2001).

    CAS  PubMed  Google Scholar 

  46. Curti, B. D. et al. Phase I trial of anti-CD3-stimulated CD4+ T cells, infusional interleukin-2, and cyclophosphamide in patients with advanced cancer. J. Clin. Oncol. 16, 2752–2760 (1998).

    CAS  PubMed  Google Scholar 

  47. Kim, J. A. et al. Cellular immunotherapy for patients with metastatic colorectal carcinoma using lymph node lymphocytes localized in vivo by radiolabeled monoclonal antibody. Cancer 86, 22–30 (1999).

    CAS  PubMed  Google Scholar 

  48. Rosenberg, S. A. et al. Prospective randomized trial of high-dose interleukin-2 alone or in conjunction with lymphokine-activated killer cells for the treatment of patients with advanced cancer. J. Natl Cancer Inst. 85, 622–632 (1993).

    CAS  PubMed  Google Scholar 

  49. Chang, A. E. et al. Phase II trial of autologous tumor vaccination, anti-CD3-activated vaccine-primed lymphocytes, and interleukin-2 in stage IV renal cell cancer. J. Clin. Oncol. 21, 884–890 (2003).

    CAS  PubMed  Google Scholar 

  50. To, W. C. et al. Systemic adoptive T-cell immunotherapy in recurrent and metastatic carcinoma of the head and neck: a phase 1 study. Arch. Otolaryngol. Head Neck Surg. 126, 1225–1231 (2000).

    CAS  PubMed  Google Scholar 

  51. Plautz, G. E. et al. T-cell adoptive immunotherapy of metastatic renal cell carcinoma. Urology 54, 617–623 (1999).

    CAS  PubMed  Google Scholar 

  52. Meijer, S. L. et al. Adoptive cellular therapy with tumor vaccine draining lymph node lymphocytes after vaccination with HLA-B7/beta2-microglobulin gene-modified autologous tumor cells. J. Immunother. 25, 359–372 (2002).

    CAS  PubMed  Google Scholar 

  53. Meidenbauer, N. et al. Survival and tumor localization of adoptively transferred melan-a-specific T cells in melanoma patients. J. Immunol. 170, 2161–2169 (2003).

    CAS  PubMed  Google Scholar 

  54. Maus, M. V. et al. Ex vivo expansion of polyclonal and antigen-specific cytotoxic T lymphocytes by artificial APCs expressing ligands for the T-cell receptor, CD28 and 4-1BB. Nature Biotechnol. 20, 143–148 (2002).

    CAS  Google Scholar 

  55. Thomas, A. K., Maus, M. V., Shalaby, W. S., June, C. H. & Riley, J. L. A cell-based artificial antigen-presenting cell coated with anti-CD3 and CD28 antibodies enables rapid expansion and long-term growth of CD4 T lymphocytes. Clin. Immunol. 105, 259–272 (2002).

    CAS  PubMed  Google Scholar 

  56. Oelke, M. et al. Ex vivo induction and expansion of antigen-specific cytotoxic T cells by HLA-Ig-coated artificial antigen-presenting cells. Nature Med. 9, 619–624 (2003).

    CAS  PubMed  Google Scholar 

  57. Sili, U. et al. Large-scale expansion of dendritic cell-primed polyclonal human cytotoxic T-lymphocyte lines using lymphoblastoid cell lines for adoptive immunotherapy. J. Immunother. 26, 241–256 (2003).

    PubMed  Google Scholar 

  58. Walter, E. A. et al. Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N. Engl. J. Med. 333, 1038–1044 (1995). This influential paper describes the use of CMV-specific cloned lymphocytes for the treatment of bone-marrow-transplant recipients, and their efficacy in prevention of CMV-induced disease.

    CAS  PubMed  Google Scholar 

  59. Dudley, M. E. et al. Adoptive transfer of cloned melanoma-reactive T lymphocytes for the treatment of patients with metastatic melanoma. J. Immunother. 24, 363–373 (2001).

    CAS  PubMed  Google Scholar 

  60. Dudley, M. E. et al. A phase I study of nonmyeloablative chemotherapy and adoptive transfer of autologous tumor antigen-specific T lymphocytes in patients with metastatic melanoma. J. Immunother. 25, 243–251 (2002). The failure of non-myeloablative chemotherapy followed by administration of highly avid T-cell clones for treatment of patients with melanoma is documented.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Yee, C. et al. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc. Natl Acad. Sci. USA 99, 16168–16173 (2002). An excellent example of the failure of highly avid T-cell clones to induce objective clinical responses in patients with metastatic solid tumours. T-cell function, transport and persistence were monitored.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Tan, R. et al. Rapid death of adoptively transferred T cells in acquired immunodeficiency syndrome. Blood 93, 1506–1510 (1999).

    CAS  PubMed  Google Scholar 

  63. Brodie, S. J. et al. in vivo migration and function of transferred HIV-1-specific cytotoxic T cells. Nature Med. 5, 34–41 (1999).

    CAS  PubMed  Google Scholar 

  64. Marzo, A. L. et al. Tumor-specific CD4+ T cells have a major 'post-licensing' role in CTL mediated anti-tumor immunity. J. Immunol. 165, 6047–6055 (2000).

    CAS  PubMed  Google Scholar 

  65. Donawho, C. K., Pride, M. W. & Kripke, M. L. Persistence of immunogenic pulmonary metastases in the presence of protective anti-melanoma immunity. Cancer Res. 61, 215–221 (2001).

    CAS  PubMed  Google Scholar 

  66. Topalian, S. L. et al. Immunotherapy of patients with advanced cancer using tumor- infiltrating lymphocytes and recombinant interleukin-2: a pilot study. J. Clin. Oncol. 6, 839–853 (1988).

    CAS  PubMed  Google Scholar 

  67. Kradin, R. L. et al. Tumour-infiltrating lymphocytes and interleukin-2 in treatment of advanced cancer. Lancet 1, 577–580 (1989).

    CAS  PubMed  Google Scholar 

  68. Dillman, R. O. et al. Continuous interleukin-2 and tumor-infiltrating lymphocytes as treatment of advanced melanoma. A national biotherapy study group trial. Cancer 68, 1–8 (1991).

    CAS  PubMed  Google Scholar 

  69. Arienti, F. et al. Adoptive immunotherapy of advanced melanoma patients with interleukin-2 (IL-2) and tumor-infiltrating lymphocytes selected in vitro with low doses of IL-2. Cancer Immunol. Immunother. 36, 315–322 (1993).

    CAS  PubMed  Google Scholar 

  70. Figlin, R. A. et al. Treatment of metastatic renal cell carcinoma with nephrectomy, interleukin-2 and cytokine-primed or CD8(+) selected tumor infiltrating lymphocytes from primary tumor. J. Urol. 158, 740–745 (1997).

    CAS  PubMed  Google Scholar 

  71. Quattrocchi, K. B. et al. Pilot study of local autologous tumor infiltrating lymphocytes for the treatment of recurrent malignant gliomas. J. Neurooncol. 45, 141–157 (1999).

    CAS  PubMed  Google Scholar 

  72. Valmori, D. et al. Circulating tumor-reactive CD8(+) T cells in melanoma patients contain a CD45RA(+)CCR7(−) effector subset exerting ex vivo tumor-specific cytolytic activity. Cancer Res. 62, 1743–1750 (2002).

    CAS  PubMed  Google Scholar 

  73. Finke, J. H. et al. Characterization of tumor-infiltrating lymphocyte subsets from human renal cell carcinoma: specific reactivity defined by cytotoxicity, interferon-gamma secretion, and proliferation. J. Immunother. Emphasis. Tumor Immunol. 15, 91–104 (1994).

    CAS  PubMed  Google Scholar 

  74. Hom, S. S., Rosenberg, S. A. & Topalian, S. L. Specific immune recognition of autologous tumor by lymphocytes infiltrating colon carcinomas: analysis by cytokine secretion. Cancer Immunol. Immunother. 36, 1–8 (1993).

    CAS  PubMed  Google Scholar 

  75. Hoshino, T. et al. HLA class-I-restricted and tumor-specific CTL in tumor-infiltrating lymphocytes of patients with gastric cancer. Int. J. Cancer 70, 631–638 (1997).

    CAS  PubMed  Google Scholar 

  76. Nakamura, H., Ishiguro, K., & Mori, T. Different immune functions of peripheral blood, regional lymph node, and tumor infiltrating lymphocytes in lung cancer patients. Cancer 62, 2489–2497 (1988).

    CAS  PubMed  Google Scholar 

  77. Schwartzentruber, D. J., Solomon, D., Rosenberg, S. A. & Topalian, S. L. Characterization of lymphocytes infiltrating human breast cancer: specific immune reactivity detected by measuring cytokine secretion. J. Immunother. 12, 1–12 (1992).

    CAS  PubMed  Google Scholar 

  78. Itoh, K., Tilden, A. B., & Balch, C. M. Interleukin 2 activation of cytotoxic T-lymphocytes infiltrating into human metastatic melanomas. Cancer Res. 46, 3011–3017 (1986).

    CAS  PubMed  Google Scholar 

  79. Topalian, S. L., Solomon, D., & Rosenberg, S. A. Tumor-specific cytolysis by lymphocytes infiltrating human melanomas. J. Immunol. 142, 3714–3725 (1989).

    CAS  PubMed  Google Scholar 

  80. Yannelli, J. R. et al. Growth of tumor-infiltrating lymphocytes from human solid cancers: summary of a 5-year experience. Int. J. Cancer 65, 413–421 (1996).

    CAS  PubMed  Google Scholar 

  81. Dudley, M. E., Wunderlich, J. R., Shelton, T. E., Even, J. & Rosenberg, S. A. Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J. Immunother. 26, 332–342 (2003).

    PubMed  PubMed Central  Google Scholar 

  82. Rosenberg, S. A. et al. Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. J. Natl Cancer Inst. 86, 1159–1166 (1994). A comprehensive summary of the clinical experience with TIL for the treatment of patients with melanoma in the absence of non-myeloablative conditioning.

    CAS  PubMed  Google Scholar 

  83. Rosenberg, S. A. et al. Gene transfer into humans: immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction. N. Engl. J. Med. 323, 570–578 (1990).

    CAS  PubMed  Google Scholar 

  84. Maine, G. N. & Mule, J. J. Making room for T cells. J. Clin. Invest. 110, 157–159 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Fry, T. J. & Mackall, C. L. Interleukin-7: master regulator of peripheral T-cell homeostasis? Trends. Immunol. 22, 564–571 (2001).

    CAS  PubMed  Google Scholar 

  86. Phan, G. Q., Attia, P., Steinberg, S. M., White, D. E. & Rosenberg, S. A. Factors associated with response to high-dose interleukin-2 in patients with metastatic melanoma. J. Clin. Oncol. 19, 3477–3482 (2001).

    CAS  PubMed  Google Scholar 

  87. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. van Elsas, A., Hurwitz, A. A. & Allison, J. P. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J. Exp. Med. 190, 355–366 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Overwijk, W. W. et al. Vaccination with a recombinant vaccinia virus encoding a 'self' antigen induces autoimmune vitiligo and tumor cell destruction in mice: requirement for CD4(+) T lymphocytes. Proc. Natl Acad. Sci. USA 96, 2982–2987 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Phan, G. Q. et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc. Natl Acad. Sci. USA 100, 8372–8377 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Clay, T. M. et al. Efficient transfer of a tumor antigen-reactive TCR to human peripheral blood lymphocytes confers anti-tumor reactivity. J. Immunol. 163, 507–513 (1999).

    CAS  PubMed  Google Scholar 

  92. Hwu, P. et al. In vivo antitumor activity of T cells redirected with chimeric antibody/T-cell receptor genes. Cancer Res. 55, 3369–3373 (1995).

    CAS  PubMed  Google Scholar 

  93. Wherry, E. J. et al. Lineage relationship and protective immunity of memory CD8 T cell subsets. Nature Immunol. 4, 225–234 (2003).

    CAS  Google Scholar 

  94. Geginat, J., Lanzavecchia, A. & Sallusto, F. Proliferation and differentiation potential of human CD8+ memory T-cell subsets in response to antigen or homeostatic cytokines. Blood 101, 4260–4266 (2003).

    CAS  PubMed  Google Scholar 

  95. Liu, K. & Rosenberg, S. A. Transduction of an IL-2 gene into human melanoma-reactive lymphocytes results in their continued growth in the absence of exogenous IL-2 and maintenance of specific antitumor activity. J. Immunol. 167, 6356–6365 (2001).

    CAS  PubMed  Google Scholar 

  96. Sun, J. C. & Bevan, M. J. Defective CD8 T cell memory following acute infection without CD4 T cell help. Science 300, 339–342 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Shedlock, D. J. & Shen, H. Requirement for CD4 T cell help in generating functional CD8 T cell memory. Science 300, 337–339 (2003).

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mark E. Dudley.

Related links

Related links

DATABASES

Cancer.gov

melanoma

LocusLink

β-catenin

CD4

CD8

CD25

CTLA-4

FASL

gp100

HLA-A2

IFN-γ

IL-2

MART-1

NY-ESO-1

TRAIL

tumour-necrosis factor-α

FURTHER INFORMATION

Steven A. Rosenberg's lab

Glossary

CYTOTOXIC T LYMPHOCYTES

(CTLs). T lymphocytes that exert a cytolytic function following engagement by their T-cell antigen receptor on target cells. CTLs express the co-receptor CD8 and recognize antigenic peptides (or CTL epitopes) that are presented by HLA-class-I molecules.

MHC-CLASS-I MOLECULES

Highly polymorphic glycoproteins that are expressed by every nucleated cell of vertebrates, and that are encoded by the gene cluster 'major histocompatibility complex' (MHC). The human MHC molecules are termed HLA (human leukocyte antigen) molecules. MHC-class-I molecules mainly present peptides from intracellular proteins to cytotoxic T cells.

HLA-RESTRICTED ANTIGENS

T-cell receptors recognize antigen peptides on the surface of antigen-presenting cells in the context of an HLA molecule. Each HLA allele can bind only a fraction of the potential peptide pool, and this 'restricts' the peptide repertoire. So, the HLA-restricted antigenic peptides provide a unique immunological 'signature' that allows immune discrimination of tumour cells, but not normal cells.

MHC-CLASS-II MOLECULES

Peptide receptors, similar to class-I molecules in structure and function, but expressed by a small set of professional antigen-presenting immune cells. They mainly present peptides from extracellular proteins to T-helper cells.

CD4+ T CELLS

T cells bearing the CD4 surface glycoprotein, which recognizes MHC-class-II molecules. These cells provide helper function to CD8+ T cells through the release of cytokines and the activation of professional antigen-presenting cells.

CD8+ T CELLS

T cells bearing the CD8 cell-surface glycoprotein, which recognizes MHC-class-I molecules on target cells. CD8+ T cells are usually cytotoxic T cells.

ANERGIC T CELLS

T cells that are unable to undergo proliferation, secretion of inflammatory cytokines or other functions in response to antigens.

SYNGENEIC TUMOURS

Tumours derived from mice with an identical genetic background; specifically, the same inbred line.

GRAFT-VERSUS-HOST DISEASE

A toxic reaction that is mediated by donor-derived T lymphocytes within the graft towards the recipient's organs. The attack is usually directed toward the skin, gut, liver and haematopoietic cells.

CO-STIMULATION

Optimal stimulation of T-cell proliferation requires two signals. Signal one is transduced through T-cell-antigen receptors. Signal two is generically referred to as co-stimulation; several receptors on T cells have been reported to mediate co-stimulation, including CD28.

LYMPHOKINE-ACTIVATED KILLER CELLS

(LAKs). Lymphocytes that have been cultured in high concentrations of cytokines (including interleukin 2). They show characteristic functional and phenotypic properties, such as in vitro lysis of tumour cells, in the absence of MHC restriction.

CD3

A multimeric component of the T-cell-receptor signalling apparatus. Direct activation of T cells by antibody cross-linking of CD3 complexes on the cell surface bypasses the requirement for T-cell receptor engagement of HLA-restricted antigens.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Dudley, M., Rosenberg, S. Adoptive-cell-transfer therapy for the treatment of patients with cancer. Nat Rev Cancer 3, 666–675 (2003). https://doi.org/10.1038/nrc1167

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc1167

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