CD4+ T cells kill HLA-class-II-antigen-positive melanoma cells presenting peptide in vitro

Purpose: Most melanoma cell lines express HLA class II antigens constitutively or can be induced to do so with interferon γ (IFNγ). We have previously demonstrated that peptide-specific CD4+ T cells proliferate in response to HLA-class-II-antigen-mediated peptide presentation by melanoma cells in vitro and produce interleukin-10 (IL-10) and (IFNγ). We asked whether the responding T cells kill the tumor cells and, if so, whether direct cell contact was required. Methods: Two HLA class II+ melanoma cell lines derived from metastases were co-cultured with a human CD4+ T cell clone specific for influenza hemagglutinin peptide (HA). T cells, melanoma, and HA were co-cultured for 48 h. Melanoma cells with and without HA and/or T cells served as controls. After 36 h, the medium was removed for cytokine analysis by enzyme-linked immunosorbent assay (ELISA). Twelve hours later non-adherent cells were washed away and the adherent melanoma cells were trypsinized and counted. Dual-chamber culture plates were used to determine whether cell contact and/or exposure to cytokine were required for tumor cell death. Results: Melanoma cell counts were over 80% lower in wells containing T cells than in wells with melanoma and peptide alone (P < 0.05). ELISA of supernatants revealed production of IFNγ and IL-10 by the responding T cells. Direct T cell contact with tumor cells was not required for tumor cell death, as melanoma cells were killed when they shared medium but had no contact with T cells responding to peptide presentation by HLA-class-II-antigen-positive melanoma cells in a separate chamber. Blocking antibody to IFNγ but not IL-10 prevented melanoma cell death at levels of cytokine similar to that present in co-culture assays. Conclusions: Peptide-specific CD4+ T cells kill melanoma cells in vitro when they recognize peptide presented by the tumor cell in the context of HLA class II antigen. Direct cell contact is not required, suggesting that it is a cytokine-mediated event. Immunotherapy, using primed CD4+ T cells and peptide, may be beneficial in patients whose tumors express HLA class II antigens or can be induced to do so with IFNγ.

[1]  S. Ferrone,et al.  DR (Ia-like) antigens on human melanoma cells. Serological detection and immunochemical characterization , 1979, The Journal of experimental medicine.

[2]  B. Dörken,et al.  Induction of HLA class‐II antigen expression on human carcinoma cell lines by IFN‐gamma , 1985, International journal of cancer.

[3]  J. Becker,et al.  Tumor escape mechanisms from immunosurveillance: induction of unresponsiveness in a specific MHC-restricted CD4+ human T cell clone by the autologous MHC class II+ melanoma. , 1993, International immunology.

[4]  B. Dréno,et al.  HLA class II‐restricted recognition of common tumor epitopes on human melanoma cells by CD4+ melanoma‐infiltrating lymphocytes , 1995, European journal of immunology.

[5]  E. Bröcker,et al.  HLA-DR antigen expression in primary melanomas of the skin. , 1984, The Journal of investigative dermatology.

[6]  S. Rosenberg,et al.  Human CD4+ T cells specifically recognize a shared melanoma-associated antigen encoded by the tyrosinase gene. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[7]  B. Giannotti,et al.  Ki67 antigen expression correlates with tumor progression and HLA-DR antigen expression in melanocytic lesions. , 1990, The Journal of investigative dermatology.

[8]  R. Locksley,et al.  Altered Immune Responses in Interleukin 10 Transgenic Mice , 1997, The Journal of experimental medicine.

[9]  D. Pardoll,et al.  The role of CD4+ T cell responses in antitumor immunity. , 1998, Current opinion in immunology.

[10]  J. McCluskey,et al.  Class II-restricted presentation of an endogenously derived immunodominant T-cell determinant of hen egg lysozyme. , 1991, Proceedings of the National Academy of Sciences of the United States of America.

[11]  S. Kimura,et al.  CD4+ T cells from peripheral blood of a melanoma patient recognize peptides derived from nonmutated tyrosinase. , 1998, Cancer research.

[12]  C. Y. Wang,et al.  Expression of Ia-like antigens on cultured human malignant melanoma cell lines. , 1978, Proceedings of the National Academy of Sciences of the United States of America.

[13]  A. Billiau,et al.  Immunomodulatory Properties of Interferon‐γ: An Update a , 1998 .

[14]  D. Ruiter,et al.  Level of HLA antigens in locoregional metastases and clinical course of the disease in patients with melanoma. , 1988, Cancer research.

[15]  J. D. de Vries,et al.  Interleukin-10 induces a long-term antigen-specific anergic state in human CD4+ T cells , 1996, The Journal of experimental medicine.

[16]  J. Kirkwood,et al.  Immune response to autologous human melanoma: implication of class I and II MHC products. , 1986, Biochimica et biophysica acta.

[17]  William Arbuthnot Sir Lane,et al.  Specificity and promiscuity among naturally processed peptides bound to HLA-DR alleles , 1993, The Journal of experimental medicine.

[18]  D. Ruiter,et al.  Immunohistochemical analysis of malignant melanomas and nevocellular nevi with monoclonal antibodies to distinct monomorphic determinants of HLA antigens. , 1984, Cancer Research.

[19]  Eric O Long,et al.  Processing pathways for presentation of cytosolic antigen to MHC class II-restricted T cells , 1992, Nature.

[20]  J S Lee,et al.  MHC class II-mediated antigen presentation by melanoma cells. , 1996, Journal of immunotherapy with emphasis on tumor immunology : official journal of the Society for Biological Therapy.

[21]  P. Marrack,et al.  Interleukin-induced increase in Ia expression by normal mouse B cells , 1984, The Journal of experimental medicine.

[22]  J. Ting,et al.  Major histocompatibility complex class II-transfected tumor cells present endogenous antigen and are potent inducers of tumor-specific immunity. , 1997, Proceedings of the National Academy of Sciences of the United States of America.

[23]  J. Woody,et al.  Human T-cell clones recognize chemically synthesized peptides of influenza haemagglutinin , 1982, Nature.

[24]  A. Rejthar,et al.  DNA content and MHC class II antigen expression in malignant melanoma: clinical course. , 1988, Journal of clinical pathology.

[25]  L. Lampson,et al.  Two populations of Ia-like molecules on a human B cell line. , 1980, Journal of immunology.

[26]  S. Ostrand-Rosenberg,et al.  MHC class II-transfected tumor cells directly present antigen to tumor-specific CD4+ T lymphocytes. , 1998, Journal of immunology.

[27]  A. Houghton,et al.  Light chain variants of an IgG3 anti-GD3 monoclonal antibody and the relationship among avidity, effector functions, tumor targeting, and antitumor activity. , 1990, Cancer research.

[28]  F. Brodsky,et al.  Class II molecules of the major histocompatibility complex considered as differentiation markers. , 1986, Human immunology.

[29]  J. Uhr,et al.  Increased expression of Ia antigens on resting B cells: an additional role for B-cell growth factor. , 1984, Proceedings of the National Academy of Sciences of the United States of America.

[30]  D. Ruiter,et al.  Differential expression of HLA-DR, DQ, and DP antigens in primary and metastatic melanoma. , 1988, The Journal of investigative dermatology.

[31]  R. Klausner,et al.  Class II MHC molecules can use the endogenous pathway of antigen presentation , 1990, Nature.

[32]  A. Billiau,et al.  Immunomodulatory properties of interferon-gamma. An update. , 1998, Annals of the New York Academy of Sciences.