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

A van Elsas, A A Hurwitz, J P Allison, A van Elsas, A A Hurwitz, J P Allison

Abstract

We examined the effectiveness of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade, alone or in combination with a granulocyte/macrophage colony-stimulating factor (GM-CSF)-expressing tumor cell vaccine, on rejection of the highly tumorigenic, poorly immunogenic murine melanoma B16-BL6. Recently established tumors could be eradicated in 80% (68/85) of the cases using combination treatment, whereas each treatment by itself showed little or no effect. Tumor rejection was dependent on CD8(+) and NK1.1(+) cells but occurred irrespective of the presence of CD4(+) T cells. Mice surviving a primary challenge rejected a secondary challenge with B16-BL6 or the parental B16-F0 line. The same treatment regimen was found to be therapeutically effective against outgrowth of preestablished B16-F10 lung metastases, inducing long-term survival. Of all mice surviving B16-BL6 or B16-F10 tumors after combination treatment, 56% (38/68) developed depigmentation, starting at the site of vaccination or challenge and in most cases progressing to distant locations. Depigmentation was found to occur in CD4-depleted mice, strongly suggesting that the effect was mediated by CTLs. This study shows that CTLA-4 blockade provides a powerful tool to enhance T cell activation and memory against a poorly immunogenic spontaneous murine tumor and that this may involve recruitment of autoreactive T cells.

Figures

Figure 1
Figure 1
Successful treatment of preestablished B16-BL6 using anti–CTLA-4 and GM-CSF–producing BL6 vaccine. C57BL/6 female mice (five per group) were injected with 104 B16-BL6 cells subcutaneously on the back, on the same day (A) or 4, 8, or 12 d (B–D) before treatment was started. Treatment consisted of three consecutive injections (in a 6-d time frame as indicated in Materials and Methods) of anti–CTLA-4 antibody 9H10 intraperitoneally (•), control hamster IgG (100, 50, 50 μg; ○), or 106 irradiated BL6/g cells subcutaneously, in combination with 9H10 (▪) or hamster IgG (□). Tumor growth (mm2) was scored by measuring perpendicular diameters and was averaged for all mice within each group. In some treatment groups, only a fraction of the mice (indicated between brackets) developed a tumor.
Figure 2
Figure 2
A single dose of GM-CSF–producing vaccine cooperates with CTLA-4 blockade to induce 100% cure of B16-BL6. Mice were inoculated subcutaneously with 104 B16-BL6 cells. On the same day, combination treatment was initiated using triple BL6/g vaccine (days 0, 3, and 6) combined with either hamster IgG (100, 50, 50 μg on days 3, 6, and 9; □) or anti–CTLA-4 (▪). Control treatments consisted of antibody injections alone: hamster IgG (○) or anti–CTLA-4 (•). Also, anti–CTLA-4 treatment was combined with a single (▴) or double injection (♦) of the BL6/g vaccine. Average tumor size was calculated for all mice within a treatment group (mm2). The fraction of mice developing tumors is shown between brackets.
Figure 3
Figure 3
Anti–CTLA-4 enhances IFN-γ production by B16-specific T cells induced in vivo. Mice (four per group) were vaccinated with irradiated BL6/g (106 per mouse) and cotreated with control hamster IgG (A) or anti–CTLA-4 (B). After 4 wk, mice were challenged with 2 × 104 B16-BL6, and 10 d later, splenocytes were pooled and restimulated in vitro using B16-BL6/B7.1 (open bars) or a mixture of B16-F10 and DC2.4 dendritic cells (filled bars). On day 8, cultures were tested for tumor-specific IFN-γ release as described in Materials and Methods. Targets included B16 sublines -F0, -F10, and -BL6, as well as unrelated H-2b tumors EL4 and MC38.
Figure 3
Figure 3
Anti–CTLA-4 enhances IFN-γ production by B16-specific T cells induced in vivo. Mice (four per group) were vaccinated with irradiated BL6/g (106 per mouse) and cotreated with control hamster IgG (A) or anti–CTLA-4 (B). After 4 wk, mice were challenged with 2 × 104 B16-BL6, and 10 d later, splenocytes were pooled and restimulated in vitro using B16-BL6/B7.1 (open bars) or a mixture of B16-F10 and DC2.4 dendritic cells (filled bars). On day 8, cultures were tested for tumor-specific IFN-γ release as described in Materials and Methods. Targets included B16 sublines -F0, -F10, and -BL6, as well as unrelated H-2b tumors EL4 and MC38.
Figure 4
Figure 4
Mice bearing B16-F10 lung metastases show enhanced survival when treated with anti–CTLA-4 and F10/g vaccine. B16-F10 cells (5 × 104 per mouse) were injected into the tail vein and 24 h later, treatment was started using control hamster IgG (10 mice, ○), anti–CTLA-4 antibody 9H10 (9 mice; •), irradiated F10/g (106 subcutaneously) in combination with hamster IgG (10 mice; □) or 9H10 (13 mice; ▪) on days 1, 4, and 7, according to the dosing schedule used for subcutaneous tumors (see Fig. 1 legend). Mice were followed for survival, and in some subjects death due to extensive pulmonary metastasis was confirmed by harvesting lungs postmortem.
Figure 5
Figure 5
B16-F10 metastases demonstrate lymphocytic infiltration after treatment with anti–CTLA-4 and F10/g vaccine. Mice injected with 105 B16-F10 intravenously and treated with control hamster IgG (A), 9H10 (B), or F10/g vaccine in combination with either hamster IgG (C) or 9H10 (D) on days 1, 4, and 7, as outlined in the Fig. 4 legend. On day 25, lungs were harvested, fixed in 10% neutral-buffered formalin, and processed for hematoxylin–eosin staining.
Figure 6
Figure 6
Rejection of B16-BL6 or B16-F10 as a result of treatment with anti–CTLA-4 and GM-CSF–producing vaccines causes autoimmune skin and hair depigmentation. After successful treatment for B16-BL6 subcutaneously or B16-F10 intravenously, C57Bl/6 mice developed skin and hair depigmentation. (A) Depigmentation of both sites of vaccination and challenge, after rejection of a day 0 tumor. (B) Progressive depigmentation found in a mouse rejecting a B16-BL6 subcutaneous tumor, established 8 d before treatment started. (C) Depigmentation at the site of vaccination of a mouse cured from preestablished B16-F10 lung metastases.
Figure 6
Figure 6
Rejection of B16-BL6 or B16-F10 as a result of treatment with anti–CTLA-4 and GM-CSF–producing vaccines causes autoimmune skin and hair depigmentation. After successful treatment for B16-BL6 subcutaneously or B16-F10 intravenously, C57Bl/6 mice developed skin and hair depigmentation. (A) Depigmentation of both sites of vaccination and challenge, after rejection of a day 0 tumor. (B) Progressive depigmentation found in a mouse rejecting a B16-BL6 subcutaneous tumor, established 8 d before treatment started. (C) Depigmentation at the site of vaccination of a mouse cured from preestablished B16-F10 lung metastases.
Figure 6
Figure 6
Rejection of B16-BL6 or B16-F10 as a result of treatment with anti–CTLA-4 and GM-CSF–producing vaccines causes autoimmune skin and hair depigmentation. After successful treatment for B16-BL6 subcutaneously or B16-F10 intravenously, C57Bl/6 mice developed skin and hair depigmentation. (A) Depigmentation of both sites of vaccination and challenge, after rejection of a day 0 tumor. (B) Progressive depigmentation found in a mouse rejecting a B16-BL6 subcutaneous tumor, established 8 d before treatment started. (C) Depigmentation at the site of vaccination of a mouse cured from preestablished B16-F10 lung metastases.

References

    1. Boon T., Cerottini J.C., Van den Eynde B., van der Bruggen P., Van Pel A. Tumor antigens recognized by T lymphocytes. Annu. Rev. Immunol. 1994;12:337–365.
    1. Rosenberg S.A. Cancer vaccines based on the identification of genes encoding cancer regression antigens. Immunol. Today. 1997;18:175–182.
    1. Liu G.Y., Fairchild P.J., Smith R.M., Prowle J.R., Kioussis D., Wraith D.C. Low avidity recognition of self-antigen by T cells permits escape from central tolerance. Immunity. 1995;3:407–415.
    1. Poplonski L.B., Vukusic B., Pawling J., Clapoff S., Roder J., Hozumi N., Whither J. Tolerance is overcome in beef insulin-transgenic mice by activation of low-affinity autoreactive T cells. Eur. J. Immunol. 1996;26:601–606.
    1. Morgan D.J., Kreuwel H.T.C., Fleck S., Levitsky H.I., Pardoll D.M., Sherman L.A. Activation of low avidity CTL for a self-epitope results in tumor rejection but not autoimmunity. J. Immunol. 1998;160:643–651.
    1. Overwijk W.W., Tsung A., Irvine K.E., Parkhurst M.R., Goletz T.J., Tsung K., Carroll M.W., Liu C., Moss B., Rosenberg S.A. gp100/pmel 17 is a murine tumor rejection antigeninduction of “self”-reactive tumoricidal T cells using high-affinity, altered peptide ligand. J. Exp. Med. 1998;188:277–286.
    1. Ehl S., Hombach J., Aichele P., Rulicke T., Odermatt B., Hengartner H., Zinkernagel R., Pircher H. Viral and bacterial infections interfere with peripheral tolerance induction and activate CD8+ T cells to cause immunopathology. J. Exp. Med. 1998;187:763–774.
    1. Allison J.P. CD28-B7 interactions in T-cell activation. Curr. Opin. Immunol. 1994;6:414–419.
    1. Schwartz R.H. A cell culture model for T lymphocyte clonal anergy. Science. 1990;248:1349–1356.
    1. Townsend S., Allison J.P. Tumor rejection after direct costimulation of CD8+ T cells by B7-transfected melanoma cells. Science. 1993;259:368–370.
    1. Chen L., Ashe S., Brady W.A., Hellstrom I., Hellstrom K.E., Ledbetter J.A., McGowan P., Linsley P.S. Costimulation of antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4. Cell. 1992;71:1093–1102.
    1. Thompson C.B., Allison J.P. The emerging role of CTLA-4 as an immune attenuator. Immunity. 1997;7:445–450.
    1. Perez V.L., Van Parijs L., Biuckians A., Zheng X.X., Strom T.B., Abbas A.K. Induction of peripheral T cell tolerance in vivo requires CTLA-4 engagement. Immunity. 1997;6:411–417.
    1. Leach D., Krummel M., Allison J.P. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271:1734–1736.
    1. Hurwitz, A.A., A. van Elsas, D.R. Leach, J. Ziskin, J. Villasenor, T. Truong, and J.P. Allison. 1999. Manipulation of T cell activation to generate anti-tumor CTL. In Cytotoxic Cells: Basic Mechanisms and Medical Applications. M.V. Sitkovsky and P.A. Henkart, editors. Lippincott, Philadelphia. In press.
    1. Kwon E.D., Hurwitz A.A., Foster B.A., Madias C., Feldhaus A.L., Greenberg N.M., Burg M.B., Allison J.P. Manipulation of T cell costimulatory and inhibitory signals for immunotherapy of prostate cancer. Proc. Natl. Acad. Sci. USA. 1997;94:8099–8103.
    1. Hurwitz A.A., Yu T.F., Leach D.R., Allison J.P. CTLA-4 blockade synergizes with tumor-derived GM-CSF for treatment of an experimental mammary carcinoma. Proc. Natl. Acad. Sci. USA. 1998;95:10067–10071.
    1. Krummel M.F., Allison J.P. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J. Exp. Med. 1995;182:459–465.
    1. Shen Z., Reznikoff G., Dranoff G., Rock K.L. Cloned dendritic cells can present exogenous antigens on both MHC class I and class II molecules. J. Immunol. 1997;158:2723–2730.
    1. Dranoff G., Jaffee E., Lazenby A., Golumbek P., Levitsky H., Brose K., Jackson V., Hamada H., Pardoll D., Mulligan R.C. Vaccination with irradiated tumor cells engineered to secrete GM-CSF stimulates potent, specific, and long lasting anti-tumor immunity. Proc. Natl. Acad. Sci. USA. 1993;90:3539–3543.
    1. Hart I.R. The selection of characterization of an invasive variant of the B16 melanoma. Am. J. Pathol. 1979;97:587–600.
    1. Hung K., Hayashi R., Lafond-Walker A., Lowenstein C., Pardoll D., Levitsky H. The central role of CD4+ T cells in the antitumor immune response. J. Exp. Med. 1998;188:2357–2368.
    1. Inaba K., Inaba M., Romani N., Aya H., Deguchi M., Ikehara S., Muramatsu S., Steinman R.M. Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J. Exp. Med. 1992;176:1693–1702.
    1. Huang A.Y., Golumbek P., Ahmadzadeh M., Jaffee E., Pardoll D., Levitsky H. Role of bone marrow-derived cells in presenting MHC class I-restricted tumor antigens. Science. 1994;264:961–965.
    1. Banchereau J., Steinman R.M. Dendritic cells and the control of immunity. Nature. 1998;392:245–252.
    1. Yang Y., Zou J., Mu J., Wijesuriya R., Ono S., Walunas T., Bluestone J., Fujiwara H., Hamaoka T. Enhanced induction of antitumor T-Cell responses by cytotoxic T lymphocyte associated molecule-4 blockadethe effect is manifested only at the restricted tumor-bearing stages. Cancer Res. 1997;57:4036–4041.
    1. Staveley-O'Carroll K., Sotomayor E., Montgomery J., Borrello I., Hwang L., Fein S., Pardoll D., Levitsky H. Induction of antigen-specific T cell anergyan early event in the course of tumor progression. Proc. Natl. Acad. Sci. USA. 1998;95:1178–1183.
    1. Sotomoyer E.M., Borrello I.M., Tubb E., Allison J.P., Levitsky H.I. In vivo blockade of CTLA-4 enhances the priming of responsive T-cells, but fails to prevent the induction of tumor antigen-specific tolerance. Proc. Natl. Acad. Sci. USA. 1999;In press
    1. Levitsky H.I., Lazenby A., Hayashi R.J., Pardoll D.M. In vivo priming of two distinct antitumor effector populationsthe role of MHC class I expression. J. Exp. Med. 1994;179:1215–1224.
    1. Wu T.C., Huang A.Y.C., Jaffee E.M., Pardoll D.M. A reassessment of the role of B7-1 expression in tumor rejection. J. Exp. Med. 1995;182:1–7.
    1. McCoy K.D., Hermans I.F., Fraser J.H., Le Gros G., Ronchese F. Cytotoxic T lymphocyte–associated antigen 4 (CTLA-4) can regulate dendritic cell–induced activation and cytotoxicity of CD8+ T cells independently of CD4+ T cell help. J. Exp. Med. 1999;189:1157–1162.
    1. Schoenberger S.P., Toes R.E., van der Voort E.I., Offringa R., Melief C.J. T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Nature. 1998;393:480–483.
    1. Bennett S.R., Carbone F.R., Karamalis F., Flavell R.A., Miller J.F., Heath W.R. Help for cytotoxic-T-cell responses is mediated by CD40 signalling. Nature. 1998;393:478–480.
    1. Ridge J.P., Di Rosa F., Matzinger P. A conditioned dendritic cell can be a temporal bridge between a CD4+ T-helper and a T-killer cell. Nature. 1998;393:474–478.
    1. Richards J.M., Mehta N., Ramming K., Skosey P. Sequential chemoimmunotherapy in the treatment of metastatic melanoma. J. Clin. Oncol. 1992;10:1338–1343.
    1. Rosenberg S.A., White D.E. Vitiligo in patients with melanomanormal tissue antigens can be targets for cancer immunotherapy. J. Immunother. Emphasis Tumor Immunol. 1996;19:81–84.
    1. Okamoto T., Irie R.F., Fujii S., Huang S.K., Nizze A.J., Morton D.L., Hoon D.S. Anti-tyrosinase-related protein-2 immune response in vitiligo patients and melanoma patients receiving active-specific immunotherapy. J. Invest. Dermatol. 1998;111:1034–1039.
    1. Ogg G.S., Rod Dunbar P., Romero P., Chen J.L., Cerundolo V. High frequency of skin-homing melanocyte-specific cytotoxic T lymphocytes in autoimmune vitiligo. J. Exp. Med. 1998;188:1203–1208.
    1. Kawakami Y., Eliyahu S., Delgado C.H., Robbins P.F., Sakaguch K., Appella E., Yannelli J.R., Adema G., Miki T., Rosenberg S.A. Identification of a human melanoma antigen recognized by tumor-infiltrating lymphocytes associated with in vivo tumor rejection. Proc. Natl. Acad. Sci. USA. 1994;91:6458–6462.
    1. Bloom M.B., Perry-Lalley D., Robbins P.F., Li Y., el-Gamil M., Rosenberg S.A., Yang J.C. Identification of tyrosinase-related protein 2 as a tumor rejection antigen for the B16 melanoma. J. Exp. Med. 1997;185:453–459.
    1. Naftzger C., Takechi Y., Kohda H., Hara I., Vijayasaradhi S., Houghton A.N. Immune response to a differentiation antigen induced by altered antigena study of tumor rejection and autoimmunity. Proc. Natl. Acad. Sci. USA. 1996;93:14809–14814.
    1. Hara I., Takechi Y., Houghton A.N. Implicating a role for immune recognition of self in tumor rejectionpassive immunization against the brown locus protein. J. Exp. Med. 1995;182:1609–1614.
    1. Overwijk W.W., Lee D.S., Surman D.R., Irvine D.R., Touloukian C.E., Shan C.-C., Carroll M.W., Moss B., Rosenberg S.A., Restifo N.P. Vaccination with a recombinant vaccinia virus encoding a “self” antigen induces autoimmune vitiligo and tumor cell destruction in micerequirement for CD4+ T lymphocytes. Proc. Natl. Acad. Sci. USA. 1999;96:2982–2987.
    1. Weber L.W., Bowne W.B., Wolchok J.D., Srinivasan R., Qin J., Moroi Y., Clynes R., Song P., Lewis J.J., Houghton A.N. Tumor immunity and autoimmunity induced by immunization with homologous DNA. J. Clin. Invest. 1998;102:1258–1264.

Source: PubMed

3
Abonner