Specific T helper cell requirement for optimal induction of cytotoxic T lymphocytes against major histocompatibility complex class II negative tumors

F Ossendorp, E Mengedé, M Camps, R Filius, C J Melief, F Ossendorp, E Mengedé, M Camps, R Filius, C J Melief

Abstract

This study shows that induction of tumor-specific CD4+ T cells by vaccination with a specific viral T helper epitope, contained within a synthetic peptide, results in protective immunity against major histocompatibility complex (MHC) class II negative, virus-induced tumor cells. Protection was also induced against sarcoma induction by acutely transforming retrovirus. In contrast, no protective immunity was induced by vaccination with an unrelated T helper epitope. By cytokine pattern analysis, the induced CD4+ T cells were of the T helper cell 1 type. The peptide-specific CD4+ T cells did not directly recognize the tumor cells, indicating involvement of cross-priming by tumor-associated antigen-presenting cells. The main effector cells responsible for tumor eradication were identified as CD8+ cytotoxic T cells that were found to recognize a recently described immunodominant viral gag-encoded cytotoxic T lymphocyte (CTL) epitope, which is unrelated to the viral env-encoded T helper peptide sequence. Simultaneous vaccination with the tumor-specific T helper and CTL epitopes resulted in strong synergistic protection. These results indicate the crucial role of T helper cells for optimal induction of protective immunity against MHC class II negative tumor cells. Protection is dependent on tumor-specific CTLs in this model system and requires cross-priming of tumor antigens by specialized antigen-presenting cells. Thus, tumor-specific T helper epitopes have to be included in the design of epitope-based vaccines.

Figures

Figure 1
Figure 1
T helper peptide vaccination protects mice against intraperitoneal tumor induction. C57BL/6 mice were vaccinated with synthetic peptide (100 μg/mouse) in a 50% PBS/IFA (vol/vol) emulsion subcutaneously at day −14 and challenged with 1,000 RMA cells intraperitoneally at day zero. The weights of the individual mice were monitored every other day and the percent weight increase is depicted. Each line is an individual mouse. (A) Six mice were injected with an emulsion of IFA with PBS without peptide as a negative control. (B) Six mice were vaccinated with the subdominant Kb-presented MuLV env–gp70 CTL epitope peptide env-Kb8 (SSWDFITV). (C) Seven mice were vaccinated with the I-Ab–presented 19-mer MuLV env–gp70 T helper epitope peptide env-H19 (EPLTSLTPRCNTAWNRLKL). (D) 10 mice were vaccinated with a mixture of env-Kb8 and env-H19 (both 100 μg) in a single vaccine depot.
Figure 2
Figure 2
(A) Tumor-specific T helper peptide vaccination increases long-term survival of mice challenged with tumor cells. C57BL/6 mice were vaccinated with synthetic peptides as described in the legend of Fig. 1. All groups received IFA (50% vol/vol) with or without 100 μg peptide subcutaneously. The percentage of surviving mice is depicted. Mice were killed when the weight increase was >25%. PBS, nonimmunized control (n = 6); SV-Kb9, nonrelated Sendai virus nucleoprotein Kb-presented CTL epitope (FAPGNYPAL; n = 6); env-Kb8, MuLV env–gp70 subdominant CTL epitope (n = 7); env-H19, 19-mer MuLV env–gp70 T helper epitope (n = 7); env-Kb8 + env-H19, mixture vaccine of MuLV CTLs and T helper epitopes (n = 10); OvaH, nonrelated ovalbumin I-Ab–presented T helper epitope (ISQAVHAAHAEINEAGR; n = 6); env-Kb8 + OvaH, mixture of MuLV subdominant CTL epitope and ovalbumin T helper epitope. Significance of differences (log rank test): PBS versus env-H19: P = 0.04; PBS versus env-Kb8 + env-H19: P = 0.0015; env-H19 versus env-Kb8 + env-H19: P = 0.28 (not significant; n.s.). (B) Combination of T helper peptide and the immunodominant gag-L CTL epitope induces highly efficient tumor protection. C57BL/6 mice were immunized with synthetic peptides in IFA subcutaneously (50 μg/mouse) at day −14 and challenged with 103 RMA tumor cells intraperitoneally. Long-term survival was monitored. gag-L-Db10, gag-L CTL epitope (LCCLCLTVFL); env-H13.3, env-gp70 T helper peptide (SLTPRCNTAWNRL). All groups: n = 10. Significance of differences (log rank test): PBS versus gag-L-Db10: P = 0.0078; PBS versus env-H13.3: P = 0.0078; PBS versus gag-L-Db10 + env-H13.3: P = 0.0001; gag-L-Db10 versus gag-L-Db10 + env-H13.3: P = 0.0108; env-H13.3 versus gag-L-Db10 + env-H13.3: P = 0.025. (C) Efficient protection by dominant CTL epitope requires specific help. Similar experiment using the 9-mer dominant CTL epitope gag-L–Db9 in combination with either the MuLV T helper peptide env-H13.3 or the nonrelated OvaH T helper peptide. All groups: n = 8. Significance of differences (log rank test): gag-L-Db9 versus gag-L-Db9 + OvaH: P = 0.169 (n.s.); gag-L-Db9 versus gag-L-Db9 + env-H13.3: P = 0.003; gag-L-Db9 + OvaH versus gag-L-Db9 + env-H13.3: P = 0.04; PBS versus gag-L-Db9 + env-H13.3: P = 0.0004.
Figure 3
Figure 3
T helper peptide vaccination protects mice against retrovirus-induced sarcoma. C57BL/6 mice were vaccinated at day −14 with (A) PBS-IFA subcutaneously (n = 7) and (B) 13-mer MuLV T helper peptide H13.3 (SLTPRCNTAWNRL), 10 μg/mouse in IFA subcutaneously (n = 7). At day zero mice were injected in the left thigh muscle with 103 focus-forming units of MoMSV. Mice were monitored for tumor size using a caliper measuring two perpendicular diameters. Each line represents one individual mouse.
Figure 4
Figure 4
Both CD4+ and CD8+ T cells are involved in the protective mechanism. C57BL/6 mice were treated with anti-CD4 (GK1.5) and anti-CD8 (2.43) monoclonal antibodies leading to selective depletion (>95%) of these T cell subsets. (A) Mice were vaccinated with 100 μg of H19 in IFA subcutaneously (day −14) and treated five times with 100 μg GK1.5 monoclonal antibody in 200 μl PBS intraperitoneally during the vaccination period (days −14, −13, −10, −7, and −4) or as a control with PBS. At day 0, 103 RMA cells were injected intraperitoneally. Significance of differences (log rank test): PBS versus env-H19: P = 0.0002; env-H19 + anti-CD4 versus env-H19: P = 0.0009; PBS versus env-H19 + anti-CD4: P = 0.577 (n.s.). (B) Mice were vaccinated with 10 μg T helper peptide H13.3 in IFA subcutaneously at day −14 and 103 RMA cells were injected intraperitoneally at day 0. Antibody treatment (three times with 100 μg of GK1.5 or 2.43 intraperitoneally) was done during the effector phase (days 0, 3, and 7). Significance of differences (log rank test): PBS versus env-H13.3: P = 0.0027; env-H13.3/anti-CD4 versus env-H13.3: P = 0.0013; env-H13.3/anti-CD8 versus env-H13.3: P = 0.0068.
Figure 5
Figure 5
MuLV-specific CD4+ T cells can be isolated from T helper peptide-immunized mice. CD4+ T cell clones were isolated from T helper peptide-immunized and RMA-challenged mice and tested in a 3-d proliferation assay. Values represent means of triplicate measurements. (A) CD4+ clone 3A12 was isolated from a spleen of a protected mouse, 2 mo after tumor challenge, that was previously immunized with the 11-mer T helper H11.1 peptide. Spleen cells were in vitro restimulated with mitomycin-treated and irradiated RMA tumor cells and CD4+ T cell clones were isolated by limiting dilution. Shaded bars from black to gray: 4 × 104, 2 × 104, 104, and 5 × 103 cells/well, respectively. env-H19 peptide was used at 10 μg/ml. SC, irradiated spleen cells (105/well). RMA cells were mitomycin treated, irradiated, and used at 2 × 104 cells/well with or without irradiated spleen cells. 771 is a B cell lymphoma cell line isolated from a tumor induced by the AKV type MCF1233 MuLV that lacks the T helper epitope sequence in env (2 × 104 irradiated cells/well). (B) Peptide fine specificity of CD4+ clone 3A12. Peptides were used at 5 μg/ml. Shaded bars from dark to light gray: 2 × 104, 104, and 5 × 103 cells/well, respectively. (C) Moloney MuLV specificity of clone 3A12. LPS B cell blasts were induced using C57BL/6 spleen cells in the presence or absence of Moloney MuLV (MoLV-LPS). After 3 d, the LPS blasts were isolated, irradiated, and used as stimulator cells (LPS, 105 cells/well) for clone 3A12 in the presence or absence of H19 peptide (10 μg/ml).
Figure 6
Figure 6
Cytotoxic T cells isolated from protected mice recognize the immunodominant MuLV gag-L peptide. CD8+ T cell bulk cultures were isolated from spleen cells of a C57BL/6 mouse that was immunized with the H11.1 T helper peptide and survived RMA tumor challenge. 2H9 was a representative clone isolated from this RMA-specific bulk culture by limiting dilution. (A) Cytotoxicity assay using RMA cells and HeLa cells selectively transfected with the MHC class I Db molecule as target cells. HeLaDb cells were incubated with the gag-L (CCLCLTVFL) peptide (1 μg/ml) during the assay. (B) No reactivity of 2H9 CTL clone with the T helper peptide sequence. EL-4 cells (H-2b) were incubated with different synthetic peptides (1 μg/ml). gag-L–Db9 is the dominant gag-L epitope CCLCLTVFL; gag-L–SIV9, another upstream gag-leader sequence reported to comprise a CTL epitope SIVLCCLCL (31).
Figure 6
Figure 6
Cytotoxic T cells isolated from protected mice recognize the immunodominant MuLV gag-L peptide. CD8+ T cell bulk cultures were isolated from spleen cells of a C57BL/6 mouse that was immunized with the H11.1 T helper peptide and survived RMA tumor challenge. 2H9 was a representative clone isolated from this RMA-specific bulk culture by limiting dilution. (A) Cytotoxicity assay using RMA cells and HeLa cells selectively transfected with the MHC class I Db molecule as target cells. HeLaDb cells were incubated with the gag-L (CCLCLTVFL) peptide (1 μg/ml) during the assay. (B) No reactivity of 2H9 CTL clone with the T helper peptide sequence. EL-4 cells (H-2b) were incubated with different synthetic peptides (1 μg/ml). gag-L–Db9 is the dominant gag-L epitope CCLCLTVFL; gag-L–SIV9, another upstream gag-leader sequence reported to comprise a CTL epitope SIVLCCLCL (31).

References

    1. Ostrand-Rosenberg S, Thakur A, Clements V. Rejection of mouse sarcoma cells after transfection of MHC class II genes. J Immunol. 1990;144:4068–4071.
    1. James R, Edwards S, Hui K, Bassett P, Grosveld F. The effect of class II gene transfection on the tumorigenicity of the H-2knegative mouse leukemia cell line K36.16. Immunology. 1991;72:213–218.
    1. Chen P, Aanathaswamy H. Rejection of K1735 murine melanoma in syngeneic hosts requires expression of MHC class I antigens and either class II antigens or IL-2. J Immunol. 1993;151:244–255.
    1. Yoshimura A, Shiku H, Nakayama E. Rejection of an IA+ variant line of FBL-3 leukemia by cytotoxic T lymphocytes with CD4+ and CD4−CD8−T cell receptor-αβ phenotypes generated in CD8-depleted C57BL/6 mice. J Immunol. 1993;150:4900–4910.
    1. Greenberg PD. Adoptive T cell therapy of tumors: mechanisms operative in the recognition and elimination of tumor cells. Adv Immunol. 1991;49:281–355.
    1. Klarnet JP, Kern DE, Okunu K, Holt C, Lilly F, Greenberg PD. FBL-reactive CD8 cytotoxic and CD4 helper T cells recognize distinct Friend murine leukemia virus–encoded antigens. J Exp Med. 1989;169:457–467.
    1. Hu J, Kindsvogel W, Busby S, Bailey MC, Shi Y, Greenberg PD. An evaluation of the potential to use tumor-associated antigens as targets for antitumor T cell therapy using transgenic mice expressing a retroviral tumor antigen in normal lymphoid tissues. J Exp Med. 1993;177:1681–1690.
    1. Lilly F, Boyse EA, Old LJ. Genetic basis of susceptibility to viral leukemogenesis. Lancet. 1964;ii:1207–1209.
    1. Vasmel WLJ, Zijlstra M, Radaszkiewicz T, Leupers CJM, De Goede REY, Melief CJM. Major histocompatibility class II regulated immunity to murine leukemia virus protects against early T- but not late B-cell lymphomas. J Virol. 1988;62:3156–3166.
    1. Chesebro B, Miyazawa B, Britt WJ. Host genetic control of spontaneous and induced immunity to Friend murine retrovirus infection. Annu Rev Immunol. 1990;8:477–499.
    1. Iwashiro M, Kondo T, Shimizu T, Yamagishi H, Takahashi K, Matsubayashi Y, Masuda T, Otaka A, Fujii N, Ishimoto A, et al. Multiplicity of virus-encoded helper T-cell epitopes expressed on FBL-3 tumor cells. J Virol. 1993;67:4533–4542.
    1. Vasmel WLE, Sijts EJAM, Leupers CJM, Matthews EA, Melief CJM. Primary virus–induced lymphomas evade T cell immunity by failure to express viral antigens. J Exp Med. 1989;169:1233–1254.
    1. Plata F, Langlade-Demoyen P, Abastado JP, Berbar T, Kourilsky P. Retrovirus antigens recognized by cytotoxic T lymphocytes activate tumor rejection in vivo. Cell. 1987;48:231–240.
    1. Sijts AJAM, De Bruijn ML, Ressing ME, Nieland JD, Mengedé EAM, Boog CJ, Ossendorp F, Kast WM, Melief CJM. Identification of an H-2 Kb–presented Moloney murine leukemia virus cytotoxic T lymphocyte epitope that displays enhanced recognition in H-2 Dbmutant bm13 mice. J Virol. 1994;68:6038–6046.
    1. Chen W, Qin H, Chesebro B, Cheever MA. Identification of a gag-encoded cytotoxic T lymphocyte epitope from FBL-3 leukemia shared by Friend, Moloney, and Rauscher murine leukemia virus–induced tumors. J Virol. 1996;70:7773–7782.
    1. Kast WM, Roux L, Curren J, Blom HJJ, Voordouw AC, Meloen RH, Kolakofski D, Melief CJM. Protection against lethal Sendai virus infection by in vivo priming of virus-specific cytotoxic T lymphocytes with an unbound peptide. Proc Natl Acad Sci USA. 1991;88:2283–2287.
    1. Schulz M, Zinkernagel RM, Hengartner H. Peptide-induced antiviral protection by cytotoxic T cells. Proc Natl Acad Sci USA. 1991;88:991–993.
    1. Feltkamp MCW, Smits HL, Vierboom MPM, Minnaar RP, De Jongh BM, Drijfhout JW, Ter J, Schegget, Melief CJM, Kast WM. Vaccination with a cytotoxic T lymphocyte epitope-containing peptide protects against a tumor induced by human Papillomavirus type 16-transformed cells. Eur J Immunol. 1993;23:2242–2249.
    1. Schild H, Norda M, Deres K, Falk K, Rötschke O, Wiesmüller K-H, Jung G, Rammensee H-G. Fine specificity of cytotoxic T lymphocytes primed in vivo either with virus or synthetic lipopeptide vaccine or primed in vitro with peptide. J Exp Med. 1991;174:1665–1668.
    1. Minev BR, McFarland BJ, Spiess PJ, Rosenberg SA, Restifo NP. Insertion signal sequence fused to minimal peptides elicits specific CD8+T-cell responses and prolongs survival of thymoma-bearing mice. Cancer Res. 1994;54:4155–4161.
    1. Mandelboim O, Vadai E, Fridkin M, Katz-Hillel A, Feldman M, Berke G, Eisenbach L. Regression of established murine carcinoma metastases following vaccination with tumour-associated antigen peptides. Nat Med. 1995;1:1179–1183.
    1. Mayordomo JI, Zorina T, Storkus WJ, Zitvogel L, Celluzzi C, Falo LD, Melief CJM, Ildstad ST, Kast WM, Deleo AB, Lotze MT. Bone marrow–derived dendritic cells pulsed with synthetic tumour peptides elicit protective and therapeutic antitumour immunity. Nat Med. 1995;1:1297–1302.
    1. Sijts AJAM, Ossendorp F, Mengedé EAM, van den Elsen PJ, Melief CJM. Immunodominant mink cell focus-inducing murine leukemia virus (MuLV)-encoded CTL epitope, identified by its MHC class I binding motif, explains MuLV-type specificity of MCF-directed cytotoxic T lymphocytes. J Immunol. 1994;152:106–116.
    1. Ljunggren H-G, Kärre K. Host resistance directed selectively against H-2 deficient lymphoma variants. J Exp Med. 1985;162:1745–1759.
    1. Stukart MJ, Vos A, Melief CJM. Cytotoxic T cell response against lymphoblasts infected with Moloney (Abelson) murine leukemia virus. Methodological aspects and H-2 requirements. Eur J Immunol. 1981;11:251–257.
    1. Gausepohl, H., M. Kraft, C. Boulin, and R.W. Frank. 1990. Automated multiple peptide synthesis with BOP activation. In Proc. of the 11th American Peptide Symposium. J.E. Rivier and G.R. Marshall, editors. ESCOM, Leiden. 1003–1004.
    1. Shimonkevitz R, Colon S, Kappler JW, Marrack P, Grey HM. Antigen recognition by H-2–restricted T cells. II. A tryptic ovalbumin peptide that substitutes for processed antigen. J Immunol. 1984;133:2067–2074.
    1. Bouaboula M, Legoux P, Pességué B, Delpech B, Dumont X, Piechaczyk M, Casellas P, Shire D. Quantitation of cytokine gene expression using a polymerase chain reaction method involving co-amplification with an internal multi-specific control. J Biol Chem. 1992;267:21830–21838.
    1. Ossendorp F, Eggers M, Neisig A, Ruppert T, Groettrup M, Sijts A, Mengedé E, Kloetzel P, Neefjes J, Koszinowski U, Melief C. A single residue exchange within a viral CTL epitope alters proteasome-mediated degradation resulting in lack of antigen presentation. Immunity. 1996;5:115–124.
    1. Shimizu T, Uenishi H, Teramura Y, Iwashiro M, Kuribayashi K, Tamamura H, Fujii N, Yamagishi H. Fine specificity of a virus-encoded helper T-cell epitope expressed on FBL-3 tumor cells. J Virol. 1994;68:7704–7708.
    1. Kondo T, Uenishi H, Shimizu T, Hirama T, Iwashiro M, Kuribayashi K, Tamamura H, Fujii N, Fujisawa R, Yamagishi H. A single retroviral gag precursor signal peptide recognized by FBL-3 tumor-specific cytotoxic T lymphocytes. J Virol. 1995;69:6735–6741.
    1. Alexander J, Sidney J, Southwood S, Ruppert J, Oseroff C, Maewal A, Snoke K, Serra HM, Kubo RT, Sette A, Grey H. Development of high potency universal DR-restricted helper epitopes by modification of high affinity DR-blocking peptides. Immunity. 1994;1:751–761.
    1. Keene JA, Forman J. Helper activity is required for the induction of cytotoxic T lymphocytes. J Exp Med. 1982;155:768–782.
    1. Gao X-M, Zheng B, Liew FY, Brett S, Tite J. Priming of influenza virus–specific cytotoxic T lymphocytes in vivo by short synthetic peptides. J Immunol. 1991;147:3168–3173.
    1. Fayolle C, Deriaud E, Leclerc C. In vivo induction of cytotoxic T cell response by a free synthetic peptide requires CD4+T cell help. J Immunol. 1991;147:4069–4073.
    1. Shirai M, Pendleton CD, Ahlers J, Takeshita T, Newman M, Berzofsky JA. Helper-cytotoxic T lymphocyte (CTL) determinant linkage required for priming of HIV CD8+CTL in vivo with peptide vaccine constructs. J Immunol. 1994;152:529–556.
    1. Bennett SRM, Carbone FR, Karamalis F, Miller JFAP, Heath WR. Induction of a CD8+ cytotoxic T lymphocyte response by cross-priming requires cognate CD4+T cell help. J Exp Med. 1997;186:65–70.
    1. Hasenkrug KJ, Brooks DM, Nishio J, Chesebro B. Differing T-cell requirements for recombinant retrovirus vaccines. J Virol. 1996;70:368–372.
    1. Miyazawa M, Fujisawa R, Ishihara C, Takei YA, Shimizu T, Uenishi H, Yamagishi H, Kuribayashi K. Immunization with a single T helper cell epitope abrogates Friend virus–induced early erythroid proliferation and prevents late leukemia development. J Immunol. 1995;155:748–758.
    1. Aichele P, Brduscha-Riem K, Zinkernagel RM, Hengartner H, Pircher H. T cell priming versus T cell tolerance induced by synthetic peptides. J Exp Med. 1995;182:261–266.
    1. Toes REM, Blom RJJ, Offringa R, Kast WM, Melief CJM. Functional deletion of tumor-specific cytotoxic T lymphocytes induced by peptide immunization can lead to the inability to reject tumors. J Immunol. 1996;156:3911–3918.
    1. Toes REM, Offringa R, Blom RJJ, Melief CJM, Kast WM. Peptide vaccination can lead to enhanced tumor outgrowth through specific T-cell tolerance induction. Proc Natl Acad Sci USA. 1996;93:7855–7860.
    1. Kearney ER, Pape KA, Loh DY, Jenkins MK. Visualization of peptide induced T cell immunity and peripheral tolerance induction in vivo. Immunity. 1994;1:327–339.
    1. Matloubian M, Concepcion RJ, Ahmed R. CD4+ T cells are required to sustain CD8+ cytotoxic T-cell responses during chronic viral infection. J Virol. 1994;68:8056–8063.
    1. Von Herrath MG, Yokoyama M, Dockter J, Oldstone MBA, Whitton JL. CD4-deficient mice have reduced levels of memory cytotoxic T lymphocytes after immunization and show diminished resistance to subsequent virus challenge. J Virol. 1996;70:1072–1079.
    1. Cardin RD, Brooks JW, Sarawar SR, Doherty PC. Progressive loss of CD8+ T cell-mediated control of a γ-herpesvirus in the absence of CD4+T cells. J Exp Med. 1996;184:863–871.

Source: PubMed

3
Subscribe