Identification of immunogenic LY6K long peptide encompassing both CD4+ and CD8+ T-cell epitopes and eliciting CD4+ T-cell immunity in patients with malignant disease

Yusuke Tomita, Akira Yuno, Hirotake Tsukamoto, Satoru Senju, Yasuhiro Kuroda, Masatoshi Hirayama, Yuya Imamura, Junji Yatsuda, Mohammad Abu Sayem, Atsushi Irie, Akinobu Hamada, Hirofumi Jono, Koji Yoshida, Takuya Tsunoda, Yataro Daigo, Hirotsugu Kohrogi, Yoshihiro Yoshitake, Yusuke Nakamura, Masanori Shinohara, Yasuharu Nishimura, Yusuke Tomita, Akira Yuno, Hirotake Tsukamoto, Satoru Senju, Yasuhiro Kuroda, Masatoshi Hirayama, Yuya Imamura, Junji Yatsuda, Mohammad Abu Sayem, Atsushi Irie, Akinobu Hamada, Hirofumi Jono, Koji Yoshida, Takuya Tsunoda, Yataro Daigo, Hirotsugu Kohrogi, Yoshihiro Yoshitake, Yusuke Nakamura, Masanori Shinohara, Yasuharu Nishimura

Abstract

Identification of peptides that activate both tumor-specific helper T (Th) cells and cytotoxic T lymphocytes (CTLs) are important for the induction of effective antitumor immune responses. We focused on a long peptide (LP) derived from lymphocyte antigen 6 complex locus K (LY6K) encompassing both candidate Th epitopes and a known CTL epitope. Using IFNγ ELISPOT assays as a marker of activated T cells, we studied the immunogenicity and cross-priming potential of LY6K-LP, assaying human immune cell responses in vitro and immunologic activities in HLA-A24 transgenic mice in vivo. We identified LY6K172-191-LP as an effective immunogen spanning naturally processed epitopes recognized by T helper type 1 (Th1) cells and CTLs. LY6K-specific CTLs were induced through cross-presentation of LY6K172-191-LP in vitro and in vivo. In addition, LY6K172-191-LP enhanced induction of LY6K-specific CTLs among the peripheral blood mononuclear cells (PBMCs) of head-and-neck malignant tumor (HNMT) patients. LY6K172-191-LP-specific Th1 immunologic response following 1 week in vitro stimulation of PBMCs with LY6K172-191-LP were detected in 16 of 21 HNMT patients (76%) vaccinated with CTL-epitope peptides and 1 of 11 HNMT patients (9%) prior to vaccination, but not in 9 healthy donors. Our results are the first to demonstrate the presence of LY6K-specific Th1 cell responses in HNMT patients and underscore the possible utility of LY6K172-191-LP for the induction and propagation of both LY6K-specific Th1 cells and CTLs.

Keywords: CTL epitope; Th1 cells; cancer testis antigen; cross-presentation; cross-priming; head and neck cancer; helper T-cell epitope; immunotherapy; lymphocyte antigen 6 complex locus K.

Figures

https://www.ncbi.nlm.nih.gov/pmc/articles/instance/4203508/bin/onci-3-e28100-g1.jpg
Figure 1. Induction of LY6K-specific helper T cells from healthy donors. (A–F) LY6K-specific helper T (Th) cells were generated from healthy donors (HDs) by stimulating purified CD4+ cells with LY6K119–142-LP or LY6K172–191-LP, as indicated. The number of IFNγ-producing Th cells was analyzed by ELISPOT assay. Data are presented as the mean ± SD of triplicate assays. The HLA class-II genotype is indicated above the panels. The underlined HLA-Class II allele encodes the element presenting the peptides to Th cells. (A) Induction of HLA-DP5-restricted LY6K119–142-LP-specific Th cells in donor HD1. Data are from at least 3 independent experiments with representative results shown. The Th cells were restimulated with autologous peripheral blood mononuclear cells (PBMCs; left panel) or L-cells (engineered to express the indicated HLA molecule) pulsed with LY6K119–142-LP antigen presenting cells (APCs; right panel). An Epstein‒Barr virus-derived nuclear antigen (EBNA-DP5 LP) was used as a control LP. B-F. Induction of HLA-restricted LY6K119–142-LP (B and C) or LY6K172–191-LP (D–F) specific Th cells, similar to (A) (above). (B) Induction of HLA-DR8-restricted LY6K119–142-LP-specific Th cells in donor HD2. (C) Induction of HLA-DR15-restricted LY6K119–142-LP-specific Th cells in donor HD3. (D and E) HLA-DR15-restricted LY6K172–191-LP-specific Th cells were generated from HD2 and HD3. (F). HLA-DQ-restricted LY6K172–191-LP-specific Th cells were generated from a DR15-negative HD4. LP, long peptide.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/4203508/bin/onci-3-e28100-g2.jpg
Figure 2. LY6K-LPs encompass naturally processed Th-cell epitopes. (A and B). The number of IFNγ-producing healthy donor (HD) derived helper T (Th) cells stimulated by antigen-presenting dendritic cells (DCs) loaded with recombinant full-length lymphocyte antigen 6 complex locus K (LY6K) protein was analyzed by ELISPOT assay. (A) HLA-DP5-restricted LY6K119–142-LP-specific bulk helper T (Th) cells established from HD1 specifically recognized autologous DCs loaded with recombinant LY6K protein (left panel). Representative data are shown from 3 independent experiments with similar results achieved. (B) HLA-DR15 (left) or HLA-DQ (right)-restricted LY6K172–191-LP-specific Th-clone (from HD2 and HD4, respectively) recognized autologous DCs loaded with a recombinant LY6K protein. Representative data are shown from 5 independent experiments with similar results achieved. The cognate LY6K-LPs were used as positive controls in the IFNγ ELISPOT assays. APC, antigen-presenting cells; LP, long peptide.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/4203508/bin/onci-3-e28100-g3.jpg
Figure 3. Presence of LY6K-LPs-specific Th1 cells in the peripheral blood of LY6K177–186-A24 SP vaccinated HNMT patients. (A–D). Fresh peripheral blood mononuclear cells (PBMCs) derived from patients vaccinated with LY6K177–186-A24 SP (refer to Table 1) were stimulated with a mixture of LY6K119–142-LP and LY6K172–191-LP plus IL-2 and IL-7 in vitro. After 1 wk, the frequency of LY6K-LP-specific T cells was detected by IFNγ ELISPOT assay. (B and C) LY6K-LP-specific-Th1 cell responses were assessed in 23 HNMT patients. Samples were tested from 21 HNMT patients vaccinated with LY6K177–186-A24 SP (After Vac.), 11 HNMT patients prior to vaccination (Before Vac.), and 9 healthy donors as controls. (B) Th-cell responses to the indicated LY6K-LP as IFNγ spots per 105 cells detected after background subtraction. Each dot represents an individual donor. Horizontal lines denote median values, and P values represent statistical results from a nonparametric Mann‒Whitney U test. n.s., not significant. (C) Column graph showing proportion of patients and healthy donors responding to LY6K172–191-LP (from B) prior to and after vaccination. P values were calculated using Fisher’s exact probability test. (D) HLA class II-restriction of LY6K-LP-specific Th1 cells. Fresh PBMCs stimulated with LY6K-LPs for 1 wk were re-stimulated with each LY6K-LP (LY6K119–142-LP or LY6K172–191-LP, as indicated) in the presence (or absence, first column) of monoclonal antibodies (mAbs) specific to HLA-DR, -DP, -DQ class II or HLA-Class I. SP, short peptide; LP, long peptide.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/4203508/bin/onci-3-e28100-g4.jpg
Figure 4. LY6K172–191-LP induces efficient expansion of LY6K177–186-A24 SP-specific CD8+ T-cells in vitro and in vivo. (A and B) Autologous dendritic cells (DCs) uptake and cross-present LY6K172–191-LP to LY6K177–186-A24 SP-specific CD8+ cytotoxic T lymphocytes (CTLs) derived from healthy donor (HD3). (A) Unfixed or fixed DCs were pulsed for 3 h with LY6K172–191-LP or LY6K177–186-A24 SP. Bulk HD3-derived LY6K177–186-A24 SP-specific CTLs were co-cultured with the pulsed DCs for 6 h and responses were measured by intracellular IFNγ staining and flow cytometry. Representative data from 3 independent experiments with similar results are shown. (B) HD3-derived LY6K177–186-A24 SP-specific bulk CTLs were stimulated with LY6K172–191-LP (middle panel) or irrelevant LP (right panel)-pulsed autologous DCs in vitro. The peptide treated cells were stained with a HLA-A24/LY6K177–186-specific tetramer in combination with an anti-CD8 mAb at the indicated time points. A pre-treatment (d0) control is shown in the left panel. Stained cells were analyzed by flow cytometry gating on CD8+ T cells. Representative data are from 3 independent experiments with similar results achieved. (C and D) LY6K172–191-LP expands LY6K177–186-A24 SP-specific CTLs derived from vaccinated HNMT patients. Fresh PBMCs from the HNMT patient (HNMT108) vaccinated with LY6K177–186-A24 SP were cultured with LY6K172–191-LP. On day 0 (ex vivo) and day 7, the PBMCs were stained with a HLA-A24/LY6K177–186-specific tetramer or control tetramer and analyzed by flow cytometry gating on CD8+ T cells. (C) Dot plot (left) of tetramer+ CD8+ T cells . On day 7, the frequency of IFNγ-expressing LY6K177–186-A24 SP-specific CTLs was also detected by ELISPOT assay (bar graph). Representative data from 4 vaccinated HNMT patients (refer to Table 1, HNMT43, 105, 108, and 110) with similar results achieved. (D) The increases (fold increase) in proportion of CD8+ tetramer+ cells (from C) are shown. E. LY6K172–191-LP cross-priming of CTLs in vitro. PBMCs obtained from HD3 were incubated with LY6K172–191-LP for 2 wk and LY6K172–191-LP was added on d0 and d7. Cells were harvested d14 and stained with a HLA-A24/LY6K177–186-specific tetramer. Representative data are from 3 independent experiments (both technical and biological replicates) with similar results achieved. (F) Induction of LY6K177–186-A24 SP-specific CTLs in mice immunized with LY6K172–191-LP. HLA-A24-expressing transgenic mice were immunized with LY6K172–191-LP. After the third vaccination with LY6K172–191-LP, murine CD8+ T-cells in the inguinal lymph nodes were stimulated with bone marrow-derived DCs pulsed with LY6K177–186-A24 SP. The number of IFNγ-producing murine CD8+ T-cells was analyzed by ELISPOT. Representative data are from 8 independent experiments with similar results achieved. HNMT, head and neck malignant tumor; LP, long peptide; SP, short peptide.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/4203508/bin/onci-3-e28100-g5.jpg
Figure 5. The synergistic effect of LY6K-LPs on induction of LY6K177–186-A24 SP-specific CTLs. (A) LY6K119–142-LP-specific or LY6K172–191-LP-specific bulk CD4+ T cells and LY6K177–186-A24 SP-specific bulk CD8+ T cells derived from HLA-A24+/DR15+ HD3 were cultured with autologous DCs in the presence of LY6K177–186-A24 SP (SP) alone, LY6K177–186-A24 SP + Control LP (Control LP + SP), or LY6K177–186-A24 SP + LY6K-LP (LY6K-LP + SP) without cytokine for 1-wk. The cultured cells were then stained with a PE-labeled HLA-A24/LY6K177–186-specific tetramer and a FITC-labeled anti-CD8 antibody and analyzed by flow cytometry. “Pre-stimulation” indicates the absolute number of tetramer+ CD8+ T cells/well of LY6K177–186-A24 SP-specific bulk CD8+ T cells in the healthy donor line. Data are the mean ± SD of triplicate assays. Representative data shown are from 3 independent experiments with similar results achieved. (B) Fresh PBMCs obtained from HNMT43 vaccinated with LY6K177–186-A24 SP were plated in a 96-well, round-bottomed culture plate (1 × 105 cells/well) in 10 μg/mL LY6K177–186-A24 SP alone, LY6K177–186-A24 SP + control LP, LY6K177–186-A24 SP + LY6K119–142-LP or LY6K177–186-A24 SP + LY6K172–191-LP without addition of cytokines. On day 7, cells were stained with a HLA-A24/LY6K177–186-specific tetramer and anti-CD8 antibody and analyzed by flow cytometry. Representative HLA-A24/LY6K177–186-specific tetramer staining is shown (dot plots). (C) Absolute number of tetramer+ CD8+ cells/well. Data are the mean ± SD of triplicate assays. Representative data are from 3 independent experiments with similar results achieved.

References

    1. Ishikawa N, Takano A, Yasui W, Inai K, Nishimura H, Ito H, Miyagi Y, Nakayama H, Fujita M, Hosokawa M, et al. Cancer-testis antigen lymphocyte antigen 6 complex locus K is a serologic biomarker and a therapeutic target for lung and esophageal carcinomas. Cancer Res. 2007;67:11601–11. doi: 10.1158/0008-5472.CAN-07-3243.
    1. Suda T, Tsunoda T, Daigo Y, Nakamura Y, Tahara H. Identification of human leukocyte antigen-A24-restricted epitope peptides derived from gene products upregulated in lung and esophageal cancers as novel targets for immunotherapy. Cancer Sci. 2007;98:1803–8. doi: 10.1111/j.1349-7006.2007.00603.x.
    1. Mizukami Y, Kono K, Daigo Y, Takano A, Tsunoda T, Kawaguchi Y, Nakamura Y, Fujii H. Detection of novel cancer-testis antigen-specific T-cell responses in TIL, regional lymph nodes, and PBL in patients with esophageal squamous cell carcinoma. Cancer Sci. 2008;99:1448–54. doi: 10.1111/j.1349-7006.2008.00844.x.
    1. Suzuki H, Fukuhara M, Yamaura T, Mutoh S, Okabe N, Yaginuma H, Hasegawa T, Yonechi A, Osugi J, Hoshino M, et al. Multiple therapeutic peptide vaccines consisting of combined novel cancer testis antigens and anti-angiogenic peptides for patients with non-small cell lung cancer. J Transl Med. 2013;11:97. doi: 10.1186/1479-5876-11-97.
    1. Ishikawa H, Imano M, Shiraishi O, Yasuda A, Peng YF, Shinkai M, Yasuda T, Imamoto H, Shiozaki H. Phase I clinical trial of vaccination with LY6K-derived peptide in patients with advanced gastric cancer. Gastric Cancer. 2014;17:173–80. doi: 10.1007/s10120-013-0258-6.
    1. Kono K, Iinuma H, Akutsu Y, Tanaka H, Hayashi N, Uchikado Y, Noguchi T, Fujii H, Okinaka K, Fukushima R, et al. Multicenter, phase II clinical trial of cancer vaccination for advanced esophageal cancer with three peptides derived from novel cancer-testis antigens. J Transl Med. 2012;10:141. doi: 10.1186/1479-5876-10-141.
    1. Kono K, Mizukami Y, Daigo Y, Takano A, Masuda K, Yoshida K, Tsunoda T, Kawaguchi Y, Nakamura Y, Fujii H. Vaccination with multiple peptides derived from novel cancer-testis antigens can induce specific T-cell responses and clinical responses in advanced esophageal cancer. Cancer Sci. 2009;100:1502–9. doi: 10.1111/j.1349-7006.2009.01200.x.
    1. Iwahashi M, Katsuda M, Nakamori M, Nakamura M, Naka T, Ojima T, Iida T, Yamaue H. Vaccination with peptides derived from cancer-testis antigens in combination with CpG-7909 elicits strong specific CD8+ T cell response in patients with metastatic esophageal squamous cell carcinoma. Cancer Sci. 2010;101:2510–7. doi: 10.1111/j.1349-7006.2010.01732.x.
    1. Dobrzanski MJ. Expanding roles for CD4 T cells and their subpopulations in tumor immunity and therapy. Front Oncol. 2013;3:63. doi: 10.3389/fonc.2013.00063.
    1. Church SE, Jensen SM, Antony PA, Restifo NP, Fox BA. Tumor-specific CD4+ T cells maintain effector and memory tumor-specific CD8+ T cells. Eur J Immunol. 2014;44:69–79. doi: 10.1002/eji.201343718.
    1. Bos R, Sherman LA. CD4+ T-cell help in the tumor milieu is required for recruitment and cytolytic function of CD8+ T lymphocytes. Cancer Res. 2010;70:8368–77. doi: 10.1158/0008-5472.CAN-10-1322.
    1. Melief CJ, van der Burg SH. Immunotherapy of established (pre)malignant disease by synthetic long peptide vaccines. Nat Rev Cancer. 2008;8:351–60. doi: 10.1038/nrc2373.
    1. Melief CJ. “License to kill” reflects joint action of CD4 and CD8 T cells. Clin Cancer Res. 2013;19:4295–6. doi: 10.1158/1078-0432.CCR-13-1432.
    1. Disis ML, Wallace DR, Gooley TA, Dang Y, Slota M, Lu H, Coveler AL, Childs JS, Higgins DM, Fintak PA, et al. Concurrent trastuzumab and HER2/neu-specific vaccination in patients with metastatic breast cancer. J Clin Oncol. 2009;27:4685–92. doi: 10.1200/JCO.2008.20.6789.
    1. Brunsvig PF, Kyte JA, Kersten C, Sundstrøm S, Møller M, Nyakas M, Hansen GL, Gaudernack G, Aamdal S. Telomerase peptide vaccination in NSCLC: a phase II trial in stage III patients vaccinated after chemoradiotherapy and an 8-year update on a phase I/II trial. Clin Cancer Res. 2011;17:6847–57. doi: 10.1158/1078-0432.CCR-11-1385.
    1. Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, Essahsah F, Fathers LM, Offringa R, Drijfhout JW, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009;361:1838–47. doi: 10.1056/NEJMoa0810097.
    1. Tomita Y, Nishimura Y. Long peptide-based cancer immunotherapy targeting tumor antigen-specific CD4(+) and CD8(+) T cells. Oncoimmunology. 2013;2:e25801. doi: 10.4161/onci.25801.
    1. Tomita Y, Yuno A, Tsukamoto H, Senju S, Yoshimura S, Osawa R, Kuroda Y, Hirayama M, Irie A, Hamada A, et al. Identification of CDCA1-derived long peptides bearing both CD4+ and CD8+ T-cell epitopes: CDCA1-specific CD4+ T-cell immunity in cancer patients. Int J Cancer. 2014;134:352–66. doi: 10.1002/ijc.28376.
    1. Wang P, Sidney J, Kim Y, Sette A, Lund O, Nielsen M, Peters B. Peptide binding predictions for HLA DR, DP and DQ molecules. BMC Bioinformatics. 2010;11:568. doi: 10.1186/1471-2105-11-568.
    1. Tomita Y, Yuno A, Tsukamoto H, Senju S, Kuroda Y, Hirayama M, Irie A, Kawahara K, Yatsuda J, Hamada A, et al. Identification of promiscuous KIF20A long peptides bearing both CD4+ and CD8+ T-cell epitopes: KIF20A-specific CD4+ T-cell immunity in patients with malignant tumor. Clin Cancer Res. 2013;19:4508–20. doi: 10.1158/1078-0432.CCR-13-0197.
    1. Saito S, Ota S, Yamada E, Inoko H, Ota M. Allele frequencies and haplotypic associations defined by allelic DNA typing at HLA class I and class II loci in the Japanese population. Tissue Antigens. 2000;56:522–9. doi: 10.1034/j.1399-0039.2000.560606.x.
    1. Mack SJ, Bugawan TL, Moonsamy PV, Erlich JA, Trachtenberg EA, Paik YK, Begovich AB, Saha N, Beck HP, Stoneking M, et al. Evolution of Pacific/Asian populations inferred from HLA class II allele frequency distributions. Tissue Antigens. 2000;55:383–400. doi: 10.1034/j.1399-0039.2000.550501.x.
    1. Harao M, Hirata S, Irie A, Senju S, Nakatsura T, Komori H, Ikuta Y, Yokomine K, Imai K, Inoue M, et al. HLA-A2-restricted CTL epitopes of a novel lung cancer-associated cancer testis antigen, cell division cycle associated 1, can induce tumor-reactive CTL. Int J Cancer. 2008;123:2616–25. doi: 10.1002/ijc.23823.
    1. Tomita Y, Harao M, Senju S, Imai K, Hirata S, Irie A, Inoue M, Hayashida Y, Yoshimoto K, Shiraishi K, et al. Peptides derived from human insulin-like growth factor-II mRNA binding protein 3 can induce human leukocyte antigen-A2-restricted cytotoxic T lymphocytes reactive to cancer cells. Cancer Sci. 2011;102:71–8. doi: 10.1111/j.1349-7006.2010.01780.x.
    1. Casazza JP, Betts MR, Price DA, Precopio ML, Ruff LE, Brenchley JM, Hill BJ, Roederer M, Douek DC, Koup RA. Acquisition of direct antiviral effector functions by CMV-specific CD4+ T lymphocytes with cellular maturation. J Exp Med. 2006;203:2865–77. doi: 10.1084/jem.20052246.
    1. Attig S, Hennenlotter J, Pawelec G, Klein G, Koch SD, Pircher H, Feyerabend S, Wernet D, Stenzl A, Rammensee HG, et al. Simultaneous infiltration of polyfunctional effector and suppressor T cells into renal cell carcinomas. Cancer Res. 2009;69:8412–9. doi: 10.1158/0008-5472.CAN-09-0852.
    1. Widenmeyer M, Griesemann H, Stevanović S, Feyerabend S, Klein R, Attig S, Hennenlotter J, Wernet D, Kuprash DV, Sazykin AY, et al. Promiscuous survivin peptide induces robust CD4+ T-cell responses in the majority of vaccinated cancer patients. Int J Cancer. 2012;131:140–9. doi: 10.1002/ijc.26365.
    1. Martorelli D, Muraro E, Merlo A, Turrini R, Rosato A, Dolcetti R. Role of CD4+ cytotoxic T lymphocytes in the control of viral diseases and cancer. Int Rev Immunol. 2010;29:371–402. doi: 10.3109/08830185.2010.489658.
    1. Corbière V, Chapiro J, Stroobant V, Ma W, Lurquin C, Lethé B, van Baren N, Van den Eynde BJ, Boon T, Coulie PG. Antigen spreading contributes to MAGE vaccination-induced regression of melanoma metastases. Cancer Res. 2011;71:1253–62. doi: 10.1158/0008-5472.CAN-10-2693.
    1. Ribas A, Timmerman JM, Butterfield LH, Economou JS. Determinant spreading and tumor responses after peptide-based cancer immunotherapy. Trends Immunol. 2003;24:58–61. doi: 10.1016/S1471-4906(02)00029-7.
    1. Hunder NN, Wallen H, Cao J, Hendricks DW, Reilly JZ, Rodmyre R, Jungbluth A, Gnjatic S, Thompson JA, Yee C. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N Engl J Med. 2008;358:2698–703. doi: 10.1056/NEJMoa0800251.
    1. Castellino F, Germain RN. Cooperation between CD4+ and CD8+ T cells: when, where, and how. Annu Rev Immunol. 2006;24:519–40. doi: 10.1146/annurev.immunol.23.021704.115825.
    1. Aarntzen EH, De Vries IJ, Lesterhuis WJ, Schuurhuis D, Jacobs JF, Bol K, Schreibelt G, Mus R, De Wilt JH, Haanen JB, et al. Targeting CD4(+) T-helper cells improves the induction of antitumor responses in dendritic cell-based vaccination. Cancer Res. 2013;73:19–29. doi: 10.1158/0008-5472.CAN-12-1127.
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–64. doi: 10.1038/nrc3239.
    1. Weide B, Zelba H, Derhovanessian E, Pflugfelder A, Eigentler TK, Di Giacomo AM, Maio M, Aarntzen EH, de Vries IJ, Sucker A, et al. Functional T cells targeting NY-ESO-1 or Melan-A are predictive for survival of patients with distant melanoma metastasis. J Clin Oncol. 2012;30:1835–41. doi: 10.1200/JCO.2011.40.2271.
    1. Godet Y, Fabre E, Dosset M, Lamuraglia M, Levionnois E, Ravel P, Benhamouda N, Cazes A, Le Pimpec-Barthes F, Gaugler B, et al. Analysis of spontaneous tumor-specific CD4 T-cell immunity in lung cancer using promiscuous HLA-DR telomerase-derived epitopes: potential synergistic effect with chemotherapy response. Clin Cancer Res. 2012;18:2943–53. doi: 10.1158/1078-0432.CCR-11-3185.
    1. Kyte JA, Gaudernack G, Dueland S, Trachsel S, Julsrud L, Aamdal S. Telomerase peptide vaccination combined with temozolomide: a clinical trial in stage IV melanoma patients. Clin Cancer Res. 2011;17:4568–80. doi: 10.1158/1078-0432.CCR-11-0184.
    1. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, et al. IMPACT Study Investigators Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–22. doi: 10.1056/NEJMoa1001294.
    1. Ding ZC, Zhou G. Cytotoxic chemotherapy and CD4+ effector T cells: an emerging alliance for durable antitumor effects. Clin Dev Immunol. 2012;2012:890178. doi: 10.1155/2012/890178.
    1. Walter S, Weinschenk T, Stenzl A, Zdrojowy R, Pluzanska A, Szczylik C, Staehler M, Brugger W, Dietrich PY, Mendrzyk R, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med. 2012;18:1254–61. doi: 10.1038/nm.2883.
    1. Tomita Y, Imai K, Senju S, Irie A, Inoue M, Hayashida Y, Shiraishi K, Mori T, Daigo Y, Tsunoda T, et al. A novel tumor-associated antigen, cell division cycle 45-like can induce cytotoxic T-lymphocytes reactive to tumor cells. Cancer Sci. 2011;102:697–705. doi: 10.1111/j.1349-7006.2011.01865.x.
    1. Demachi-Okamura A, Ito Y, Akatsuka Y, Tsujimura K, Morishima Y, Takahashi T, Kuzushima K. Epstein-Barr virus nuclear antigen 1-specific CD4+ T cells directly kill Epstein-Barr virus-carrying natural killer and T cells. Cancer Sci. 2008;99:1633–42. doi: 10.1111/j.1349-7006.2008.00852.x.
    1. Ramduth D, Day CL, Thobakgale CF, Mkhwanazi NP, de Pierres C, Reddy S, van der Stok M, Mncube Z, Nair K, Moodley ES, et al. Immunodominant HIV-1 Cd4+ T cell epitopes in chronic untreated clade C HIV-1 infection. PLoS One. 2009;4:e5013. doi: 10.1371/journal.pone.0005013.
    1. Tabata H, Kanai T, Yoshizumi H, Nishiyama S, Fujimoto S, Matsuda I, Yasukawa M, Matsushita S, Nishimura Y. Characterization of self-glutamic acid decarboxylase 65-reactive CD4+ T-cell clones established from Japanese patients with insulin-dependent diabetes mellitus. Hum Immunol. 1998;59:549–60. doi: 10.1016/S0198-8859(98)00050-0.
    1. Zarour HM, Storkus WJ, Brusic V, Williams E, Kirkwood JM. NY-ESO-1 encodes DRB1*0401-restricted epitopes recognized by melanoma-reactive CD4+ T cells. Cancer Res. 2000;60:4946–52.
    1. Imai K, Hirata S, Irie A, Senju S, Ikuta Y, Yokomine K, Harao M, Inoue M, Tsunoda T, Nakatsuru S, et al. Identification of a novel tumor-associated antigen, cadherin 3/P-cadherin, as a possible target for immunotherapy of pancreatic, gastric, and colorectal cancers. Clin Cancer Res. 2008;14:6487–95. doi: 10.1158/1078-0432.CCR-08-1086.
    1. Jung KO, Khan AM, Tan BY, Hu Y, Simon GG, Nascimento EJ, Lemonnier F, Brusic V, Miotto O, Tan TW, et al. West Nile virus T-cell ligand sequences shared with other flaviviruses: a multitude of variant sequences as potential altered peptide ligands. J Virol. 2012;86:7616–24. doi: 10.1128/JVI.00166-12.
    1. Britten CM, Janetzki S, Butterfield LH, Ferrari G, Gouttefangeas C, Huber C, Kalos M, Levitsky HI, Maecker HT, Melief CJ, et al. T cell assays and MIATA: the essential minimum for maximum impact. Immunity. 2012;37:1–2. doi: 10.1016/j.immuni.2012.07.010.

Source: PubMed

3
購読する