Functional differences between low- and high-affinity CD8(+) T cells in the tumor environment

Rinke Bos, Kristi L Marquardt, Jocelyn Cheung, Linda A Sherman, Rinke Bos, Kristi L Marquardt, Jocelyn Cheung, Linda A Sherman

Abstract

Weak T-cell antigen receptor (TCR)-ligand interactions are sufficient to activate naïve CD8(+) T cells, but generally do not result in tumor eradication. How differences in TCR affinity affect the regulation of T-cell function in an immunosuppressive tumor environment has not been investigated. We have examined the functional differences of high- vs. low-affinity CD8(+) T cells and we observed that infiltration, accumulation, survival and cytotoxicity within the tumor are severely impacted by the strength of TCR-ligand interactions. In addition, high-affinity CD8(+) T cells were found to exhibit lower expression of inhibitory molecules including PD-1, LAG-3 and NKG2A, thus being less susceptible to suppressive mechanisms. Interferon γ and autocrine interleukin-2 were both found to influence the level of expression of these molecules. Interestingly, although high-affinity CD8(+) T cells were superior to low-affinity CD8(+) T cells in their ability to effect tumor eradication, they could be further improved by the presence of tumor specific CD4(+) T cells. These findings illustrate the importance of both TCR affinity and tumor-specific CD4 help in tumor immunotherapy.

Figures

https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3518496/bin/onci-1-1239-g1.jpg
Figure 1. Antitumor efficacy of high and low-affinity CD8+ T cells. (A and B) 8–9 week old RIP-Tag2-HA mice were immunized with cognate peptide and polyI:C in IFA and Clone 1 (A) or Clone 4 cells (B) (2 × 105) were injected i.v. Glucose levels in the blood were measured at the indicated time points, and each line represents one mouse. Data are representative of two independent experiments.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3518496/bin/onci-1-1239-g2.jpg
Figure 2. High-affinity CD8+ T cells are superior to low-affinity CD8+ T cells in the tumor milieu. 8–9 week old RIP-Tag2-HA mice were immunized with peptide and polyI:C in IFA and Clone 1 or Clone 4 cells (3 × 104) were injected i.v. (A–H) Pancreata and spleens were analyzed at day 7 by flow cytometry to assess percentage of CD8+Thy1.1+ cells, the percentage of cells exhibiting granzyme B, percentage of dividing cells and the expression level of Bim. Data are cumulative from 2 independent experiments with 3 mice per group. I-J, Pancreas derived CD8+Thy1.1+ cells were analyzed by qRT-PCR for perforin and Bcl-2 mRNA levels. Delta Ct values were compared using actin as the normalization control. Data are from 1 experiment with 3 independent samples per group. *p < 0.05, **p < 0.005, ***p < 0.0005.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3518496/bin/onci-1-1239-g3.jpg
Figure 3. Expression of inhibitory molecules in the spleen and tumor microenvironment. 8–9 week old RIP-Tag2-HA mice were treated as described in Figure 2. Pancreata and spleens were analyzed at day 7 by flow cytometry to assess the expression of PD-1, LAG-3 and NKG2A on CD8+Thy1.1+ cells. Histograms are representative of 3 independent experiments with 3 mice per group. Isotype control = gray line, Clone 4 = black line, Clone 1 = dashed line. Mean MFI ± SD, PD-1: Clone 4 2428.0 ± 216.4, Clone 1 2784.0 ± 234.7. LAG-3: Clone 4 446.5 ± 98.2, Clone 1 664.0 ± 135.76. NKG2A: Clone 4 724.5 ± 151.0, Clone 1 1001.0 ± 186.9.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3518496/bin/onci-1-1239-g4.jpg
Figure 4. Infiltration of high- vs. low-affinity CD8+ T cells. (A) Purified CD8+ Clone 1 or Clone 4 cells (3 × 105) were incubated with HA110–119 peptide pulsed splenocytes for 15 h. Cells were analyzed for the expression of integrins and lectins by flow cytometry. Data shown are representative of 2 independent experiments. (B–E) Clone 1 or Clone 4 cells were activated in vitro with HA110–119 peptide and after 6 d cells (5 × 106) were injected into InsHA (B and C) or RIP-Tag2-HA mice (14 weeks old, D,E). Pancreata, tumors and spleens were isolated 40 h after injection. In panels B and C, cumulative data are shown from 5 experiments with 15 mice total. Data shown in D and E are representative of 2 independent experiments with 3 mice per group. **p < 0.005, ***p < 0.0005.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3518496/bin/onci-1-1239-g5.jpg
Figure 5. Effects of autocrine IL-2 and IFNγ on function of Clone 4 cells in the tumor microenvironment. 8–9 week old RIP-Tag2-HA mice were immunized with peptide, polyI:C in IFA and Clone 4 or Clone 4 IL-2−/− (3x104) were injected i.v.. One group receiving Clone 4 cells was also injected with IFNγ neutralizing antibodies at days 4,5 and 6. (A–D) CD8+Thy1.1+ cells from pancreata were analyzed at day 7 by flow cytometry to assess the number of cells in the pancreas, the percentage of cells exhibiting granzyme B, the expression level of Bim and the percentage of dividing cells. Cumulative data are shown from 3 experiments with 2–3 mice per group per experiment. (E) Histograms are representative of 3 independent experiments with 3 mice per group. Isotype control = grey line, Clone 4 = black line, Clone 4 IL2-/- = dashed line, Clone 4 + anti-IFNγ = dotted line.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3518496/bin/onci-1-1239-g6.jpg
Figure 6. High-affinity CD8+ T cells benefit from CD4 help in the tumor milieu. 8–9 week old RIP-Tag2-HA mice were immunized with peptide, polyI:C in IFA and 3 x 104 Clone 4 cells with or without 2 × 105 SFE cells were injected i.v.. (A–D) Pancreata were analyzed at day 7 by flow cytometry to assess the number of cells in the pancreas, the percentage of cells exhibiting granzyme B, the expression level of Bim and the percentage of dividing cells. Data are cumulative of 2 experiments with 2–3 mice per group and are representative of 4 independent experiments. (E) Pancreata were analyzed at day 7 by flow cytometry to assess the expression of PD-1, LAG-3 and NKG2A on CD8+Thy1.1+ cells. Histograms are representative of 3 independent experiments with 2–3 mice per group. Isotype control = gray line, Clone 4 = black line, Clone 4 + SFE = dashed line. Mean MFI ± SD, PD-1: Clone 4 2428.0 ± 216.4; Clone 4+SFE 1569.0 ± 72.1. LAG-3: Clone 4 446.5 ± 98.2; Clone 4 + SFE 353.0 ± 33.9. NKG2A: Clone 4 724.5 ± 151.0; Clone 4 + SFE 681.0 ± 84.4.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/3518496/bin/onci-1-1239-g7.jpg
Figure 7. Improvement of antitumor efficacy of high-affinity CD8+ T cells by the presence of tumor-specific CD4+ T cells. 8–9 week old RIP-Tag2-HA mice were immunized with peptide and polyI:C in IFA and Clone 4 cells (A–B: 2 × 105, C–D: 3 × 104) with or without 2 × 105 SFE cells were injected i.v.. Glucose levels in the blood were measured at the indicated time points, and each line represents one mouse. Data are representative of two independent experiments. (A) compared with (B): p < 0.05, (C) compared with (D): p < 0.0005.

References

    1. Kyewski B, Klein L. A central role for central tolerance. Annu Rev Immunol. 2006;24:571–606. doi: 10.1146/annurev.immunol.23.021704.115601.
    1. Sprent J, Kishimoto H. The thymus and negative selection. Immunol Rev. 2002;185:126–35. doi: 10.1034/j.1600-065X.2002.18512.x.
    1. Miller JF, Kurts C, Allison J, Kosaka H, Carbone F, Heath WR. Induction of peripheral CD8+ T-cell tolerance by cross-presentation of self antigens. Immunol Rev. 1998;165:267–77. doi: 10.1111/j.1600-065X.1998.tb01244.x.
    1. Bouneaud C, Kourilsky P, Bousso P. Impact of negative selection on the T cell repertoire reactive to a self-peptide: a large fraction of T cell clones escapes clonal deletion. Immunity. 2000;13:829–40. doi: 10.1016/S1074-7613(00)00080-7.
    1. Nugent CT, Morgan DJ, Biggs JA, Ko A, Pilip IM, Pamer EG, et al. Characterization of CD8+ T lymphocytes that persist after peripheral tolerance to a self antigen expressed in the pancreas. J Immunol. 2000;164:191–200.
    1. Pardoll D. Does the immune system see tumors as foreign or self? Annu Rev Immunol. 2003;21:807–39. doi: 10.1146/annurev.immunol.21.120601.141135.
    1. Willimsky G, Blankenstein T. Sporadic immunogenic tumours avoid destruction by inducing T-cell tolerance. Nature. 2005;437:141–6. doi: 10.1038/nature03954.
    1. Klebanoff CA, Acquavella N, Yu Z, Restifo NP. Therapeutic cancer vaccines: are we there yet? Immunol Rev. 2011;239:27–44. doi: 10.1111/j.1600-065X.2010.00979.x.
    1. Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10:909–15. doi: 10.1038/nm1100.
    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. Wong SB, Bos R, Sherman LA. Tumor-specific CD4+ T cells render the tumor environment permissive for infiltration by low-avidity CD8+ T cells. J Immunol. 2008;180:3122–31.
    1. Merhavi-Shoham E, Haga-Friedman A, Cohen CJ. Genetically modulating T-cell function to target cancer. Semin Cancer Biol. 2012;22:14–22. doi: 10.1016/j.semcancer.2011.12.006.
    1. Jorritsma A, Gomez-Eerland R, Dokter M, van de Kasteele W, Zoet YM, Doxiadis II, et al. Selecting highly affine and well-expressed TCRs for gene therapy of melanoma. Blood. 2007;110:3564–72. doi: 10.1182/blood-2007-02-075010.
    1. Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. 2009;114:535–46. doi: 10.1182/blood-2009-03-211714.
    1. Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006;314:126–9. doi: 10.1126/science.1129003.
    1. Lyman MA, Nugent CT, Marquardt KL, Biggs JA, Pamer EG, Sherman LA. The fate of low affinity tumor-specific CD8+ T cells in tumor-bearing mice. J Immunol. 2005;174:2563–72.
    1. Dutoit V, Rubio-Godoy V, Dietrich PY, Quiqueres AL, Schnuriger V, Rimoldi D, et al. Heterogeneous T-cell response to MAGE-A10(254-262): high avidity-specific cytolytic T lymphocytes show superior antitumor activity. Cancer Res. 2001;61:5850–6.
    1. Alexander-Miller MA, Leggatt GR, Berzofsky JA. Selective expansion of high- or low-avidity cytotoxic T lymphocytes and efficacy for adoptive immunotherapy. Proc Natl Acad Sci U S A. 1996;93:4102–7. doi: 10.1073/pnas.93.9.4102.
    1. Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol. 2007;7:678–89. doi: 10.1038/nri2156.
    1. Marelli-Berg FM, Cannella L, Dazzi F, Mirenda V. The highway code of T cell trafficking. J Pathol. 2008;214:179–89. doi: 10.1002/path.2269.
    1. Zehn D, Lee SY, Bevan MJ. Complete but curtailed T-cell response to very low-affinity antigen. Nature. 2009;458:211–4. doi: 10.1038/nature07657.
    1. Zeh HJ, 3rd, Perry-Lalley D, Dudley ME, Rosenberg SA, Yang JC. High avidity CTLs for two self-antigens demonstrate superior in vitro and in vivo antitumor efficacy. J Immunol. 1999;162:989–94.
    1. Bullock TN, Mullins DW, Colella TA, Engelhard VH. Manipulation of avidity to improve effectiveness of adoptively transferred CD8(+) T cells for melanoma immunotherapy in human MHC class I-transgenic mice. J Immunol. 2001;167:5824–31.
    1. Savinov AY, Wong FS, Stonebraker AC, Chervonsky AV. Presentation of antigen by endothelial cells and chemoattraction are required for homing of insulin-specific CD8+ T cells. J Exp Med. 2003;197:643–56. doi: 10.1084/jem.20021378.
    1. Savinov AY, Wong FS, Chervonsky AV. IFN-gamma affects homing of diabetogenic T cells. J Immunol. 2001;167:6637–43.
    1. Refaeli Y, Van Parijs L, Alexander SI, Abbas AK. Interferon gamma is required for activation-induced death of T lymphocytes. J Exp Med. 2002;196:999–1005. doi: 10.1084/jem.20020666.
    1. Badovinac VP, Tvinnereim AR, Harty JT. Regulation of antigen-specific CD8+ T cell homeostasis by perforin and interferon-gamma. Science. 2000;290:1354–8. doi: 10.1126/science.290.5495.1354.
    1. Dalton DK, Haynes L, Chu CQ, Swain SL, Wittmer S. Interferon gamma eliminates responding CD4 T cells during mycobacterial infection by inducing apoptosis of activated CD4 T cells. J Exp Med. 2000;192:117–22. doi: 10.1084/jem.192.1.117.
    1. D’Souza WN, Lefrançois L. IL-2 is not required for the initiation of CD8 T cell cycling but sustains expansion. J Immunol. 2003;171:5727–35.
    1. Feau S, Arens R, Togher S, Schoenberger SP. Autocrine IL-2 is required for secondary population expansion of CD8(+) memory T cells. Nat Immunol. 2011;12:908–13. doi: 10.1038/ni.2079.
    1. Verdeil G, Marquardt K, Surh CD, Sherman LA. Adjuvants targeting innate and adaptive immunity synergize to enhance tumor immunotherapy. Proc Natl Acad Sci U S A. 2008;105:16683–8. doi: 10.1073/pnas.0805054105.
    1. Verdeil G, Puthier D, Nguyen C, Schmitt-Verhulst AM, Auphan-Anezin N. STAT5-mediated signals sustain a TCR-initiated gene expression program toward differentiation of CD8 T cell effectors. J Immunol. 2006;176:4834–42.
    1. Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, Beck A, Miller A, Tsuji T, et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci U S A. 2010;107:7875–80. doi: 10.1073/pnas.1003345107.
    1. Grosso JF, Kelleher CC, Harris TJ, Maris CH, Hipkiss EL, De Marzo A, et al. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. J Clin Invest. 2007;117:3383–92. doi: 10.1172/JCI31184.
    1. Woo SR, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72:917–27. doi: 10.1158/0008-5472.CAN-11-1620.
    1. Sheu BC, Chiou SH, Lin HH, Chow SN, Huang SC, Ho HN, et al. Up-regulation of inhibitory natural killer receptors CD94/NKG2A with suppressed intracellular perforin expression of tumor-infiltrating CD8+ T lymphocytes in human cervical carcinoma. Cancer Res. 2005;65:2921–9. doi: 10.1158/0008-5472.CAN-04-2108.
    1. Kessels HW, Schepers K, van den Boom MD, Topham DJ, Schumacher TN. Generation of T cell help through a MHC class I-restricted TCR. J Immunol. 2006;177:976–82.
    1. Morris EC, Tsallios A, Bendle GM, Xue SA, Stauss HJ. A critical role of T cell antigen receptor-transduced MHC class I-restricted helper T cells in tumor protection. Proc Natl Acad Sci U S A. 2005;102:7934–9. doi: 10.1073/pnas.0500357102.
    1. Kuball J, Schmitz FW, Voss RH, Ferreira EA, Engel R, Guillaume P, et al. Cooperation of human tumor-reactive CD4+ and CD8+ T cells after redirection of their specificity by a high-affinity p53A2.1-specific TCR. Immunity. 2005;22:117–29. doi: 10.1016/j.immuni.2004.12.005.
    1. Lyman MA, Aung S, Biggs JA, Sherman LA. A spontaneously arising pancreatic tumor does not promote the differentiation of naive CD8+ T lymphocytes into effector CTL. J Immunol. 2004;172:6558–67.
    1. Rozen S, Skaletsky H. Primer3 on the WWW for general users and for biologist programmers. Methods Mol Biol. 2000;132:365–86.

Source: PubMed

3
Suscribir