One obstacle in eliciting potent antitumor immune responses is the induction of tolerance to tumor antigens. TCRlo mice bearing a TCR transgene specific for the melanoma antigen tyrosinase-related protein-2 (TRP-2, Dct) harbor T cells that maintain tumor antigen responsiveness but lack the ability to control melanoma outgrowth. We used this model to determine whether higher avidity T cells could control tumor growth without becoming tolerized. As a part of the current study, we developed a second TRP-2–specific TCR transgenic mouse line (TCRhi) that bears higher avidity T cells and spontaneously developed autoimmune depigmentation. In contrast to TCRlo T cells, which were ignorant of tumor-derived antigen, TCRhi T cells initially delayed subcutaneous B16 melanoma tumor growth. However, persistence in the tumor microenvironment resulted in reduced IFN-γ production and CD107a (Lamp1) mobilization, hallmarks of T-cell tolerization. IFN-γ expression by TCRhi T cells was critical for upregulation of MHC-I on tumor cells and control of tumor growth. Blockade of PD-1 signals prevented T-cell tolerization and restored tumor immunity. Depletion of tumor-associated dendritic cells (TADC) reduced tolerization of TCRhi T cells and enhanced their antitumor activity. In addition, TADCs tolerized TCRhi T cells but not TCRlo T cells in vitro. Our findings show that T-cell avidity is a critical determinant of not only tumor control but also susceptibility to tolerization in the tumor microenvironment. For this reason, care should be exercised when considering T-cell avidity in designing cancer immunotherapeutics. Cancer Res; 73(2); 595–604. ©2012 AACR.

Many tumor antigens are derived from self-antigens that are tissue-specific differentiation antigens. These include melanoma antigens such as Melan-A, Gp-100, tyrosinase, tyrosinase-related protein-1, and tyrosinase-related protein-2 (TRP-2; refs. 1–3). These melanocyte differentiation antigens (MDA) are expressed by both melanoma cells and normal melanocytes. Immune responses against MDAs are regulated by central and peripheral tolerance mechanisms. Most high-avidity, self-reactive CD8+ T cells are deleted during thymic selection (4, 5). Some lower avidity, self-reactive T cells escape thymic deletion and persist in the periphery under the control of various peripheral tolerance mechanisms, including anergy, suppression, ignorance, and deletion (6–8). We and others have reported that tolerance to TRP-2 could be broken with various vaccination protocols, and provision of granulocyte macrophage colony-stimulating factor (GM-CSF) was able to improve the weaker autoimmune response to TRP-2 into a more potent antitumor response (9, 10).

While a longstanding goal of cancer immunotherapy is the generation of an adequate number of tumor-specific CD8+ T cells that are capable of effectively clearing the tumor, not only the quantity but also the “quality” of the cytotoxic T lymphocytes (CTL) is critical. Accordingly, CTLs can be classified on the basis of their avidity for antigen-bearing targets. High-avidity CTLs require lower antigen concentration for activation and effector function and are thought to be more effective than low-avidity cells in both antiviral and antitumor immunity (11–13). In fact, a significant effort in adoptive T cell therapy has focused on generating tumor-specific T cells with high avidity for tumor antigens either by isolating them based on MHC/antigen tetramer staining, or more recently, through transduction of genes encoding high-affinity, tumor-reactive T cell receptors (TCR) into patients' peripheral blood T cells (14, 15).

Adoptive immunotherapy with ex vivo expanded tumor-infiltrating lymphocytes (TIL) has achieved objective clinical responses in a significant fraction of patients with metastatic melanoma (16, 17). However, the failure of many patients to develop long-term tumor control may be, in part, due to tolerization of transferred T cells (18). Our laboratory has reported that in an experimental model of prostate cancer, tumor antigen–specific CD8+ T cells become tolerized after infiltrating tumor tissue (19–21). One previous study suggested that recognition of a xenogeneic, tumor-associated antigen by higher avidity T cells leads to increased susceptibility to tolerization (22). Given the recent excitement about genetic transfer of higher affinity TCRs to confer higher avidity to T cells for adoptive immunotherapy (23, 24), a better understanding of the role of T-cell avidity in T-cell tolerance to self/tumor antigen would be beneficial to generating more durable antitumor immune responses.

Using 2 CD8+ T-cell clones that recognized the same self-antigen (TRP-2180–188) but differed in their functional avidity, we generated 2 lines of TRP-2–specific TCR transgenic (Tg) mice—TCRlo and TCRhi. Our previous studies showed that despite infiltrating B16 melanoma tumors and remaining reactive against TRP-2, the lower avidity TCRlo T cells did not reduce subcutaneous B16 tumor growth (25). In the current study, we compared the difference between T cells derived from the 2 lines of TCR Tg mice. We report that the higher avidity TCRhi T cells generated superior antitumor activity and caused autoimmune depigmentation. However, they were more susceptible to tolerization, both in the tumor microenvironment (TME) as well as ex vivo following stimulation by tumor-associated dendritic cells (TADC). Given the trend for exploiting higher avidity T cells for cancer therapy, these findings suggest that selection of T cells based solely on elevated avidity may not be optimal for maintaining immunity to tumor antigens. Rather, strategies that target the tolerization of T cells and sustain antigen responsiveness may yield more durable antitumor responses.

Experimental mice

C57BL/6 female mice were purchased from National Cancer Institute Animal Production Area Facility (Frederick, MD). The TCR Tg mouse strain 37B7 (TCRlo mice) bears a TCR transgene that recognizes an H-2Kb–restricted epitope of TRP-2180–188 and was described previously (25). Mice were housed under specific pathogen-free conditions and were treated in accordance with NIH Guidelines under protocols approved by the Animal Care and Use Committee of the NCI-Frederick facility.

Generation of high-avidity TRP-2180–188 TCR Tg mice (TCRhi mice)

Vα5 and Vβ7 TCR chain usage by a TRP-2180–188 peptide-specific CD8+ T cell clone (clone 24; ref. 26) was identified by spectratype analysis (27) and were subsequently cloned and sequenced. TCRhi mice were generated by similar method as described for TCRlo mice (25). In some experiments, TCRhi mice were crossed to IFN-γ–deficient background (gift, Dr. Robert Wiltrout, NCI, Frederick, MD) to generate IFN-γ−/−TCRhi mice.

Cell line and peptide

B16-BL6, hereafter called B16, a TRP-2+ murine melanoma cell line, was maintained in culture media as previously described (25). TRP-2180–188 (SVYDFFVWL) peptides were purchased from New England Peptide.

Tetramer decay assay

The tetramer decay assay and subsequent analysis were conducted as described previously (28, 29). Briefly, splenocytes from TCR Tg mice were incubated with anti-CD8 and clonotype-specific anti-Vβ antibodies for 30 minutes at 4°C and then washed 3 times with fluorescence-activated cell-sorting (FACS) buffer [PBS + 0.5% bovine serum albumin + 0.1% sodium azide]. Cell suspensions were then incubated with TRP-2 tetramer (courtesy, NIAID Tetramer facility, Emory University, Atlanta, GA) for 2 hours at 4°C. A competing unlabeled anti-H2Kb Ab (clone Y3) was added to the cell suspension and aliquots were taken at 0, 10, 20, 40, 60 minutes up to 24 hours afterward. Cells were fixed in 1% paraformaldehyde/FACS buffer for analysis on BD LSR II.

IFN-γ ELISPOT Assay

Multiscreen plates (Millipore) were coated with 100 μL of IFN-γ capture antibody (BD Biosciences) overnight at 4°C. T cells were added to increasing concentrations of TRP-2180–188. After incubation, plates were washed and processed as previously described (19).

51Cr Release assay

The 51Cr release assay was conducted as described previously with some modification (25). Briefly, B16 cells were treated with IFN-γ (20 ng/mL) overnight and labeled with 51Cr and used as targets. Effector cells were generated by culturing TCR Tg T cells with TRP-2 Ag (1 μmol/L) and 20 IU interleukin (IL)-2 for 6 days and purified using CD8+ T Lymphocyte Enrichment Set (BD Biosciences) according to the manufacturer's instructions. Graded numbers of effectors were added to target cells in a 96-well plate to achieve the indicated effector: target (E:T) ratio. Four hours later, supernatants were harvested and radioactivity assessed using a WALLAC 1470 Gamma counter.

CFSE labeling and flow cytometric analysis

Lymph node (LN) cells from TCRhi-Thy1.1+ mice were dispersed into a single-cell suspension. CD8+CD44lo were enriched using biotin-conjugated anti-CD44 antibody (clone: IM7) and biotinylated CD8 T cell enrichment cocktail, followed by streptavidin magnetic beads (BD Pharmingen). The resulting cell population was labeled with 5,6-carboxyfluorescein-diacetate succinimidyl ester (CFSE) as previously described (25), and 2.0 × 106 antigen-specific CD8+ T cells were transferred into recipient mice by tail vein injection. Mice were euthanized on indicated days after adoptive transfer. Tumor or vaccine-draining lymph nodes (axillary, brachial, and inguinal) or spleens were incubated with antibodies directed against Thy1.1, CD8, and CD44. Intracellular IFN-γ, Granzyme B, and CD107a expression from TILs were analyzed as described previously (25). MHC-I (H-2Kb) expression on tumor cells was analyzed by immunostaining of enzymatically digested tumors, gating on CD44+CD45 tumor cells.

Adoptive transfer of transgenic T cells to treat subcutaneous B16 tumor

A total of 2.5 × 106 antigen-specific CD8+ lymph node T cells from TCRhi or TCRlo Rag−/− mice were adoptively transferred into B6 mice. The day after T cell transfer, mice were vaccinated s.c. with TRP-2 peptide–pulsed, bone marrow–derived dendritic cells (BMDC) as previously described (25). Alternatively, mice were injected with similar number of transgenic T cells 1 day after tumor challenge. Similar outcomes were noted irrespective of T cell transfer. In some studies, to study T cell responses in the context of treating established tumors, T cells were transferred into mice 9 days after tumor implantation.

Some mice were treated with in vitro activated TCR Tg T cells. In those studies, mice were injected s.c. with B16 tumor cells (1 × 105). Three, 7, and 11 days after tumor challenge, mice received an intravenous injection of in vitro generated TRP-2–specific effector cells (1 × 107). Effector cells were generated as described above for the 51Cr release assay. In all studies, tumor size was estimated by measuring perpendicular diameters using a caliper. Mice were euthanized when tumor area exceeded 250 mm2 and tumor size was recorded as 250 mm2 thereafter.

Blocking and depletion antibodies

A blocking antibody directed against IFN-γ (XMG6, kindly provided by Dr. Giorgio Trinchieri, NCI, Frederick, MD) was injected intraperitoneally (i.p.; 0.5 mg) on days 9 and 13 after B16 tumor challenge. Anti-CD317 (PDCA-1) antibody (0.5 mg/injection; kindly provided by Drs. Trinchieri and Marco Colonna, Washington University, St. Louis, MO) was injected i.p. on day 7, 8, and 15, with respect to B16 tumor challenge (30). Anti-PD-1 (clone RMP1-14) antibody was generously provided by Dr. Hideo Yagita (Juntendo University, Tokyo, Japan). Mice received 250 μg of anti-PD-1 starting 10 days after tumor challenge and every 3 days thereafter.

TADC isolation and in vitro tolerance assay

Subcutaneous B16 tumors were digested in 5 mL dissociation buffer (100 U/mL collagenase IV and 100 μg/mL DNase in RPMI/10% FBS) for 30 minutes at 37°C. DCs were isolated from single-cell suspensions of the tumor using phycoerythirn (PE)-coupled anti-PDCA-1 antibody and anti-PE magnetic beads with the Miltenyi MACS cell separation system (31). Splenic plasma DCs were isolated in the same way and used as control. Cell separations were conducted according to the manufacturer's instructions and consistently yielded purity of more than 95% CD11c+/CD317+.

For the in vitro tolerance assay, naïve TCRhi or TCRlo Thy1.1+ cells were cocultured for 72 hours with TRP-2 antigen–pulsed B16 TADCs (CD11c+/B220+/BST2+) isolated from subcutaneous B16 tumors. The T cells were re-isolated via negative selection against the DCs using magnetic beads (31). TCR Tg T cells were delivered secondary stimulation using splenocytes pulsed with TRP-2 peptide. After 48 hours, wells were pulsed with 1 μCi [3H] thymidine (Amersham) and harvested 16 hours later. Alternatively, intracellular staining for IFN-γ expression was tested as described above.

Statistical analysis

Statistical analyses for differences between group means were conducted by unpaired Student t test or ANOVA. Tumor growth was compared using a 2-way ANOVA. P < 0.05 was considered statistically significant. PRISM 5.0 software was used to analyze the data (GraphPad Software, Inc.).

TCRhi T cells display higher functional avidity than TCRlo T cells

Our previous studies showed that TRP-2-specific TCRlo T cells were unable to control B16 tumor growth despite infiltration of the tumor and retention of cytolytic ability (25). We subsequently prepared a second TCR transgenic line based on a T cell clone described to have high functional avidity (26); these T cells required lower doses of antigen for stimulation than the T cell clone from which the TCRlo mice were derived and displayed greater IFN-γ expression at indicated antigen doses (Supplementary Fig. S1). We observed a striking phenotypic difference in TCRhi mice compared with TCRlo mice (Fig. 1A): TCRhi mice spontaneously developed autoimmune depigmentation as initially revealed by diminished pigmentation of the ear at weaning. Over time, progressive vitiligo-like depigmentation was observed in the coat hair of TCRhi mice, which was rarely observed in TCRlo mice (Fig. 1A). Consistent with these observations, we detected T cells in the skin of TCRhi mice, but not in the skin of TCRlo mice (Supplementary Fig. S2).

Figure 1.

TCRhi T cells display higher functional avidity than TCRlo T cells. A, TCRhi Tg mice spontaneously developed autoimmune depigmentation. B, tetramer decay assay revealed T cells from TCRhi mice bound tetramer with higher avidity. Data are representative of 4 experiments with similar results.

Figure 1.

TCRhi T cells display higher functional avidity than TCRlo T cells. A, TCRhi Tg mice spontaneously developed autoimmune depigmentation. B, tetramer decay assay revealed T cells from TCRhi mice bound tetramer with higher avidity. Data are representative of 4 experiments with similar results.

Close modal

We next characterized the phenotype of CD8+ T cells from TCRhi mice. We observed a distinct population of cells that were CD25, CD44hi, and Ly6c+ that was not observed in TCRlo mice, which instead displayed a uniformly naïve phenotype: CD62Lhi (data not shown), CD25, CD44lo, and Ly6cneg while displaying comparable levels of TCR (Supplementary Fig. S3A). This phenotype suggested that these were antigen-experienced TCRhi T cells, presumably responding to endogenous TRP-2 antigen.

To confirm that TCRhi T cells respond to endogenous antigen, we enriched the CD44lo TCR Tg T cells using magnetic beads by negative selection. TCR Tg T cells were labeled with CFSE to monitor proliferation and transferred to immunocompetent naïve C57BL/6 mice that express TRP-2. Consistent with our previous findings from the TCRlo transgenic mice (25), only T cells from TCRhi mice transferred to WT mice showed detectable CFSE dilution. Among those cells that diluted CSFE, most were CD44hi (Supplementary Fig. S3B). These findings confirm that only TCRhi T cells responded to endogenous TRP-2 antigen in the absence of exogenous priming, presumably due to their elevated avidity.

We next conducted tetramer decay analysis to confirm that TCRhi T cells truly possess higher avidity than TCRlo T cells. This assay measures the binding strength of a soluble form of the TCR ligand, a TRP-2180–188/H-2Kb tetramer. The decay time to 50% maximal tetramer binding for the TCRlo T cells was 65 minutes but was 2,610 minutes for the TCRhi T cells (Fig. 1B). Thus, as predicted, T cells from TCRhi mice bound tetramer ligand with higher avidity than TCRlo T cells.

Given this difference in avidity for TCR ligand, we next assessed the functional differences between TCRlo and TCRhi T cells. We generated CD8+ effector cells by antigen stimulation in vitro and tested antigen-specific cytotoxicity. Consistent with higher functional avidity of the original T-cell clones (Supplementary Fig. S1), TCRhi effector T cells displayed greater cytotoxicity than TCRlo effector T cells (Supplementary Fig. S3C). Taken together, these data show that TCRhi T cells represented a higher avidity population than TCRlo T cells and this increased avidity resulted in increased responsiveness to TRP-2 Ag, including increased IFN-γ production and Ag-specific lytic potential.

T cell avidity determines the magnitude of response to B16-derived antigen

On the basis of their different responses to endogenous antigen as well as in vitro function, we next sought to determine the response of TCRhi and TCRlo T cells to B16 tumor–derived TRP-2 antigen. CFSE-labeled TCR Tg T cells were transferred into WT B6 mice, and the following day, mice were challenged with B16 tumor cells. This enabled us to study T cell responses to tumor-derived TRP-2 antigen. Twenty days later, we analyzed CFSE dilution and expression of CD44 and IFN-γ by tumor-infiltrating TCR T cells. As shown in Fig. 2A, only TCRhi T cells recognized B16 tumor–derived Ag, as shown by a prominent fraction of tumor-infiltrating TCRhi T cells displaying diminished CFSE levels and elevated CD44 expression. Approximately 15% of TCRhi T cells that infiltrate the B16 tumors expressed IFN-γ. When comparing CFSE dilution, TCRhi T cells in the draining lymph node displayed more robust proliferative responses to B16 tumor–derived antigen than to endogenous TRP-2 antigen (Fig. 2A, top right, naïve vs. tumor); IFN-γ production was similarly elevated in LN TCRhi cells from tumor-bearing mice (22.6% vs. 5.7%, P < 0.0001). In contrast, in the absence of any exogenous stimulation, TCRlo T cells did not infiltrate B16 tumors (Fig. 2A), dilute CFSE, or generate effector function as measured by IFN-γ production, as we previously reported (25). Not surprisingly, vaccination with TRP-2 peptide–pulsed DCs 15 days after T-cell transfer induced a marked increase in infiltration of TCRhi T cells within the tumor and an elevated frequency of TCRhi T cells that expressed IFN-γ (Fig. 2B). This was again consistent with our previous report showing infiltration of TCRlo T cell into B16 tumors following DC vaccination (25).

Figure 2.

T-cell avidity determines the magnitude of response to B16-tumor derived antigen. A, CFSE-labeled CD8+CD44lo TCRhi and unsorted TCRlo Tg T cells were transferred into naïve B6 mice on day 0. The following day, mice were challenged s.c. with 1 × 105 B16 tumor cells. Twenty days after tumor challenge, the adoptively transferred T cells in B16 tumors were analyzed by testing for CD8, Thy1.1, CD44, and IFN-γ expression. Top right, TCRhi T cells from draining lymph nodes were similarly analyzed. **, P < 0.0001. B, mice were treated as in A except for receiving a s.c. TRP-2–pulsed BMDC vaccine 15 days after adoptive T-cell transfer. C, TCRhi T cells were injected i.v. and B16 tumor cells were injected s.c. into naïve B6 mice. Mice were vaccinated with TRP-2–pulsed BMDCs 9 days after tumor challenge. Tumor size was monitored. Data are representative of 3 separate experiments.

Figure 2.

T-cell avidity determines the magnitude of response to B16-tumor derived antigen. A, CFSE-labeled CD8+CD44lo TCRhi and unsorted TCRlo Tg T cells were transferred into naïve B6 mice on day 0. The following day, mice were challenged s.c. with 1 × 105 B16 tumor cells. Twenty days after tumor challenge, the adoptively transferred T cells in B16 tumors were analyzed by testing for CD8, Thy1.1, CD44, and IFN-γ expression. Top right, TCRhi T cells from draining lymph nodes were similarly analyzed. **, P < 0.0001. B, mice were treated as in A except for receiving a s.c. TRP-2–pulsed BMDC vaccine 15 days after adoptive T-cell transfer. C, TCRhi T cells were injected i.v. and B16 tumor cells were injected s.c. into naïve B6 mice. Mice were vaccinated with TRP-2–pulsed BMDCs 9 days after tumor challenge. Tumor size was monitored. Data are representative of 3 separate experiments.

Close modal

Given our observation that TCRhi T cells generated more robust responses than TCRlo T cells to both endogenous and tumor-derived antigen, we next tested whether transfer of TCRhi T cells could control B16 tumor. Despite modest generation of effector function (Fig. 2A), no reduction of tumor growth was noted in mice that were transferred with TCRhi T cells alone (Fig. 2C). However, transfer of TCRhi T cells in combination with a TRP-2–pulsed DC vaccine 9 days after tumor challenge significantly delayed tumor progression. This effect was not observed for TCRlo T cells (Fig. 2C; ref. 25). Similar results were obtained when treating s.c. B16 tumor cells with in vitro activated effector cells (Supplementary Fig. S4A). In addition, we also observed that s.c. B16 tumor growth was delayed in TCRhi Tg mice, but not in TCRlo Tg mice (Supplementary Fig. S4B). Taken together, these results indicate that TCRhi T cells generate superior antitumor activity compared with the lower avidity TCRlo T cells.

TCRhi T cells infiltrating subcutaneous B16 tumors are tolerized

Given that both in vivo and in vitro primed TCRhi T cells initially slowed subcutaneous B16 tumor growth, but all mice eventually developed progressive tumor growth, we hypothesized that TCRhi T cells may be progressively tolerized within the developing B16 TME. Therefore, we sequentially analyzed TCRhi T cell reactivity after tumor infiltration. As indicated in Fig. 3 and Supplementary Fig. S5, TCRhi TILs were highly responsive to TRP-2 antigen 5 days after vaccination. This included expression of IFN-γ and Granzyme B as well as CD107a mobilization, an indicator of CTL granule exocytosis. These observations were consistent with our previous studies examining TCRlo TILs (25). However, at later time points (e.g., 15 days after vaccination), a significant reduction in the frequency of TCRhi TILs expressing IFN-γ and mobilizing CD107a was observed in comparison to the earlier time point, or when compared with TCRhi T cells isolated from the spleen of naïve and tumor-bearing mice (Fig. 3A and B). Interestingly, we noted a loss of responsiveness in TCRhi T cells isolated from the spleen of tumor-bearing mice relative to those isolated from spleens of naïve mice. Total GrB expression remained consistent throughout the course of the experiments (Fig. 3C), indicative of prior activation of the TILs. These data show that high-avidity TCRhi T cells gradually lost their effector functions during tumor progression, which is in contrast to our previously published findings showing that lower avidity TCRlo TILs retain effector function within the TME, despite their inability to retard B16 tumor growth (25).

Figure 3.

TCRhi T cells infiltrating subcutaneous B16 tumors become tolerized. CD8+CD44loThy1.1+ TCRhi T cells were adoptively transferred into wild-type (WT) B6 mice. The following day, mice were challenged s.c. with B16 tumor cells. Nine days later, mice were vaccinated with TRP-2–pulsed BMDCs. At the indicated time after vaccination, mice were euthanized and splenocytes (SPL) or TILs were analyzed for IFN-γ expression (A), CD107a mobilization (B), and Granzyme B (Gr-B) expression (C). A and B, data from 3 experiments were combined (mean ± SD). C, data are presented from 1 of 2 identical experiments using 3 mice per group, pooled together for the assay. *, P < 0.05; **, P < 0.01; ***, P < 0.005.

Figure 3.

TCRhi T cells infiltrating subcutaneous B16 tumors become tolerized. CD8+CD44loThy1.1+ TCRhi T cells were adoptively transferred into wild-type (WT) B6 mice. The following day, mice were challenged s.c. with B16 tumor cells. Nine days later, mice were vaccinated with TRP-2–pulsed BMDCs. At the indicated time after vaccination, mice were euthanized and splenocytes (SPL) or TILs were analyzed for IFN-γ expression (A), CD107a mobilization (B), and Granzyme B (Gr-B) expression (C). A and B, data from 3 experiments were combined (mean ± SD). C, data are presented from 1 of 2 identical experiments using 3 mice per group, pooled together for the assay. *, P < 0.05; **, P < 0.01; ***, P < 0.005.

Close modal

IFN-γ expression by TCRhi T cells is critical for their antitumor immunity

One consequence of IFN-γ expression by TILs is the upregulation of MHC-I expression by tumor cells, which may affect susceptibility of tumor cells to T-cell–mediated lysis. Given the progressive loss of IFN-γ expression by TCRhi T cells, we next tested whether this loss of antigen responsiveness by TCRhi T cells correlated with a decrease in MHC-I expression by B16 tumor cells. While infiltration by both TCRhi and TCRlo T cells led to an increase in MHC-I expression by B16 tumor cells (Supplementary Fig. S6), we noted that there was a progressive decrease in MHC-I expression by B16 tumor cells that paralleled the loss of IFN-γ expression by TCRhi T cells (Fig. 4A, P = 0.002). To confirm the connection between IFN-γ expression and increases in MHC-I expression on tumor cells, we blocked IFN-γ using an IFN-γ neutralizing antibody. We observed that upregulation of MHC-I on tumor cells was significantly reduced following treatment with TCRhi T cells in combination with IFN-γ blockade (Fig. 4B).

Figure 4.

IFN-γ expression by TCRhi T cells is critical for antitumor immunity. A, TCRhi T cells were injected i.v. and B16 tumor cells were injected s.c. into naïve B6 mice. Mice were vaccinated with TRP-2-pulsed BMDCs 9 days after tumor challenge. Five and 15 days after DC vaccination, subcutaneous B16 tumors were excised. MHC-I (H-2Kb) expression on tumor cells was analyzed by gating on CD44+CD45 tumor cells. B, mice were challenged with tumor and treated as in A. Anti-IFN-γ antibody or an isotype control antibody was delivered i.p. on days 9 and 13 after tumor challenge. On day 15, MHC-I expression on tumor cells was analyzed by FACS staining. Some tumor-bearing mice received no T-cell transfer or DC vaccine (Ctrl). Data are representative of 3 independent experiments (mean ± SD). *, P < 0.005. C + D, TCRhi Tg mice were backcrossed to the Ifng-deficient background and T cells from these mice were transferred into 9-day tumor-bearing B6 mice. C, MHC-I expression on tumor cells was compared. D, tumor growth was monitored. *, P < 0.05; **, P < 0.0001. Representative of 3 separate experiments, 4 to 5 mice per group.

Figure 4.

IFN-γ expression by TCRhi T cells is critical for antitumor immunity. A, TCRhi T cells were injected i.v. and B16 tumor cells were injected s.c. into naïve B6 mice. Mice were vaccinated with TRP-2-pulsed BMDCs 9 days after tumor challenge. Five and 15 days after DC vaccination, subcutaneous B16 tumors were excised. MHC-I (H-2Kb) expression on tumor cells was analyzed by gating on CD44+CD45 tumor cells. B, mice were challenged with tumor and treated as in A. Anti-IFN-γ antibody or an isotype control antibody was delivered i.p. on days 9 and 13 after tumor challenge. On day 15, MHC-I expression on tumor cells was analyzed by FACS staining. Some tumor-bearing mice received no T-cell transfer or DC vaccine (Ctrl). Data are representative of 3 independent experiments (mean ± SD). *, P < 0.005. C + D, TCRhi Tg mice were backcrossed to the Ifng-deficient background and T cells from these mice were transferred into 9-day tumor-bearing B6 mice. C, MHC-I expression on tumor cells was compared. D, tumor growth was monitored. *, P < 0.05; **, P < 0.0001. Representative of 3 separate experiments, 4 to 5 mice per group.

Close modal

While multiple cell populations could deliver IFN-γ to increase tumor MHC-I expression, we determined the role of TCRhi T-cell–derived IFN-γ by crossing TCRhi mice onto the IFN-γ–deficient background. Using T cells from these mice for adoptive transfer into B16 tumor–bearing mice, we noted that Ifng−/−TCRhi T cells were unable to upregulate MHC-I expression on tumor cells as efficiently as WT TCRhi T cells (Fig. 4C). Loss of IFN-γ production by TCRhi T cells also resulted in the loss of their ability to slow B16 tumor growth (Fig. 4D). Taken together, these data indicate that IFN-γ expression by TCRhi T cells was critical for their antitumor activity and their ability to enhance MHC-I expression on their tumor targets.

PD-1 blockade improves TCRhi T-cell responses and reduced tumor burden

PD-1 is an inhibitory receptor expressed on activated T cells (23). We and others have reported that PD-1 blockade prevents T-cell exhaustion and tolerization, which confers improved immunity to tumors (24, 31). To determine whether PD-1 ligation contributes to TCRhi T-cell tolerization, we used an anti-PD-1 antibody to block PD-1–mediated signals. As shown in Supplementary Fig. S7A and S7B, PD-1 blockade improved T-cell responses and prevented tolerization of TCRhi TILs. In addition, we also observed restoration of responsiveness of TCRhi T cells in the spleen of tumor-bearing mice. This retention of T-cell responsiveness was associated with reduced tumor burden (Supplementary Fig. S7C). While these findings implicate a role for PD-1 in tolerization of TCRhi T cells, we cannot rule out the possibility that PD-1 blockade also targets another effector cell population, including natural killer cells (32) and/or endogenous B16-reactive T cells.

B16 tumor–associated dendritic cells tolerize TCRhi but not TCRlo T cells

Given the differential tolerization of TCRhi and TCRlo T cells infiltrating B16 tumors, we tested the possibility that B16 TADCs may preferentially tolerize TCRhi T cells. We recently identified a population of plasmacytoid-like TADCs with immunosuppressive function in both human and murine tumors, including B16 melanoma (31). These TADCs were CD11clo/B220+/BST2 (CD317)+ (Supplementary Fig. S8). To test the tolerogenic ability of the B16 TADCs, TCR Tg T cells were cocultured with TRP-2 peptide–pulsed TADCs for 4 days before re-isolation and subsequent restimulation with TRP-2–pulsed splenic antigen-presenting cells (APC). TCRhi T cells initially cultured with B16 TADCs did not proliferate (Fig. 5A) and had reduced IFN-γ production (Fig. 5B) in response to secondary antigenic stimulation, whereas marked proliferative and cytokine responses were observed when splenic plasmacytoid DCs (pDC) isolated using an identical approach were used as APCs for the primary stimulation. In contrast, TCRlo T cells initially cultured with either B16 TADCs or splenic pDCs displayed robust proliferative and IFN-γ responses after secondary stimulation (Fig. 5A and B). Furthermore, TADCs from Foxo3−/− mice did not tolerize TCRhi T cells (Supplementary Fig. S9), which is consistent with our recent finding that FOXO3 may program TADCs to become tolerogenic (31). Here, we further showed that T-cell avidity also contributes T-cell tolerance. Taken together, these data indicate that only TCRhi T cells were tolerized by B16 TADCs, consistent with our observations on tolerance induction following TCRhi T-cell infiltration into B16 tumors.

Figure 5.

B16 TADCs tolerize TCRhi T cells but not TCRlo T cells in vitro. TADCs were isolated from 15- to 20-day-old subcutaneous B16 tumors for use in the tolerization assay. Naïve TCR Tg T cells were cocultured for 4 days in vitro with TRP-2 peptide and B16 TADCs or, as a positive control, wild-type (WT) splenic plasmacytoid DCs (pDC) isolated using an identical protocol. TCR Tg T cells were re-isolated and tested for secondary stimulation with peptide-pulsed splenic APCs and proliferation was monitored by measuring [3H] thymidine incorporation (A) and IFN-γ production by intracellular staining (B). A, *, P = 0.0001 each point 1, 5, 10; B, *, P < 0.05; **, P < 0.005.

Figure 5.

B16 TADCs tolerize TCRhi T cells but not TCRlo T cells in vitro. TADCs were isolated from 15- to 20-day-old subcutaneous B16 tumors for use in the tolerization assay. Naïve TCR Tg T cells were cocultured for 4 days in vitro with TRP-2 peptide and B16 TADCs or, as a positive control, wild-type (WT) splenic plasmacytoid DCs (pDC) isolated using an identical protocol. TCR Tg T cells were re-isolated and tested for secondary stimulation with peptide-pulsed splenic APCs and proliferation was monitored by measuring [3H] thymidine incorporation (A) and IFN-γ production by intracellular staining (B). A, *, P = 0.0001 each point 1, 5, 10; B, *, P < 0.05; **, P < 0.005.

Close modal

On the basis of the above findings that TADCs preferentially tolerized TCRhi T cells in vitro, we next sought to determine whether depletion of TADCs in vivo would prevent TCRhi T-cell tolerization and promote tumor regression. Administration of an anti-CD317 antibody 1 week before T-cell transfer and vaccination, which was previously reported to deplete pDCs (30) and prostatic TADCs (31), depleted approximately 80% of TADCs in B16 melanoma tumors (Supplementary Fig. S10). Administration of anti-CD317 alone was not sufficient to alter tumor growth (Supplementary Fig. S11A), consistent with our observation that B16 tumor cells do not express CD317 (Supplementary Fig. S11B) and delivery of tumor-specific T cells is required for enhancing immunity to B16. Following TADC depletion, the frequency of IFN-γ–secreting TCRhi T cells from B16 tumors was significantly higher than those isolated from mice injected with a control antibody (Fig. 6A). CD107a mobilization was slightly but not significantly increased after TADC depletion (Fig. 6B), suggesting that the reduction of TADCs in TME at least partially prevented tolerization of TCRhi TILs. Consistent with the restoration of IFN-γ production, we noted significant upregulation of MHC-I on tumor cell after the depletion of TADCs (Fig. 6C). Moreover, B16 tumor burden was reduced in TADC-depleted mice compared with those in control antibody–treated mice (Fig. 6D). Taken together, these data show that selective tolerization of TCRhi T cells may ultimately contribute to their loss of tumor growth control.

Figure 6.

Depletion of B16 TADCs reduces tolerization of adoptively transferred TCRhi T cells in vivo. Wild-type (WT) B6 mice were injected s.c. with 1 × 105 B16 tumor cells on day 0. At days 7, 8, and 15, mice were injected i.p. with anti-CD317 antibody. TCRhi T cells were adoptively transferred into tumor-bearing mice on day 9 and mice were vaccinated with TRP-2–pulsed BMDCs the following day. A and B, mice were euthanized on day 21 and splenocytes or TILs were analyzed for IFN-γ expression (left) and CD107a mobilization (right). Data from 3 experiments were combined (mean ± SD). C, MHC-I (H-2Kb) expression on tumor cells was analyzed by gating on CD44+CD45 cells. Data were pooled from 2 independent trials with 3 mice per group. *, P < 0.05. D, WT B6 mice were injected s.c. with 1 × 105 B16 tumor cells on day 0. At days 9, 10, 17, 21, and 24, mice were injected i.p with anti-CD317 antibody (0.5 mg/injection). TCRhi T cells were adoptively transferred into tumor-bearing mice on day 12 and mice were vaccinated with TRP-2–pulsed BMDCs on day 13. Tumor size was monitored. Data are representative of 2 independent trials, with 4 to 5 mice per group.

Figure 6.

Depletion of B16 TADCs reduces tolerization of adoptively transferred TCRhi T cells in vivo. Wild-type (WT) B6 mice were injected s.c. with 1 × 105 B16 tumor cells on day 0. At days 7, 8, and 15, mice were injected i.p. with anti-CD317 antibody. TCRhi T cells were adoptively transferred into tumor-bearing mice on day 9 and mice were vaccinated with TRP-2–pulsed BMDCs the following day. A and B, mice were euthanized on day 21 and splenocytes or TILs were analyzed for IFN-γ expression (left) and CD107a mobilization (right). Data from 3 experiments were combined (mean ± SD). C, MHC-I (H-2Kb) expression on tumor cells was analyzed by gating on CD44+CD45 cells. Data were pooled from 2 independent trials with 3 mice per group. *, P < 0.05. D, WT B6 mice were injected s.c. with 1 × 105 B16 tumor cells on day 0. At days 9, 10, 17, 21, and 24, mice were injected i.p with anti-CD317 antibody (0.5 mg/injection). TCRhi T cells were adoptively transferred into tumor-bearing mice on day 12 and mice were vaccinated with TRP-2–pulsed BMDCs on day 13. Tumor size was monitored. Data are representative of 2 independent trials, with 4 to 5 mice per group.

Close modal

Using TCR Tg T cells that recognize the same tumor/self-antigen but display different avidity, we provide direct evidence that while higher avidity CD8+ T cells induce autoimmunity, they also display superior antitumor activity. However, higher avidity T cells are more susceptible to tolerization in the tumor microenvironment.

In the current study, we show that higher avidity CD8+ T cells from TCRhi Tg mice had a small but detectable population of cells that were “antigen-experienced” and these cells persisted in the periphery as putative auto-reactive T cells, which is supported by our observation that TCRhi mice develop spontaneous autoimmune depigmentation. Recognition of cognate antigen in the periphery can be regulated by either the avidity of responding T cells and/or antigen dose (33). Consistent with this idea, TCRhi T cell proliferation was more vigorous in response to tumor-derived antigen than endogenous antigen. This is presumably due to higher levels of TRP-2 antigen presentation in the tumor-draining lymph nodes. However, providing improved antigen priming through peptide-pulsed DC vaccination was capable of augmenting both TCRhi and TCRlo T cell responses and promoting tumor infiltration by both populations.

More importantly, our studies show that T cell avidity correlated with induction of tolerance, a significant obstacle to successful cancer immunotherapy (34). We show that unlike lower avidity TCRlo T cells, the higher avidity TCRhi T cells that persisted in the TME lost their ability to produce IFN-γ and to mobilize CD107a, hallmarks of tolerance. These data confirm our previous report showing loss of CTL function among tumor-specific CD8+ T cells that infiltrate prostate tumors (21). Morgan and colleagues also reported that higher avidity, Flu-HA–specific T cells were more readily tolerized than lower avidity T cells with identical specificity (22). Thus, our findings show that avidity may be critical in determining the fates of TILs within TME.

The impact of T-cell tolerization on antitumor immunity was amplified by the reduction of MHC expression by tumor cells, which correlated with loss of IFN-γ expression by TCRhi T cells. Loss of MHC-I expression represents a major impediment to successful immunotherapy and is a well-described mechanism of immune escape in many cancer types (35). The correlation between TCRhi T cell-derived IFN-γ and increased MHC-I expression by B16 tumor cells was further supported by in vivo neutralization of IFN-γ as well as studies using IFN-γ–deficient TCRhi T cells. These findings are in agreement with previous work implicating IFN-γ as an important factor for retention of antitumor immunity (35) and underscore the importance of maintaining T cell responsiveness for maintaining MHC expressing by tumors and tumor immunity. In separate studies, hydrodynamic delivery cDNA encoding IFN-γ only partially restored MHC-I expression by tumor cells and had minimal effect on restoring TCRhi-mediated antitumor activity (data was not shown). These observations are in agreement with the finding of Esumi and colleagues (36), suggesting that MHC-I expression is necessary but not sufficient for the induction of a host response to tumor and also imply that the source of IFN-γ may be critical, as well.

The mechanisms by which the tolerance induction occurs in tumors are only partially understood. It is widely accepted that the TME does not favor infiltrating tumor-specific T cells (37). In the present study, TADCs isolated from the B16 TME tolerized TCRhi, but not TCRlo T cells in vitro. Enhanced TCRhi T cell reactivity following depletion of B16 TADCs supports the idea that TADCs are at least one component of the TME responsible for tolerizing TCRhi T cells. Incomplete depletion of TADCs may explain partial restoration of TCRhi T-cell function and partial enhancement of immunity to B16 tumor. However, we cannot rule out the possibility that anti-CD317 may deplete another cell population with immunosuppressive functions. Furthermore, TADCs isolated from B16 tumors growing in Foxo3−/− mice were not tolerogenic, which is consistent with our previous finding (31). It remains unclear whether other components of the TME, such as myeloid-deprived suppressor cells (MDSC; ref. 38), regulatory T (Treg) cells (39), macrophages (40), or mast cells (41), also contribute to the tolerization of infiltrating TCRhi T cells. Similarly, blockade of PD-1 reduced tolerization of T cells and improved tumor immunity. As PD-1 has been associated with T cell exhaustion, it remains possible that the loss of T-cell function may be indicative of chronic T cell stimulation in the TME, a feature of T cell exhaustion. Strategies to eliminate TADCs or target their suppressive pathways such as FOXO3 or PD-1 may prevent high avidity T cell tolerization and thus enhance antitumor immunity.

Taken together, our findings show that while higher avidity T cells may initially confer better protection to tumors, they are more susceptible to tolerization in the TME. Many clinical trials are testing the use of transgenic TCRs for conferring melanoma specificity (42, 43). Specifically, an emphasis on higher avidity T cell populations has been proposed (44, 45). As T cell tolerance and exhaustion is also a feature of viral infections, these findings may also be applicable to anti-viral immune-based therapies (46). Therefore, caution should be exercised when selecting T cell populations for use in cancer immunotherapy. If low-avidity T cells are targeted, it will be necessary to optimize their effector function and overcome their reduced tumoricidal activity. However, identifying mechanisms that prevent or reverse tolerization and combat the potentially adverse results of autoimmune reactivity may also be acceptable alternatives for using higher avidity T cells. In addition, ongoing studies that elucidate the different signals transduced by these divergent T cell populations may further reveal novel therapeutic targets for maintaining durable and effective antitumor immunity.

No potential conflicts of interest were disclosed.

Conception and design: Z. Zhu, V. Singh, V. Bronte, A.A. Hurwitz

Development of methodology: Z. Zhu, V. Singh, S. Watkins, A.A. Hurwitz

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Z. Zhu, V. Singh, S. Watkins, L. Feigenbaum, J. Shoe

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Z. Zhu, V. Singh, S. Watkins, A.A. Hurwitz

Writing, review, and/or revision of the manuscript: Z. Zhu, V. Singh, S. Watkins, V. Bronte, A.A. Hurwitz

Study supervision: A.A. Hurwitz

The authors thank Dr. Joost Oppenheim for critical review of the manuscript and Terri Stull for her technical assistance.

This work was supported in-part by the intramural research program of the NCI, NIH.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Engelhard
VH
,
Bullock
TN
,
Colella
TA
,
Sheasley
SL
,
Mullins
DW
. 
Antigens derived from melanocyte differentiation proteins: self-tolerance, autoimmunity, and use for cancer immunotherapy
.
Immunol Rev
2002
;
188
:
136
46
.
2.
Cloosen
S
,
Arnold
J
,
Thio
M
,
Bos
GM
,
Kyewski
B
,
Germeraad
WT
. 
Expression of tumor-associated differentiation antigens, MUC1 glycoforms and CEA, in human thymic epithelial cells: implications for self-tolerance and tumor therapy
.
Cancer Res
2007
;
67
:
3919
26
.
3.
Rongcun
Y
,
Salazar-Onfray
F
,
Charo
J
,
Malmberg
KJ
,
Evrin
K
,
Maes
H
, et al
Identification of new HER2/neu-derived peptide epitopes that can elicit specific CTL against autologous and allogeneic carcinomas and melanomas
.
J Immunol
1999
;
163
:
1037
44
.
4.
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
.
5.
Kawai
K
,
Ohashi
PS
. 
Immunological function of a defined T-cell population tolerized to low-affinity self antigens
.
Nature
1995
;
374
:
68
9
.
6.
Hernandez
J
,
Aung
S
,
Redmond
WL
,
Sherman
LA
. 
Phenotypic and functional analysis of CD8(+) T cells undergoing peripheral deletion in response to cross-presentation of self-antigen
.
J Exp Med
2001
;
194
:
707
17
.
7.
Herndon
JM
,
Stuart
PM
,
Ferguson
TA
. 
Peripheral deletion of antigen-specific T cells leads to long-term tolerance mediated by CD8+ cytotoxic cells
.
J Immunol
2005
;
174
:
4098
104
.
8.
Redmond
WL
,
Sherman
LA
. 
Peripheral tolerance of CD8 T lymphocytes
.
Immunity
2005
;
22
:
275
84
.
9.
Ji
Q
,
Gondek
D
,
Hurwitz
AA
. 
Provision of granulocyte-macrophage colony-stimulating factor converts an autoimmune response to a self-antigen into an antitumor response
.
J Immunol
2005
;
175
:
1456
63
.
10.
Schreurs
MW
,
Eggert
AA
,
de Boer
AJ
,
Vissers
JL
,
van Hall
T
,
Offringa
R
, et al
Dendritic cells break tolerance and induce protective immunity against a melanocyte differentiation antigen in an autologous melanoma model
.
Cancer Res
2000
;
60
:
6995
7001
.
11.
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
.
12.
Derby
M
,
Alexander-Miller
M
,
Tse
R
,
Berzofsky
J
. 
High-avidity CTL exploit two complementary mechanisms to provide better protection against viral infection than low-avidity CTL
.
J Immunol
2001
;
166
:
1690
7
.
13.
Zeh
HJ
 III
,
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
.
14.
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
.
15.
Yee
C
,
Thompson
JA
,
Byrd
D
,
Riddell
SR
,
Roche
P
,
Celis
E
, et al
Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells
.
Proc Natl Acad Sci U S A
2002
;
99
:
16168
73
.
16.
Dudley
ME
,
Wunderlich
JR
,
Robbins
PF
,
Yang
JC
,
Hwu
P
,
Schwartzentruber
DJ
, et al
Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes
.
Science
2002
;
298
:
850
4
.
17.
Dudley
ME
,
Wunderlich
JR
,
Yang
JC
,
Sherry
RM
,
Topalian
SL
,
Restifo
NP
, et al
Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma
.
J Clin Oncol
2005
;
23
:
2346
57
.
18.
Rosenberg
SA
,
Restifo
NP
,
Yang
JC
,
Morgan
RA
,
Dudley
ME
. 
Adoptive cell transfer: a clinical path to effective cancer immunotherapy
.
Nat Rev Cancer
2008
;
8
:
299
308
.
19.
Shafer-Weaver
KA
,
Watkins
SK
,
Anderson
MJ
,
Draper
LJ
,
Malyguine
A
,
Alvord
WG
, et al
Immunity to murine prostatic tumors: continuous provision of T-cell help prevents CD8 T-cell tolerance and activates tumor-infiltrating dendritic cells
.
Cancer Res
2009
;
69
:
6256
64
.
20.
Shafer-Weaver
KA
,
Anderson
MJ
,
Stagliano
K
,
Malyguine
A
,
Greenberg
NM
,
Hurwitz
AA
. 
Cutting edge: tumor-specific CD8+ T cells infiltrating prostatic tumors are induced to become suppressor cells
.
J Immunol
2009
;
183
:
4848
52
.
21.
Anderson
MJ
,
Shafer-Weaver
K
,
Greenberg
NM
,
Hurwitz
AA
. 
Tolerization of tumor-specific T cells despite efficient initial priming in a primary murine model of prostate cancer
.
J Immunol
2007
;
178
:
1268
76
.
22.
Janicki
CN
,
Jenkinson
SR
,
Williams
NA
,
Morgan
DJ
. 
Loss of CTL function among high-avidity tumor-specific CD8+ T cells following tumor infiltration
.
Cancer Res
2008
;
68
:
2993
3000
.
23.
Keir
ME
,
Butte
MJ
,
Freeman
GJ
,
Sharpe
AH
. 
PD-1 and its ligands in tolerance and immunity
.
Ann Rev Immunol
2008
;
26
:
677
704
.
24.
Sakuishi
K
,
Apetoh
L
,
Sullivan
JM
,
Blazar
BR
,
Kuchroo
VK
,
Anderson
AC
. 
Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity
.
J Exp Med
2010
;
207
:
2187
94
.
25.
Singh
V
,
Ji
Q
,
Feigenbaum
L
,
Leighty
RM
,
Hurwitz
AA
. 
Melanoma progression despite infiltration by in vivo-primed TRP-2-specific T cells
.
J Immunother
2009
;
32
:
129
39
.
26.
De Palma
R
,
Marigo
I
,
Del Galdo
F
,
De Santo
C
,
Serafini
P
,
Cingarlini
S
, et al
Therapeutic effectiveness of recombinant cancer vaccines is associated with a prevalent T-cell receptor alpha usage by melanoma-specific CD8+ T lymphocytes
.
Cancer Res
2004
;
64
:
8068
76
.
27.
Currier
JR
,
Robinson
MA
. 
Spectratype/immunoscope analysis of the expressed TCR repertoire
.
Curr Protoc Immunol
2001
;
Chapter 10:Unit 10 28
.
28.
Savage
PA
,
Boniface
JJ
,
Davis
MM
. 
A kinetic basis for T cell receptor repertoire selection during an immune response
.
Immunity
1999
;
10
:
485
92
.
29.
Wang
XL
,
Altman
JD
. 
Caveats in the design of MHC class I tetramer/antigen-specific T lymphocytes dissociation assays
.
J Immunol Methods
2003
;
280
:
25
35
.
30.
Blasius
AL
,
Giurisato
E
,
Cella
M
,
Schreiber
RD
,
Shaw
AS
,
Colonna
M
. 
Bone marrow stromal cell antigen 2 is a specific marker of type I IFN-producing cells in the naive mouse, but a promiscuous cell surface antigen following IFN stimulation
.
J Immunol
2006
;
177
:
3260
5
.
31.
Watkins
SK
,
Zhu
Z
,
Riboldi
E
,
Shafer-Weaver
KA
,
Stagliano
KE
,
Sklavos
MM
, et al
FOXO3 programs tumor-associated DCs to become tolerogenic in human and murine prostate cancer
.
J Clin Invest
2011
;
121
:
1361
72
.
32.
Terme
M
,
Ullrich
E
,
Aymeric
L
,
Meinhardt
K
,
Desbois
M
,
Delahaye
N
, et al
IL-18 induces PD-1-dependent immunosuppression in cancer
.
Cancer Res
2011
;
71
:
5393
9
.
33.
Heath
WR
,
Karamalis
F
,
Donoghue
J
,
Miller
JF
. 
Autoimmunity caused by ignorant CD8+ T cells is transient and depends on avidity
.
J Immunol
1995
;
155
:
2339
49
.
34.
Rosenberg
SA
. 
Overcoming obstacles to the effective immunotherapy of human cancer
.
Proc Natl Acad Sci USA
2008
;
105
:
12643
4
.
35.
Dunn
GP
,
Koebel
CM
,
Schreiber
RD
. 
Interferons, immunity and cancer immunoediting
.
Nat Rev Immunol
2006
;
6
:
836
48
.
36.
Esumi
N
,
Hunt
B
,
Itaya
T
,
Frost
P
. 
Reduced tumorigenicity of murine tumor-cells secreting gamma-interferon is due to nonspecific host responses and is unrelated to class-I major histocompatibility complex expression
.
Cancer Res
1991
;
51
:
1185
9
.
37.
Zou
W
. 
Immunosuppressive networks in the tumour environment and their therapeutic relevance
.
Nat Rev Cancer
2005
;
5
:
263
74
.
38.
Marigo
I
,
Dolcetti
L
,
Serafini
P
,
Zanovello
P
,
Bronte
V
. 
Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells
.
Immunol Rev
2008
;
222
:
162
79
.
39.
Zou
W
. 
Regulatory T cells, tumour immunity and immunotherapy
.
Nat Rev Immunol
2006
;
6
:
295
307
.
40.
Stout
RD
,
Watkins
SK
,
Suttles
J
. 
Functional plasticity of macrophages: in situ reprogramming of tumor-associated macrophages
.
J Leukoc Biol
2009
;
86
:
1105
9
.
41.
Wasiuk
A
,
de Vries
VC
,
Hartmann
K
,
Roers
A
,
Noelle
RJ
. 
Mast cells as regulators of adaptive immunity to tumours
.
Clin Exp Immunol
2009
;
155
:
140
6
.
42.
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
.
43.
Roszkowski
JJ
,
Lyons
GE
,
Kast
WM
,
Yee
C
,
Van Besien
K
,
Nishimura
MI
. 
Simultaneous generation of CD8 +and CD4+ melanoma-reactive T cells by retroviral-mediated transfer of a single T-cell receptor
.
Cancer Res
2005
;
65
:
1570
6
.
44.
Zhao
Y
,
Bennett
AD
,
Zheng
Z
,
Wang
QJ
,
Robbins
PF
,
Yu
LY
, et al
High-affinity TCRs generated by phage display provide CD4+ T cells with the ability to recognize and kill tumor cell lines
.
J Immunol
2007
;
179
:
5845
54
.
45.
Kuball
J
,
Hauptrock
B
,
Malina
V
,
Antunes
E
,
Voss
RH
,
Wolfl
M
, et al
Increasing functional avidity of TCR-redirected T cells by removing defined N-glycosylation sites in the TCR constant domain
.
J Exp Med
2009
;
206
:
463
75
.
46.
Wherry
EJ
,
Ha
SJ
,
Kaech
SM
,
Haining
WN
,
Sarkar
S
,
Kalia
V
, et al
Molecular signature of CD8+ T cell exhaustion during chronic viral infection
.
Immunity
2007
;
27
:
670
84
.