To understand why vaccine-activated tumor-specific T cells often fail to generate antitumor effects, we studied two α-fetoprotein–specific CD8+ T cells (Tet499 and Tet212) that had different antitumor effects. We found that Tet499 required high antigen doses for reactivation, but could survive persistent antigen stimulation and maintain their effector functions. In contrast, Tet212 had a low threshold of reactivation, but underwent exhaustion and apoptosis in the presence of persistent antigen. In vivo, Tet499 cells expanded more than Tet212 upon reencountering antigen and generated stronger antitumor effects. The different antigen responsiveness and antitumor effects of Tet212 and Tet499 cells correlated with their activation and differentiation states. Compared with Tet212, the population of Tet499 cells was less activated and contained more stem-like memory T cells (Tscm) that could undergo expansion in vivo. The TCR signaling strength on Tet499 was weaker than Tet212, correlating with more severe Tet499 TCR downregulation. Weak TCR signaling may halt T-cell differentiation at the Tscm stage during immune priming and also explains why Tet499 reactivation requires a high antigen dose. Weak TCR signaling of Tet499 cells in the effector stage will also protect them from exhaustion and apoptosis when they reencounter persistent antigen in tumor lesion, which generates antitumor effects. Further investigation of TCR downregulation and manipulation of TCR signaling strength may help design cancer vaccines to elicit a mix of tumor-specific CD8+ T cells, including Tscm, capable of surviving antigen restimulation to generate antitumor effects. Cancer Immunol Res; 5(10); 908–19. ©2017 AACR.

The success of using checkpoint blockade to unleash antitumor immunity (1) confirms that activation of the immune system can control cancer growth. Intensive efforts by vaccine researchers have yielded a variety of immunization approaches that can elicit tumor-specific immunity (2). Despite induction of tumor-specific T cells, the antitumor effect of cancer vaccines remains disappointing (3). Evidence from both animal models (4, 5) and human trials (6) suggests that the presence of tumor antigen–specific T cells does not necessarily translate into antitumor effects. The limited antitumor effect of tumor-specific T cells has been attributed to immune suppression (7) as well as to T-effector cell (Teff) exhaustion (8, 9). However, the lack of correlation between the frequency of antigen-specific T cells and antitumor effects (10–12) also suggests that intrinsic cell attributes may play a role in determining their antitumor outcome (13). Although high “functional avidity” of antigen-specific T cells might be a measure of antitumor efficacy (14–16), contrasting evidence shows that high avidity T cells are associated with weaker antitumor effects (17, 18). Indeed, in chronic infections (19, 20), the competitiveness of high-avidity T cells comes at a cost, i.e., overactivation, exhaustion, and apoptosis of Teff cells in the presence of persistent antigen stimulation, a scenario that also exists in tumor lesions. Thus, the attributes of CD8+ T cells and mechanisms underpinning the antitumor efficacy of tumor-specific immunity remain unclear and require in-depth investigation in order to improve the efficacy of current immunotherapies.

Lentivector (lv) activates CD8+ T-cell responses (21, 22) because of its transduction of dendritic cells (23). We found that immunization with lv-expressing epitope-optimized α-fetoprotein (opt-AFP) activated CD8+ cells to prevent carcinogen-induced autochthonous hepatocellular carcinoma (HCC) in mice (24). But it is not clear whether different epitope-specific CD8+ cells all have antitumor effect. In the current study, we investigated the antitumor effects of two AFP epitope-specific CD8+ cells that could be reproducibly induced by lv immunization. We found that epitope AFP212- and AFP499-specific CD8+ cells (hereafter as Tet212 and Tet499) generated different antitumor effects. We thus studied the molecular and cellular attributes of vaccine-activated T cells that are associated with their antitumor effects. We found that Tet499 were insensitive to antigen stimulation and survived persistent in vitro antigen restimulation and maintained their effector functions. In contrast, Tet212 were sensitive to antigen restimulation, resulting in more expansion at low doses of antigen, but became exhausted and apoptotic in the presence of high and persistent antigen. In vivo, Tet499 generated more expansion than Tet212 upon reencountering antigen. The different antigen responsiveness of Tet212 and Tet499 cells correlated to their different TCR signaling strength and the presence of stem-like memory T cells (Tscm), which had robust in vivo expansion. The TCR signaling on Tet499 was weaker, associated with severe TCR downregulation. Thus, TCR downregulation and weakened TCR signaling likely prevent Tet499 from overactivation and enhances Tscm responses in the priming phase, but also protect Teff from exhaustion and apoptosis at effector phase. Our data suggest that further investigation into the molecular mechanisms of how to control TCR downregulation and TCR signaling strength may help design cancer vaccines to elicit a diverse and healthy mix of tumor-specific CD8+ T cells, especially Tscm, to generate antitumor effects.

Mice

C57BL/6 (CD45.2 and CD45.1) and C3H mice were from Charles River. Nur77GFP mice (25) were from The Jackson Laboratory. Mice were bred and maintained in specific pathogen-free facility at Augusta University. Animal protocols were approved by the Institutional Animal Care and Use Committee.

Tetramers

The PE-labeled H-2Db/AFP212 and PE- or APC-labeled H-2Kb-Db/AFP499 tetramers were prepared by NIH Tetramer Core Facility. In the H-2Kb-Db/AFP499 tetramer, the α3 domain of H-2Kb is replaced with α3 domain of H-2Db to reduce nonspecific binding. The AFP212 and AFP499 peptides bind to H-2Db and H-2Kb, respectively. Wild-type AFP peptides were used to prepare tetramers.

Cell lines, tumor challenge, and tumor induction

EL4 and 293T cells were purchased from ATCC in 2010. Cells received from ATCC were immediately expanded and stored in liquid nitrogen. Each time, one vial of cells were thawed and used for less than 6 passages to maintain their authenticity. EL4-AFP tumor cells were established previously (24) by transducing parental EL4 cells with lv expressing mouse AFP. Cell lines were checked for mycoplasma by PCR test (Fisher Scientific). For tumor challenge, 1 × 105 EL4-AFP cells were injected subcutaneously into the flank of C57BL/6 mice. The induction of autochthonous HCC with diethylnitrosamine (DEN; Sigma) was conducted as described (24). Briefly, 2-week-old F1 mice of B6XC3H cross-bred were intraperitoneally injected with 50 μg of DEN per gram of mouse weight.

Recombinant viral vectors and immunization

The plasmid expressing epitope-optimized mouse AFP (opt-AFP) was described (24). The shorter AFP fragments of AFP142 and AFP164 were cloned into pLenti6 (Invitrogen) by PCR cloning. The lvs were prepared by transient cotransfection of 293T cells, and the vectors were concentrated and titered by measuring the p24 level as described (21).

To construct recombinant vv expressing the opt-AFP, a shuttle plasmid vector pG10 was used (26). The opt-AFP gene was cloned into the vector behind the p7.5 early gene promoter to create pG10-opt-AFP. CV-1 cells in 6-well plates were infected with a wild-type vaccinia virus of WR strain at multiplicity of infection of 0.1, and then transfected with pG10-opt-AFP by SuperFect reagent (Qiagen). The recombinant vv was selected in human TK-143 cells with addition of BrdUrd in the medium. After three rounds of plaque purification, the purity of the virus was verified by PCR assays for the presence of the transgene and deletion of the viral thymidine kinase gene, and by fluorescence of DsRED in the infected cells. The virus, designated as opt-AFP-vv, was amplified in HeLa cells and purified by a standard procedure (26).

For immunization, 2 × 107 transduction units of opt-AFP-lv were injected via footpad. To boost immune responses, 1.5 × 107 infectious units of recombinant opt-AFP-vv were injected intraperitoneally. For immunization in the autochthonous HCC model, 2-month-old F1 mice of B6XC3H that had been induced by carcinogen DEN were immunized with opt-AFP-lv and then boosted with opt-AFP-vv at 3 months.

In vitro restimulation

The splenocytes (6 million) were restimulated for the indicated time with various concentration of wild-type AFP212 (GSMLNEHVC) or AFP499 (SSYSNRRL) peptide in the presence of 20 IU/mL IL-2 (ProSpec-Tany Technogene). The wild-type AFP peptides were used to make sure that the restimulated T cells would recognize and kill tumor cells expressing wild-type AFP. In some experiments, 10 ng/mL of IL-7 and IL-15 were also added. To maintain the peptide level, half of the RPMI medium containing the indicated concentrations of peptides was changed every other day.

In the second setting, the splenic CD8 T cells were purified by magnetic beads from immunized CD45.2 mice. The splenocytes of naïve CD45.1 congenic mice were pulsed with indicated concentrations of AFP212 or AFP499 peptides for 2 hours. Free peptides were washed away. Two millions of the purified CD8 T cells were then restimulated with four millions of peptide-loaded CD45.1 splenocytes in a 24-well plate in RPMI media containing 20 IU/mL IL-2. After 48 hours, 4 million peptide-loaded CD45.1 fresh splenocytes were added to continue stimulation for another 2 days before analysis.

Immunological staining and flow-cytometry analysis

Peripheral mouse blood cells and the splenocytes (fresh or stimulated) were stained with indicated combinations of antibodies plus H-2Db/AFP212 or H-2Kb-Db/AFP499 tetramers. For tetramer staining after restimulation with peptide in the media, the stimulated cells need 12 hours rest to recover the TCR. DAPI was added to exclude dead cells. Intracellular staining of IFNγ was conducted as described (24) after the cells were restimulated with peptides in the presence of GolgiStop for 3.5 hours. Events were collected on the LSR II, and the data were analyzed using FCS Express III software. The antibodies used in this study include antibodies to mouse CD8 (Clone: 53-6.7, Biolegend), CD45.1 (Clone: A20, Biolegend), CD45.2 (Clone: 104, BD Bioscience), CD44 (Clone: 1M7, Biolegend), CD62L (Clone: MEL-14, Biolegend), CD127 (IL-7Ra; Clone: A7R34, Biolegend), PD-1 (Clone: 29F.1A12, Biolegend), CCR7 (Clone: 4B12, Biolegend), CD122 (IL-2Rβ; Clone: TM-β1, Biolegend), Sca-1 (Clone: D7, Biolegend,), CD95 (Clone: SA367H8, Biolegend), IFN-γ (Clone: XMG1.2, Biolegend), Bcl-2 (Clone: BCL/10C4, Biolegend), H-2K b/H-2Db (Clone: 28-8-6, Biolegend), and TCRβ (Clone: H57-597, BD Bioscience). The Annexin-V and DAPI (4,6-diamidino-2 phenylinole dilactate) were from Biolegend.

CTL assay

The in vitro CTL assay was performed as previously described (21, 24). The splenocytes from immunized mice were restimulated with 2 μg/mL of AFP212 or AFP499 peptide for indicated times in the RPMI media containing 20 units/mL of IL-2. The EL4 and EL4-AFP tumor cells were labeled with 1 μmol/L and 0.03 μmol/L of 5-(and 6)-carboxyfluorescein diacetate succinimidyl ester (CFSE; Invitrogen), respectively, for 10 minutes at 37°C. After washing, equal numbers (5 × 104) of CFSElo-labeled target EL4-AFP cells and CFSEhi-labeled EL4 control cells were cocultured in triplicate with the in vitro peptide-stimulated splenocytes at the indicated E/T ratios for 6 hours. The killing of target EL4-AFP cells was analyzed by flow cytometry. The specific killing activity was calculated using the formula [1-(ratio of CFSEhi/CFSElo in the absence of CTL)/(ratio of CFSEhi/CFSElo in the presence of CTL)] × 100, as previously described (21).

Cell sorting and adaptive cell transfer

The splenic CD8 T cells were enriched by magnetic beads (Stemcell Technologies) and then stained with anti-CD8, CD44, and CD62L antibodies plus H-2Db/mAFP212 or H-2Kb-Db/mAFP499 tetramer. The tetramer+ cells and their subsets were sorted on FACSAria (BD Biosciences). The sorted cells of indicated number were injected into CD45.1 congenic mice, which were then immunized with opt-AFP-lv. The CD45.2+CD8+Tet+ cells in the peripheral blood of CD45.1 mice were monitored once a week by tetramer staining.

Tetramer binding and dissociation assay

The tetramer binding and dissociation assays were conducted as described (27). For tetramer binding assay, 1 million splenocytes of the immunized mice were stained with anti-CD8 antibody and the indicated concentrations of H-2Db/AFP212 or H-2Kb-Db/AFP499 tetramer. Between the tetramer concentrations of 6.5 and 13 μg/mL, the percentage of Tet212 and Tet499 cells did not increase further, suggesting that all the tetramer+ cells were stained. The Tet212 and Tet499 cells detected at lower tetramer concentration were compared with those detected at the concentration of 13 μg/mL, which was considered as 100%.

For the tetramer dissociation assay, the cells were stained with 6.5 μg/mL of the tetramer. After washing away the free tetramers, the cells were left at room temperature for the indicated times in the presence of anti-H-2Kb/H-2Db antibody before they were analyzed for the percent and MFI of the Tet+ cells.

Statistical analysis

Statistical analyses were performed using t test or ANOVA (GraphPad Inc.).

Tet212 and Tet499 have different CTL activity and in vivo antitumor effects

To study whether different AFP epitope–specific CD8+ T cells could kill AFP+ tumor cells, we restimulated the splenocytes with AFP212 and AFP499 peptide in media and used them as effectors for in vitro CTL assays (Fig. 1A). After 6-day restimulation, splenocytes of naïve and wild-type-AFP-lv–immunized mice showed no CTL activity (Fig. 1B), consistent with our recent report (24). On the other hand, splenocytes from the opt-AFP-lv–immunized mice had CTL activity after AFP499 peptide restimulation (Fig. 1B and Supplementary Fig. S1A and S1B). However, after AFP212 peptide restimulation, splenocytes of the opt-AFP-lv–immunized mice generated no CTL activity. Kinetic study demonstrated that the CTL activity of opt-AFP-lv–immunized splenocytes was diminished with AFP212 peptide, but was enhanced with AFP499 peptide, restimulation (Supplementary Fig. S1C).

To study whether Tet212 and Tet499 generated different antitumor effects in vivo, we constructed two lvs, AFP142-lv and AFP164-lv, to activate Tet212 and Tet499 separately (Fig. 1C). We found that, like opt-AFP-lv, AFP164-lv immunization protected mice from AFP+ tumor challenge. In contrast, all mice immunized with AFP142-lv developed tumors (Fig. 1D). The lack of antitumor effect by Tet212 was not due to its incapability of recognizing AFP+ tumor cells because both Tet212 and Tet499 cells produced IFNγ after coculture with EL4-AFP cells (Supplementary Fig. S1D). Together, these data suggest that Tet212 and Tet499 cells have different responses to tumor antigen in vitro and in vivo, resulting in different antitumor effects.

Tet212 cells differ from Tet499 in reactivation threshold, exhaustion, and apoptosis

To investigate why Tet212 and Tet499 generate such different CTL activities and antitumor effects, we studied their antigen responsiveness in vitro. To generate more Tet212 and Tet499 cells, we boosted the lv-immunized mice with vv. The purified CD8+ cells from immunized CD45.2 mice were restimulated with naïve CD45.1 splenocytes pulsed with different peptides. After 4 days, we observed a dose-dependent increase of Tet212 cells between the pulsed peptide ranges of 0.1–2 μg/mL (Fig. 2A). However, at 10 μg/mL of AFP212 peptide, the percentage of Tet212 among total CD45.2 CD8+ cells began to decline. Furthermore, restimulation with 2 μg/mL of AFP212 peptide in media reduced Tet212 to ∼1% (Fig. 2A). In contrast, the percentage of Tet499 increased only slightly between the pulsed AFP499 peptide ranges of 0.1 to 10 μg/mL and increased more so after restimulation with AFP499 peptide in media (Fig. 2A). In addition, we analyzed the PD1 and Annexin-V level on Tet212 and Tet499 after stimulation. We found that, compared with Tet499, the percentage of PD1+ Tet212 and the MFI of PD1 on Tet212 was higher at every dose point (Fig. 2B). Similarly, the percentage and MFI of Annexin-V were also higher on Tet212 (Fig. 2C). The production of cytokine IFNγ by Tet212 cells was inversely related to AFP212 peptide dose (Fig. 2D and E). In contrast, the percentage and MFI of IFNγ+ Tet499 cells were directly related to the dose of AFP499 peptide.

The exhaustion and apoptosis of Tet212 and Tet499 cells were also studied when peptides were added directly to the restimulation media (Supplementary Fig. S2A). The data showed ∼10% of Tet212 and Tet499 cells being apoptotic prior to restimulation. AFP212 peptide restimulation caused Tet212 undergo dose-dependent apoptosis, ∼70% of Tet212 cell being apoptotic with 2 μg/mL of peptide stimulation (Supplementary Fig. S2B and S2C). In contrast, only ∼17% of Tet499 cells were apoptotic after AFP499 peptide restimulation. Consistent with the data, only 5% of Tet212 cells, but 80% of Tet499 cells, survived 4-day peptide restimulation (Supplementary Fig. S2D). Again, more Tet212 expressed higher level of PD1 in a dose-dependent manner (Supplementary Fig. S2E). Furthermore, Tet212 lost their cytokine production and CTL activity after restimulation (Supplementary Fig. S2F). In contrast, the majority of Tet499 was not exhausted and maintained their effector function. The exhaustion and apoptosis were different from activation-induced anergy (28) as it could not be rescued by the addition of IL2 or even IL7 and IL15.

In summary, Tet212 have a low threshold of reactivation and can be expanded by low-dose antigen. But its high sensitivity to antigen restimulation leads to progressive upregulation of PD1 and reciprocal decrease of IFNγ and apoptosis with increasing dose of antigen. In contrast, Tet499 have a high reactivation threshold and thus is difficult to activate and expand. But the Tet499′s insensitivity to antigen also protects them from exhaustion and activation-induced apoptosis. As a result, persistent antigen restimulation (such as peptide in media) decreased Tet212 but increased Tet499 cells.

Tet499 proliferate better in vivo than Tet212 in response to emerging tumor antigen

We then studied in vivo reexpansion of Tet212 and Tet499 upon reencountering antigen. Tet212 and Tet499 cells were sorted from immunized CD45.2 mice and transferred into CD45.1 congenic mice followed by immunization (Fig. 3A). The data showed a significant more increase of Tet499 than Tet212 cells in the CD45.1 mice (Fig. 3B). In addition, the carcinogen-induced autochthonous HCC model was utilized to study in vivo antigen responsiveness of Tet212 and Tet499 cells to emerging AFP tumor antigen (Fig. 3C). We first found that the magnitude of Tet499 responses after vv boost was greater than that of Tet212 (Fig. 3D), suggesting Tet499 cells primed by lv underwent more extensive expansion when reencountering antigen expressed from vv. Tet499 cells were also able to detect emerging AFP antigen in autochthonous HCC and responded by expansion (the spike of Tet499 at day 140 in Fig. 3D). In contrast, Tet212 cells did not generate expansion by vv boost and could not generate productive response to emerging AFP antigen.

Tet499 contain more Tscm, Tcm, and express more antiapoptotic Bcl-2 than Tet212

We next studied the possible mechanisms underpinning the different antigen responsiveness of Tet212 and Tet499 cells. Memory T cells, especially Tscm and Tcm, are critical for generating antitumor effect as they can undergo expansion in response to tumor antigen (29–31). To study if the antigen responsiveness and antitumor effect of Tet212 and Tet499 cells were related to Tscm and Tcm, we stained Tet212 and Tet499 with CD44 and CD62L. As demonstrated in Fig. 4A, the Tet+ CD8+ cells could be grouped into CD44+CD62L Teff (or effector memory, hereafter referred to as Teff), CD44+CD62L+ Tcm, and the CD44CD62L+ naïve-like Tscm. We found that Tet499 contained more Tscm and Tcm than Tet212 (Fig. 4A). Even after in vitro stimulation, the percentage of Tcm and Tscm was still higher among Tet499 (Fig. 4B). Further analysis showed that, different from the counterpart of naïve CD8+ T cells, the Tet499 Tscm expressed more of stem-like cell markers of ScaI, CD122, and CCR7 (Fig. 4C). To minimize nonspecific binding, beads enriched CD8+ T cells were pre-blocked by Fc blocker CD16/CD32 antibody. After taking these measures, we verified that Tet499 contained more Tscm than Tet212 (Supplementary Fig. S3A). The CD44CD62L+ Tscm also expressed higher level of CD95 than naïve CD8+ T cells, another potential marker of Tscm (Supplementary Fig. S3B). The presence of Tscm was then confirmed in the Tet499 cells by dual-color tetramer staining (Supplementary Fig. S3C).

The resistance to antigen-induced Tet499 apoptosis may also be due to the Bcl-2 molecules (32). Indeed, we found that the Tscm of Tet499 expressed the highest level of Bcl-2 compared with Tcm and Teff, nearly to the same level as naïve CD8+ cells (Supplementary Fig. S3D). In addition, the Bcl-2 level in the Tcm and Teff of Tet499 cells was higher than the counterparts of Tet212, suggesting that even at the Tcm and Teff stages, Tet499 cells can better survive antigen-induced cell death.

Tscm undergo more in vivo expansion than Tcm and Teff upon reencountering antigen

In Fig. 3B, we showed that Tet499 cells generated more reexpansion. To determine which subsets of Tet499 cells could undergo in vivo expansion, we sorted the Teff, Tcm, and Tscm and transferred them into CD45.1 mice (Fig. 5A). Because the number of Tscm and Tcm cells in the Tet499+ population was too low to be efficiently collected, we sorted the Teff, Tcm, and Tscm based on the total CD8+ cells (see sorting strategy and purity of Tscm, Tcm, and Teff in Supplementary Fig. S4A and S4B). The number of Tet499+Tscm, Tet499+Tcm, and Tet499+Teff was calculated using the percentage of Tet499+ in the sorted CD8+ Tscm, Tcm, and Teff population (Supplementary Fig. S4C). Equal numbers (2,000) of Tet499+Tscm, Tet499+Tcm, and Tet499+Teff were transferred into CD45.1 mice. Teff generated no measurable expansion. In contrast, the CD45.2+ CD8+ cells were detected in the CD45.1 congenic mice receiving Tscm or Tcm cells (Fig. 5B and C). The percentage of CD45.2+Tet499 cells in the mice receiving Tscm increased by more than 30-fold, from ∼1% to ∼35%, whereas the percentage of CD45.2+Tet499 cells in the mice receiving Tcm increased only 5 times, from ∼1% to ∼5% (Supplementary Fig. S4C and Fig. 5B and C). These data suggest the Tet499+Tscm undergo more expansion than Tet499+Tcm in vivo after reencountering antigen. In addition, after adoptive transfer of Tscm, three populations of cells, Tscm, Tcm, and Teff, were found in the mice (Fig. 5D), suggesting that Tscm could differentiate into Tcm and Teff and maintained self-renewal capability.

The TCR signaling strength is weaker on Tet499 than on Tet212

It is postulated that TCR signaling strength affects antigen-induced T-cell differentiation, including induction of Tscm and Tcm (29, 33, 34), which have a greater potential of expansion when reencountering the same antigen (35). To determine the TCR signaling strength of Tet212 and Tet499 cells, we utilized the Nur77GFP mice (25), in which the TCR signaling strength is reflected by GFP intensity on T cells. We found that, fresh Tet212 and Tet499 cells from immunized Nur77GFP mice were GFP−/low prior to stimulation (Fig. 6A). After restimulation with 2 μg/mL peptide in media for 4 hours, all Tet212 and ∼90% of Tet499 cells became GFP+, with a higher GFP level on Tet212 cells (Fig. 6A and B). The remaining GFP−/low Tet499 after peptide stimulation contained more Tscm and Tcm cells; ∼40% of them were Tscm and Tcm. Even in the GFP+ population, Tet499 contained more Tscm and Tcm than Tet212 (Supplementary Fig. S5A–S5C). Peptide titration study revealed that the percentage and MFI of GFP+ Tet212 were higher than Tet499 at any concentration (Fig. 6C). For example, at 0.1 ng/mL, 80% of Tet212 cells were GFP+, whereas only 20% of Tet499 cells were GFP+. These data suggest that TCR signaling on Tet499 is weaker than Tet212. Furthermore, Tet212 GFP+ cells had higher CD44 expression than the Tet499 GFP+ cells (Fig. 6D), indicating that the Tet499 cells are in a lower activation state.

The TCR signaling strength of Tet212 and Tet499 cells was further studied by restimulating the purified CD45.2+CD8+ T cells from immunized Nur77GFP mice with peptide-pulsed CD45.1 splenocytes. We found that, prior to restimulation, fresh Tet212 of immunized mice express higher basal level GFP than Tet499 cells (Fig. 6E). Tet212 could become GFP+ even at low concentration of peptide (Fig. 6F). The Tet212 cells restimulated by splenocytes pulsed with 0.1 μg/mL of AFP212 peptide expressed higher level of GFP than Tet499 restimulated by splenocytes pulsed with 10 μg/mL of AFP499 peptide, suggesting that Tet499 TCR signaling is at least 100 times weaker than Tet212 TCR signaling.

Compared with Tet212, Tet499, TCRs have higher avidity for MHC/peptide but are more downregulated

The low reactivation threshold and stronger TCR signaling on Tet212 cells (Figs. 2 and 6) may be caused by high T-cell avidity for cognate MHC/peptide. Thus, we conducted a tetramer binding (Fig. 7A) and dissociation (Fig. 7B) assays, which measure the TCR avidity for the MHC/peptide complex. The data showed that approximately 90% of the Tet499 cells were stained with a low concentration (0.65 μg/mL) of H-2Kb-Db/AFP499 tetramer. In contrast, to achieve 90% staining of the Tet212 cells, 10 times more (6.5 μg/mL) of the H-2Db/AFP212 tetramer was required. In addition, the Tet499 TCR binding to MHC/peptide was dissociated more slowly (Fig. 7B). Thus, the Tet499 TCRs have a higher avidity for MHC/peptide.

Another factor that can influence the TCR signaling strength is the amount of TCR on the cell surface, which is normally downregulated after activation (36). Thus, we examined the TCR levels on Tet212 and Tet499 cells. The splenocytes from immunized mice were stained with antibodies to CD8, CD44, CD62L, TCR Vβ (Clone 57-597), plus tetramer. The TCR Vβ levels on Tet+ CD8+ T cells were compared with TetCD44CD62L+ naïve CD8+ T cells. Our data demonstrated that TCRs on Tet499 were more downregulated (Fig. 7C–E). Tet499 had only 40% of the TCRs as naïve CD8+ cells. In contrast, Tet212 had ∼80% of the TCRs as naïve CD8+ cells. By direct comparison, the Tet499 had approximately half the TCRs of Tet212 cells (Fig. 7F). The kinetic analysis of TCR downregulation showed that TCR downregulation on Tet212 was mild and transient (Fig. 7G). In contrast, TCR downregulation on Tet499 was severe and persistent. As the antibody to TCR Vβ staining may be interfered with by tetramers that compete for the same TCR, the CD3 level on Tet212 and Tet499 cells was thus measured to determine TCR downregulation. The data showed that CD3 was also more downregulated on Tet499 than on Tet212 (Supplementary Fig. S6A–S6D). Together, our data showed that, the Tet499 TCRs have higher avidity for MHC/peptide, but its level is more downregulated after activation, which may result in ensuing weaker TCR signaling and higher threshold of reactivation.

Our findings associate the antitumor effect of vaccine-activated CD8+ T cells with their antigen sensitivity and activation/differentiation states. First, the antigen sensitivity of two AFP-specific CD8+ T cells, Tet212 and Tet499, is different. Tet212 have a lower threshold of reactivation and are sensitive to antigen stimulation, but become exhausted and apoptotic when antigen persists and generate no antitumor effect in vivo. In contrast, Tet499 have a higher threshold of reactivation and are more resistant to antigen stimulation, but can survive persistent tumor antigen, maintain their effector function, and generate a potent antitumor effect in vivo. Secondly, the vaccine-activated Tet212 and Tet499 cells are at different differentiation and activation state. Tet499 cells are at a lower activation state and consist of diverse subsets at different differentiation stages, including Tscm and Tcm. In contrast, there are no Tscm and few Tcm in Tet212. The Tet499 Tscm can undergo extensive in vivo expansion when reencountering antigen. Thirdly, the TCR signaling on Tet212 is stronger and associated with mild and transient TCR downregulation. In contrast, the Tet499 TCR signaling is weaker, likely due to severe and persistent TCR downregulation.

TCR signaling strength correlates to the activation and magnitude of antigen-specific T cells (37–39). Indeed, data from Nur77GFP mice showed that the TCR signaling on Tet212 was stronger, corresponding to a higher magnitude of Tet212 response by immunization (24). But, on the other hand, strong TCR signaling can negatively affect the induction of Tscm (29, 40). In agreement with this argument, the Tet499, but not Tet212, contains Tscm. Stem-like memory immune cells were proposed by Fearon and colleagues (41). The Tscm was found to cause graft versus host disease (35). Gattinoni and colleagues showed that the TCR transgenic gp100-specific Tscm generated in vitro by inhibiting Wnt signaling had a better antitumor effect (42). They further found that human Tscm mediated strong antitumor effect in mice bearing human tumors (43). In a retrospective study, Speiser and colleagues found that yellow fever vaccine-induced Tscm in human correlated to long-term protection (44). Here, we show that cancer vaccine can elicit Tscm in vivo, which is associated with TCR downregulation and weak TCR signaling, and correlates with stronger antitumor effect.

The engagement of TCR and MHC/peptide and proper TCR signaling are also needed for the reactivation of vaccine-induced T cells to recognize and kill tumor cells. T cells with higher basal level of TCR signaling are more ready for activation (45, 46). Even though the Tet212 and Tet499 activated by vaccine are not naïve cells, their reactivation seems also correlate to their basal TCR signaling. Tet212 have a higher TCR signaling strength and are more ready for reactivation and consequently also suffers from activation induced cell death when antigen persists. In contrast, the Tet499 cells with weaker TCR signaling survive persistent antigen stimulation and generate a stronger antitumor effect. This is consistent with previous hypothesis that TCR downregulation may protect T cells from excessive signaling (47). Thus, TCR downregulation and weaker TCR signaling on the activated T cells in the effector phase may protect them from antigen-induced exhaustion and apoptosis, especially in the tumor lesions where the antigen is abundant and the stimulation is likely persistent. But the high concentration of antigens in tumor lesion and the prolonged engagement of T cells and tumor cells will compensate for the lower TCR level and generate sufficient accumulative signaling strength to activate Tet499 cells to kill tumor cells.

The association of weak TCR signaling with stronger antitumor effect of T cells seems counterintuitive. Our finding of Tet499 TCR downregulation associated with a strong antitumor effect is also in contrast to a report showing that TCR downregulation limits their efficacy of controlling microbial infection (48). We reason that this is likely due to the difference of antigen level and antigen stimulation duration between microbial infections and tumor lesion. In acute infections, the microbial antigen is transient, and thus T cells with higher level or sensitive TCR should not be exhausted and are more effective to eliminate the infection because of the stronger engagement of TCR and MHC/peptide. On the other hand, in tumor lesion, the T cells are immersed in a pool of tumor antigens for a prolonged period of time; thus, T cells with sensitive threshold of reactivation will likely be driven to exhaustion. Only the T cells with weaker TCR signaling may survive persistent antigen stimulation and generate antitumor effects. In agreement with this theory, in chronic persistent viral infection, T cells are exhausted (49). The report that T cells activated with intermediate affinity peptides (thus intermediate strength of TCR signaling) generate a better antitumor effect (50) is also in agreement with our data.

Based on these findings, we propose a model (Supplementary Fig. S7), in which the TCR signaling strength and the threshold of activation controls the state of T-cell activation, the generation of Tscm, antigen responsiveness, and the antitumor effect of the vaccine-activated CD8+ T cells. In this model, initial high-affinity TCR–MHC/peptide complex engagement results in T-cell activation and concurrently generates feedback to downregulate TCR on activated T cells, weakening the TCR signaling to avoid overactivation of T cells in the priming phase. The weakened TCR signaling will allow some activated T cells to halt differentiation at Tscm stage to reserve their potential to generate better responsiveness and stronger antitumor effect when they reencounter cognate antigen in tumor lesions. Thirdly, the weakened TCR signaling on Teff will protect them from exhaustion and apoptosis upon reencountering persistent antigen in tumor lesion. In contrast, if the TCR downregulation is mild and transient, TCR signaling will likely drive all activated T cells to become terminal Teff, which not only decrease Tscm, but also predispose the Teff to exhaustion and apoptosis when they reencounter antigen in tumor lesion.

In this model, we attribute the different TCR signaling strength and antigen sensitivity of Tet212 and Tet499 cells to their different TCR downregulation. However, these differences may also be due to the inherent difference of internal signaling pathways inside the Tet212 and Tet499 cells. Whether the high-affinity TCR–MHC/peptide engagement also reduces the internal signaling pathway is not clear, nor is the inherent signaling difference between naïve Tet212 and Tet499 cells. Even though our data support that the different antigen responsiveness of vaccine-activated Tet212 and Tet499 cells relates to their different TCR downregulation and signaling strength, further studies should be done to investigate whether the naïve Tet212 and Tet499 cells are different in their antigen responsiveness. If naïve Tet499 and Tet212 cells indeed have inherent signaling difference that is beyond the TCR level, the T cells with higher threshold of activation and capability of resisting antigen-induced cell death may be the ideal host cells for generating TCR and chimeric antigen receptor (CAR) gene–engineered T cells for immunotherapy of solid tumors, where the TCR or CAR gene–engineered T cells need to survive persistent antigen stimulation and generate antitumor effect.

This current study also raises questions in cancer vaccines and immunotherapy. The first question is whether TCR signaling strength on any antigen-specific T cells can be manipulated to enhance Tscm generation and to prevent exhaustion in tumor lesion. Secondly, how should cancer vaccine and immunization strategies be devised to find the right balance between eliciting sufficient number of tumor-specific T cells while inducing Tscm to achieve the maximal antitumor effect? Investigation into these questions should help develop more effective cancer vaccines and enhance the antitumor efficacy of current immunotherapies.

No potential conflicts of interest were disclosed.

Conception and design: S. Wu, W. Zhu, L. Huang, D. Bartlett, N. Fu, Y. He

Development of methodology: S. Wu, W. Zhu, Y. Hong, L. Huang, Z.S. Guo, Y. He

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Wu, W. Zhu, Y. Peng, L. Wang, Y. Hong, D. Dong, E. Kruse, Y. He

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Wu, W. Zhu, Y. Peng, Y. Hong, L. Huang, D. Dong, J. Xie, N. Fu, Y. He

Writing, review, and/or revision of the manuscript: S. Wu, L. Huang, D. Dong, T. Merchen, E. Kruse, D. Bartlett, Y. He

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): W. Zhu, Y. Peng, L. Huang, D. Dong, D. Bartlett, Y. He

Study supervision: Y. He

We acknowledge the stimulating discussion with Dr. Bjoern Peters of La Jolla Institute for Allergy and Immunology and Drs. Rafal Pacholczyk, David Munn, and Esteban Celis at Augusta University. The excellent service of NIH Tetramer Core Facility is greatly appreciated. We thank Dr. Rhea-Beth Markowitz of Augusta University for editing the manuscript.

This work is funded by NCI grant 1R01CA168912 and Georgia Cancer Center Grant to Y. He.

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.
Topalian
SL
,
Drake
CG
,
Pardoll
DM
.
Immune checkpoint blockade: a common denominator approach to cancer therapy
.
Cancer Cell
2015
;
27
:
450
61
.
2.
Melief
CJ
,
van Hall
T
,
Arens
R
,
Ossendorp
F
,
van der Burg
SH
.
Therapeutic cancer vaccines
.
J Clin Invest
2015
;
125
:
3401
12
.
3.
Rosenberg
SA
,
Yang
JC
,
Restifo
NP
.
Cancer immunotherapy: moving beyond current vaccines
.
Nat Med
2004
;
10
:
909
15
.
4.
Overwijk
WW
,
Theoret
MR
,
Finkelstein
SE
,
Surman
DR
,
de Jong
LA
,
Vyth-Dreese
FA
, et al
.
Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells
.
J Exp Med
2003
;
198
:
569
80
.
5.
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
.
6.
Rosenberg
SA
,
Sherry
RM
,
Morton
KE
,
Scharfman
WJ
,
Yang
JC
,
Topalian
SL
, et al
.
Tumor progression can occur despite the induction of very high levels of self/tumor antigen-specific CD8+ T cells in patients with melanoma
.
J Immunol
2005
;
175
:
6169
76
.
7.
Rabinovich
GA
,
Gabrilovich
D
,
Sotomayor
EM
.
Immunosuppressive strategies that are mediated by tumor cells
.
Annu Rev Immunol
2007
;
25
:
267
96
.
8.
Ahmadzadeh
M
,
Johnson
LA
,
Heemskerk
B
,
Wunderlich
JR
,
Dudley
ME
,
White
DE
, et al
.
Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired
.
Blood
2009
;
114
:
1537
44
.
9.
Zhou
Q
,
Xiao
H
,
Liu
Y
,
Peng
Y
,
Hong
Y
,
Yagita
H
, et al
.
Blockade of programmed death-1 pathway rescues the effector function of tumor-infiltrating T cells and enhances the antitumor efficacy of lentivector immunization
.
J Immunol
2010
;
185
:
5082
92
.
10.
Lee
PP
,
Yee
C
,
Savage
PA
,
Fong
L
,
Brockstedt
D
,
Weber
JS
, et al
.
Characterization of circulating T cells specific for tumor-associated antigens in melanoma patients
.
Nat Med
1999
;
5
:
677
85
.
11.
Dunbar
PR
,
Smith
CL
,
Chao
D
,
Salio
M
,
Shepherd
D
,
Mirza
F
, et al
.
A shift in the phenotype of melan-A-specific CTL identifies melanoma patients with an active tumor-specific immune response
.
J Immunol
2000
;
165
:
6644
52
.
12.
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
.
13.
Appay
V
,
Douek
DC
,
Price
DA
.
CD8+ T cell efficacy in vaccination and disease
.
Nat Med
2008
;
14
:
623
8
.
14.
Aranda
F
,
Llopiz
D
,
Diaz-Valdes
N
,
Riezu-Boj
JI
,
Bezunartea
J
,
Ruiz
M
, et al
.
Adjuvant combination and antigen targeting as a strategy to induce polyfunctional and high-avidity T-cell responses against poorly immunogenic tumors
.
Cancer Res
2011
;
71
:
3214
24
.
15.
Vigano
S
,
Utzschneider
DT
,
Perreau
M
,
Pantaleo
G
,
Zehn
D
,
Harari
A
.
Functional avidity: a measure to predict the efficacy of effector T cells?
Clin Dev Immunol
2012
;
2012
:
153863
.
16.
Lovgren
T
,
Baumgaertner
P
,
Wieckowski
S
,
Devevre
E
,
Guillaume
P
,
Luescher
I
, et al
.
Enhanced cytotoxicity and decreased CD8 dependence of human cancer-specific cytotoxic T lymphocytes after vaccination with low peptide dose
.
Cancer Immunol Immunother
2012
;
61
:
817
26
.
17.
McMahan
RH
,
McWilliams
JA
,
Jordan
KR
,
Dow
SW
,
Wilson
DB
,
Slansky
JE
.
Relating TCR-peptide-MHC affinity to immunogenicity for the design of tumor vaccines
.
J Clin Invest
2006
;
116
:
2543
51
.
18.
Morgan
DJ
,
Kreuwel
HT
,
Fleck
S
,
Levitsky
HI
,
Pardoll
DM
,
Sherman
LA
.
Activation of low avidity CTL specific for a self epitope results in tumor rejection but not autoimmunity
.
J Immunol
1998
;
160
:
643
51
.
19.
Price
DA
,
Brenchley
JM
,
Ruff
LE
,
Betts
MR
,
Hill
BJ
,
Roederer
M
, et al
.
Avidity for antigen shapes clonal dominance in CD8+ T cell populations specific for persistent DNA viruses
.
J Exp Med
2005
;
202
:
1349
61
.
20.
Lichterfeld
M
,
Yu
XG
,
Mui
SK
,
Williams
KL
,
Trocha
A
,
Brockman
MA
, et al
.
Selective depletion of high-avidity human immunodeficiency virus type 1 (HIV-1)-specific CD8+ T cells after early HIV-1 infection
.
J Virol
2007
;
81
:
4199
214
.
21.
He
Y
,
Zhang
J
,
Mi
Z
,
Robbins
P
,
Falo
LD
Jr
.
Immunization with lentiviral vector-transduced dendritic cells induces strong and long-lasting T cell responses and therapeutic immunity
.
J Immunol
2005
;
174
:
3808
17
.
22.
Esslinger
C
,
Chapatte
L
,
Finke
D
,
Miconnet
I
,
Guillaume
P
,
Levy
F
, et al
.
In vivo administration of a lentiviral vaccine targets DCs and induces efficient CD8(+) T cell responses
.
J Clin Invest
2003
;
111
:
1673
81
.
23.
He
Y
,
Zhang
J
,
Donahue
C
,
Falo
LD
Jr
.
Skin-derived dendritic cells induce potent CD8(+) T cell immunity in recombinant lentivector-mediated genetic immunization
.
Immunity
2006
;
24
:
643
56
.
24.
Hong
Y
,
Peng
Y
,
Guo
ZS
,
Guevara-Patino
J
,
Pang
J
,
Butterfield
LH
, et al
.
Epitope-optimized alpha-fetoprotein genetic vaccines prevent carcinogen-induced murine autochthonous hepatocellular carcinoma
.
Hepatology
2014
;
59
:
1448
58
.
25.
Moran
AE
,
Holzapfel
KL
,
Xing
Y
,
Cunningham
NR
,
Maltzman
JS
,
Punt
J
, et al
.
T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse
.
J Exp Med
2011
;
208
:
1279
89
.
26.
Guo
ZS
,
Naik
A
,
O'Malley
ME
,
Popovic
P
,
Demarco
R
,
Hu
Y
, et al
.
The enhanced tumor selectivity of an oncolytic vaccinia lacking the host range and antiapoptosis genes SPI-1 and SPI-2
.
Cancer Res
2005
;
65
:
9991
8
.
27.
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
.
28.
Mescher
MF
,
Popescu
FE
,
Gerner
M
,
Hammerbeck
CD
,
Curtsinger
JM
.
Activation-induced non-responsiveness (anergy) limits CD8 T cell responses to tumors
.
Semin Cancer Biol
2007
;
17
:
299
308
.
29.
Gattinoni
L
,
Klebanoff
CA
,
Restifo
NP
.
Paths to stemness: building the ultimate antitumour T cell
.
Nat Rev Cancer
2012
;
12
:
671
84
.
30.
Flynn
JK
,
Gorry
PR
.
Stem memory T cells (TSCM)-their role in cancer and HIV immunotherapies
.
Clin Transl Immunology
2014
;
3
:
e20
.
31.
Xu
Y
,
Dotti
G
.
Selection bias: maintaining less-differentiated T cells for adoptive immunotherapy
.
J Clin Invest
2016
;
126
:
35
7
.
32.
Guerrero
AD
,
Welschhans
RL
,
Chen
M
,
Wang
J
.
Cleavage of anti-apoptotic Bcl-2 family members after TCR stimulation contributes to the decision between T cell activation and apoptosis
.
J Immunol
2013
;
190
:
168
73
.
33.
Joshi
NS
,
Kaech
SM
.
Effector CD8 T cell development: a balancing act between memory cell potential and terminal differentiation
.
J Immunol
2008
;
180
:
1309
15
.
34.
Lugli
E
,
Dominguez
MH
,
Gattinoni
L
,
Chattopadhyay
PK
,
Bolton
DL
,
Song
K
, et al
.
Superior T memory stem cell persistence supports long-lived T cell memory
.
J Clin Invest
2013
;
123
:
594
9
.
35.
Zhang
Y
,
Joe
G
,
Hexner
E
,
Zhu
J
,
Emerson
SG
.
Host-reactive CD8+ memory stem cells in graft-versus-host disease
.
Nat Med
2005
;
11
:
1299
305
.
36.
Itoh
Y
,
Hemmer
B
,
Martin
R
,
Germain
RN
.
Serial TCR engagement and down-modulation by peptide:MHC molecule ligands: relationship to the quality of individual TCR signaling events
.
J Immunol
1999
;
162
:
2073
80
.
37.
Hofmann
M
,
Radsak
M
,
Rechtsteiner
G
,
Wiemann
K
,
Gunder
M
,
Bien-Grater
U
, et al
.
T cell avidity determines the level of CTL activation
.
Eur J Immunol
2004
;
34
:
1798
806
.
38.
Labrecque
N
,
Whitfield
LS
,
Obst
R
,
Waltzinger
C
,
Benoist
C
,
Mathis
D
.
How much TCR does a T cell need?
Immunity
2001
;
15
:
71
82
.
39.
Corse
E
,
Gottschalk
RA
,
Allison
JP
.
Strength of TCR-peptide/MHC interactions and in vivo T cell responses
.
J Immunol
2011
;
186
:
5039
45
.
40.
Gattinoni
L
,
Speiser
DE
,
Lichterfeld
M
,
Bonini
C
.
T memory stem cells in health and disease
.
Nat Med
2017
;
23
:
18
27
.
41.
Fearon
DT
,
Manders
P
,
Wagner
SD
.
Arrested differentiation, the self-renewing memory lymphocyte, and vaccination
.
Science
2001
;
293
:
248
50
.
42.
Gattinoni
L
,
Zhong
XS
,
Palmer
DC
,
Ji
Y
,
Hinrichs
CS
,
Yu
Z
, et al
.
Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells
.
Nat Med
2009
;
15
:
808
13
.
43.
Gattinoni
L
,
Lugli
E
,
Ji
Y
,
Pos
Z
,
Paulos
CM
,
Quigley
MF
, et al
.
A human memory T cell subset with stem cell-like properties
.
Nat Med
2011
;
17
:
1290
7
.
44.
Fuertes Marraco
SA
,
Soneson
C
,
Cagnon
L
,
Gannon
PO
,
Allard
M
,
Abed Maillard
S
, et al
.
Long-lasting stem cell-like memory CD8+ T cells with a naive-like profile upon yellow fever vaccination
.
Sci Transl Med
2015
;
7
:
282ra48
.
45.
Fulton
RB
,
Hamilton
SE
,
Xing
Y
,
Best
JA
,
Goldrath
AW
,
Hogquist
KA
, et al
.
The TCR's sensitivity to self peptide-MHC dictates the ability of naive CD8(+) T cells to respond to foreign antigens
.
Nat Immunol
2015
;
16
:
107
17
.
46.
Mandl
JN
,
Monteiro
JP
,
Vrisekoop
N
,
Germain
RN
.
T cell-positive selection uses self-ligand binding strength to optimize repertoire recognition of foreign antigens
.
Immunity
2013
;
38
:
263
74
.
47.
Cai
Z
,
Kishimoto
H
,
Brunmark
A
,
Jackson
MR
,
Peterson
PA
,
Sprent
J
.
Requirements for peptide-induced T cell receptor downregulation on naive CD8+ T cells
.
J Exp Med
1997
;
185
:
641
51
.
48.
Gallegos
AM
,
Xiong
H
,
Leiner
IM
,
Susac
B
,
Glickman
MS
,
Pamer
EG
, et al
.
Control of T cell antigen reactivity via programmed TCR downregulation
.
Nat Immunol
2016
;
17
:
379
86
.
49.
Barber
DL
,
Wherry
EJ
,
Masopust
D
,
Zhu
B
,
Allison
JP
,
Sharpe
AH
, et al
.
Restoring function in exhausted CD8 T cells during chronic viral infection
.
Nature
2006
;
439
:
682
7
.
50.
McMahan
RH
,
Slansky
JE
.
Mobilizing the low-avidity T cell repertoire to kill tumors
.
Semin Cancer Biol
2007
;
17
:
317
29
.

Supplementary data