Adoptive cellular therapy (ACT) using T-cell receptor (TCR)–engineered lymphocytes holds promise for eradication of disseminated tumors but also an inherent risk of pathologic autoimmunity if targeted antigens or antigenic mimics are expressed by normal tissues. We evaluated whether modulating TCR affinity could allow CD8+ T cells to control tumor outgrowth without inducing concomitant autoimmunity in a preclinical murine model of ACT. RIP-mOVA mice express a membrane-bound form of chicken ovalbumin (mOVA) as a self-antigen in kidney and pancreas. Such mice were implanted with OVA-expressing ID8 ovarian carcinoma cells and subsequently treated with CD8+ T lymphocytes (CTL) expressing either a high-affinity (OT-I) or low-affinity (OT-3) OVA-specific TCR. The effects on tumor growth versus organ-specific autoimmunity were subsequently monitored. High-affinity OT-I CTLs underwent activation and proliferation in both tumor-draining and pancreatic lymph nodes, leading to both rapid eradication of ID8-OVA tumors and autoimmune diabetes in all treated mice. Remarkably, the low-affinity OT-3 T cells were activated only by tumor-derived antigen and mediated transient regression of ID8-OVA tumors without concomitant autoimmunity. The OT-3 cells eventually upregulated inhibitory receptors PD-1, TIM-3, and LAG-3 and became functionally unresponsive, however, allowing the tumors in treated mice to reestablish progressive growth. Antibody-mediated blockade of the inhibitory receptors prevented exhaustion and allowed tumor clearance, but these mice also developed autoimmune diabetes. The findings reveal that low-affinity TCRs can mediate tumor regression and that functional avidity can discriminate between tumor-derived and endogenous antigen, while highlighting the risks involved in immune-checkpoint blockade on endogenous self-reactive T cells.

Adoptive T-cell therapy (ACT) is a potent and flexible cancer treatment modality that can induce complete and durable regression of solid tumors (1, 2). Current cell therapy options include modifying autologous T cells to express either tumor-specific T-cell receptors (TCRs) or chimeric antigen receptors (CARs), which target surface proteins via a single-chain molecule containing an antigen-binding domain fused to transmembrane and cytoplasmic domains required for surface expression and T-cell triggering (3, 4). Although target recognition by CARs occurs in an HLA-unrestricted manner, TCRs have the ability to recognize peptides derived from exogenous, membrane-bound, and intracellular proteins, thus allowing a far greater repertoire of antigens to be targeted. Pioneering studies by Rosenberg and colleagues have demonstrated that ACT with tumor-infiltrating lymphocytes (TILs) can be effective against a variety of solid malignancies, including metastatic melanoma, gastrointestinal, and human papillomavirus–induced cancers, and this extends to bulky invasive tumors at multiple sites involving liver, lung, soft tissue, and brain (1, 5–8). These responses are durable in approximately 20% of treated patients who experience prolonged remissions of 5 years or more. However, a major limitation to the widespread application of TIL therapy is the difficulty in routinely generating human T cells with antitumor activity outside of a limited number of highly specialized laboratories.

An alternative approach to TIL-based ACT is the use of high-affinity TCRs against tumor-associated antigens (TAA) that can be expressed in a patient's normal T cells prior to expansion and reinfusion (9–13). Such genetically modified T cells have the potential to overcome the variability in the specificity, potency, and longevity of TIL. A major limitation of adoptive cell therapy utilizing such genetically modified cytotoxic T lymphocytes (CTLs) has been “on-target, off-tumor” toxicities associated when the transferred cells recognize antigens expressed on both tumor cells and normal tissue (14–17). In the first clinical trial using such engineered T cells targeting the melanoma TAA MART-1, objective responses were seen in 30% of melanoma patients receiving MART-1–targeted T cells, although over 80% of patients (29/36) developed an erythematous skin rash, and others developed anterior uveitis or hearing loss attributable to recognition of low expression of MART-1 in normal melanocytes, retina, and inner ear (9). Similarly, a phase I trial, using genetically modified T cells targeting carcinoembryonic antigen (CEA) resulted in severe transient inflammatory colitis as a result of “on-target” effects of the engineered T cells against colonic tissue expressing CEA (15). Another trial with genetically modified T cells targeting the cancer testis antigen (CTA) MAGE-A3, known to be expressed in a range of epithelial malignancies but not most normal tissues, resulted in severe neurologic toxicity from periventricular infiltration of adoptively transferred CD3+ CD8+ T cells with resultant necrotizing leukoencephalopathy and death in 2 of 9 treated patients due to previously unrecognized expression of MAGE antigens in the human brain (14).

To better understand the risks involved in ACT, we developed a new preclinical model to study the phenomenon of “on-target, off-tumor” toxicities in the context of ACT, which features adoptively transferred T cells specific for an antigen expressed on both tumor and normal tissues. This system is based on a well-characterized murine model (RIP-mOVA), which features steady-state surface expression of a model antigen (membrane-bound chicken ovalbumin or mOVA) on proximal tubular cells of the kidney and beta islets of the pancreas. Although these mice lack high-affinity OVA-specific CD8+ T cells in their peripheral repertoire due to central and peripheral tolerance mechanisms, adoptive transfer of high-affinity OT-I cells specific for the OVA257-264/H-2Kb complex rapidly leads to their activation with subsequent destruction of beta cells, and autoimmune diabetes. RIP-mOVA mice do possess a low-affinity OVA-specific T-cell repertoire, which can be elicited by vigorous vaccination and can occasionally cause autoimmune diabetes (18). OT-3 mice are transgenic for one such low-affinity OVA257-264/Kb–specific TCR that was isolated from immunized RIP-mOVA mice (18). In contrast to OT-I, OT-3 T cells become neither activated nor anergic in response to mOVA expression in pancreas or kidney tubulointerstitium following adoptive transfer to RIP-mOVA mice, and instead maintain their potential for response. In this way, OT-3 T cells are functionally analogous to the postthymic repertoire of self-reactive T cells that exists in the periphery of healthy human subjects. This model is therefore distinct from previous examples in which low-affinity T cells against a shared tumor/self-antigen are rendered profoundly anergic by expression of the target antigen on self-tissues (19).

Our objective in these studies was to determine whether TCR affinity could discriminate between self and tumor tissues expressing the same antigen. To address this, we established tumors in RIP-mOVA mice using an ovarian cancer model modified to express membrane-bound OVA (ID8-mOVA) and monitored tumor control versus autoimmune diabetes development following ACT with OT-I or OT-3 CD8+ T cells (Supplementary Fig. S1). We found that the low-affinity OT-3 T cells could be cross-primed and activated by antigen originating from the tumor, but not from normal self-tissues, and that these cells could thence mediate the selective eradication of tumor cells without concomitant autoimmune beta cell destruction. OT-3 T cells eventually became functionally unresponsive, and treatment with checkpoint blockade immunotherapy both restored their functionality with respect to tumor control, but also led to diabetes induction. These findings demonstrate that TCR affinity can allow ACT to discriminate between tumor and self, while highlighting the risks that exist for the use of systemic immune-checkpoint blockade immunotherapy.

Mice

Mice were maintained under specific pathogen-free conditions in accordance with guidelines of the Association for Assessment and Accreditation of Laboratory Animal Care International. The studies described in this paper conform to the principles outlined by the Animal Welfare Act and the National Institutes of Health guidelines for the care and use of animals in biomedical research. C57BL/6J (B6; CD45.2+), B6.SJL (CD45.1+), RIP-mOVA, and Rag1−/− mice were purchased from The Jackson Laboratory. Ovalbumin (OVA)-specific OT-I TCR-transgenic (Tg) CD45.1+ and OVA-specific OT-3 mice on a B6 background have been previously described. Six- to 12-week-old mice were used in all experiments.

Cell lines

ID8-OVA cells were a kind gift from Dr. Dietmar Zehn (Technical University Munich) in 2014. The B3Z T-cell hybridoma was a kind gift from Dr. Nilabh Shastri (University of California, Berkley). All tumor cell lines were tested prior to in vivo use and found to be free of Mycoplasma. In addition, model antigen expression for OVA was confirmed by flow cytometry. Due to the potential for cross-contamination of cell lines, ID8-OVA cells were reauthenticated by flow cytometry at least every 3 months, and cells were maintained in culture for no more than 30 passages. Mycoplasma testing was repeated at least every 6 months.

Flow cytometry, intracellular cytokine staining, and cytokine analysis

Single-cell suspensions were prepared from spleens, inguinal lymph nodes (LN), and tumors. Cells were stained with fluorescent-labeled antibodies (BioLegend; BD- Bioscience Pharmingen; or eBiosciences) and analyzed by either FACSCalibur or LSR II flow cytometer (BD). The following clones were used: CD4 (RM4-5), CD8 (5H1), CD11c (HL3), Cd11b (M170), CD25 (PC61.5), SIINFEKL/H-2Kb (25-D1.16), CD44 (IM7), CD62L (MEL-14), IFNγ (XMG1.2), TNFα (MP6-XT22), FAS (2495), CD40 (3/23), and Foxp3 (FJK-16s). For intracellular cytokine staining, cells were activated with OVA peptide-pulsed splenocytes or with PMA (100 ng/mL) plus ionomycin (500 ng/mL) for 4 hours in the presence of GolgiPlugTM (BD Biosciences), processed with a Cytofix/CytopermTM kit (BD Biosciences), and stained as indicated. Gates and quadrants were set based on isotype control staining unless otherwise indicated.

Adoptive transfer experiments and CTV labeling

T cells from spleens and LNs of OT1-Rag KO and OT-3 TCRalpha KO mice were used for adoptive cell transfer experiments. CD8+ T cells were isolated by negative selection of single-cell suspensions from homogenized donor tissue using a MACS CD8 isolation kit (Miltenyi Biotec). Purity of isolated CD8+ T cells was confirmed by flow cytometry analysis and was routinely greater than 95%, with less than 0.1% CD4+ T cell contamination. Donor T cells were obtained from CD45.1 mice, whereas recipient mice were CD45.2, thereby permitting in vivo isolation via this congenic marker. For in vivo cell proliferation studies, T cells (25 × 106) were washed twice in PBS and labeled in 5 nmol/L CTV for 8 minutes at 37°C on a shaker followed by quenching with ice-cold media. Cells were washed twice in PBS, and 5 × 106 cells were injected per mouse via retro-orbital injection. Mice were sacrificed 3 days later, and the spleen and LN cell suspensions were analyzed via flow cytometry. For other adoptive transfer experiments, donor T cells were harvested from the draining LN and spleens at the indicated time points for analysis.

Ectopic ID8 tumor model, intravital imaging (IVIS), and tumor vaccinations

Previously published methods were used for ectopic implantation of ID8 tumor cells (20). Briefly, while anesthetized, mice were inoculated s.c. with single-cell suspensions of 5 × 106 tumor cells suspended in 200 μL of PBS into the right flank. Three weeks later, the mice were monitored for tumor growth by tumor bioluminescence using an IVIS system and in response to treatment thereafter at the indicated time points. To monitor for tumor bioluminescence, mice received 300 μg of D-luciferin (Promega) by intraperitoneal injection 10 minutes before imaging ID8-OVA-luc tumors using a Xenogen IVIS 100 to maximum tumor radiance. Results are reported as the percent change from baseline tumor radiance on the day of treatment. For tumor vaccination studies, single-cell suspensions of tumor cells (107/mL) in PBS were subjected to 150 Gy of gamma ionizing radiation using an irradiator prior to s.c. flank inoculations of 5 × 106 cells into the right flank of recipient mice.

Antigen-specific immune response assay

Spleen and LN cells were incubated for 5 hours with the OVA257–264 (SIINFEKL) at 5 μg/mL final concentration in the presence of Brefeldin A (BD Biosciences) directly ex vivo and were stained for surface expression of CD8 and CD3, fixed and permeabilized using the Cytofix/Cytoperm kit (BD Biosciences) and stained for intracellular IFNγ according to the manufacturer's protocol.

B3Z assays

B3Z is a lacZ-inducible CD8+ T-cell hybridoma expressing TCR specific for OVA257–264 (SIINFEKL), presented on the murine H2Kb MHC class I molecule (21). ID8-OVA-luc, SAMBOK, or MECI cells were cocultured with B3Z for 3 hours at 37°C in a 5% CO2/air atmosphere, after which cells were washed, and β-galactosidase activity was detected by addition of substrate (CPRG, Sigma-Aldrich) and absorption readings at 600 nm using a spectrophotometer plate reader.

Monoclonal antibodies for in vivo immune-checkpoint studies

The following mAbs were purchased from Bio X Cell: InVivoPlus anti-mouse PD-1 (J43; cat. #BP0033-2), InVivoPlus anti-mouse LAG-3 (C9B7W; cat. #BP0174), and InVivoPlus anti-mouse TIM-3 (RMT3-23; cat. #BP0115). A total of 250 μg of each antibody was transferred intravenously in 200 μL of PBS into recipient mice.

Statistical analysis

Statistical analysis was performed using Prism 5 (GraphPad). One-way ANOVA and unpaired two-tailed Student t tests were conducted and considered statistically significant at P values ≤ 0.05 (*), 0.01 (**) and 0.001 (***).

Induction of autoimmune diabetes by OT-3 CTLs

We first sought to determine the quantitative threshold for diabetes induction by fully activated OT-3 in RIP-mOVA mice in the setting of ACT. Naïve OT-3 CD8+ T cells were harvested from donor mice by negative selection of homogenized splenocytes, and graded doses (103, 105, or 106) were transferred to recipient RIP-mOVA mice, which were subsequently infected by an OVA-expressing strain of wild-type Listeria monocytogenes (Lm-OVA) by intravenous injection. Mice were then monitored for diabetes induction (blood glucose > 200 mg/dL) daily after transfer. Despite the lower functional avidity for the OVA antigen expressed by the pancreatic beta cells, systemic activation of 106 OT-3 cells by Lm-OVA resulted in autoimmune destruction of beta islets in all RIP-mOVA mice (Fig. 1A). In addition, 106 OT-3 cells were also activated by an attenuated strain of Lm-OVA (ActA) that is unable to spread laterally via actin polymerization, demonstrating that a single priming stimulus, and not continued activation by proliferating Lm-OVA within reticuloendothelial cells, is sufficient to result in autoimmune diabetes (Fig. 1B). Finally, this phenomenon was dose-dependent, requiring 105 OT-3 cells to develop diabetes at 100% penetrance of the phenotype (Fig. 1C). These data indicate that OT-3 cells can be activated and induce autoimmunity in RIP-mOVA mice reproducibly with the appropriate activating stimulus.

Figure 1.

Induction of autoimmune diabetes by OT-3 cytotoxic T lymphocytes. A, Systemic activation of 106 adoptively transferred OT-3 CD8+ T cells by intravenous (i.v.) cotransfer of 3,000 CFU of OVA-expressing Listeria monocytogenes (Lm-OVA) resulted in autoimmune destruction of beta islets in RIP-mOVA mice. B, Systemic activation of OT-3 CD8 T cells by i.v. cotransfer of 106 CFU of Lm-OVA deficient for Act A (Lm-OVA Act A−/−) is sufficient to result in autoimmune diabetes. C, Autoimmune diabetes induced by adoptively transferred OT-3 CD8+ T cotransferred with 3,000 CFU of Lm-OVA cells is dose-dependent, requiring 105 OT-3 T cells to develop diabetes at 100% penetrance of the phenotype.

Figure 1.

Induction of autoimmune diabetes by OT-3 cytotoxic T lymphocytes. A, Systemic activation of 106 adoptively transferred OT-3 CD8+ T cells by intravenous (i.v.) cotransfer of 3,000 CFU of OVA-expressing Listeria monocytogenes (Lm-OVA) resulted in autoimmune destruction of beta islets in RIP-mOVA mice. B, Systemic activation of OT-3 CD8 T cells by i.v. cotransfer of 106 CFU of Lm-OVA deficient for Act A (Lm-OVA Act A−/−) is sufficient to result in autoimmune diabetes. C, Autoimmune diabetes induced by adoptively transferred OT-3 CD8+ T cotransferred with 3,000 CFU of Lm-OVA cells is dose-dependent, requiring 105 OT-3 T cells to develop diabetes at 100% penetrance of the phenotype.

Close modal

ID8-OVA tumors induce proliferation of adoptively transferred OT-3 CTLs via cross-presentation

Although OT-3 cells can induce autoimmune diabetes in RIP-mOvA after priming with a strong stimulus such as Lm-OVA infection, it was not known if a progressively growing tumor could also do so. To address this, OT-3 cells were labeled with the viable dye CTV (which allows visualization of cellular division by its portioning into daughter cells) and adoptively transferred to RIP-mOVA harboring ID8-OVA tumors. The mice were euthanized 96 hours following transfer, and the transferred cells were analyzed within the tumor-draining LN for proliferation using their congenic marker (CD45.1). The OT-3 T cells within RIP-mOVA mice bearing ID8-OVA tumors both proliferated (Fig. 2A) and acquired effector functionality (Fig. 2B), whereas those transferred to RIP-mOVA mice lacking tumors did neither. These results indicate that naïve OT-3 cells can be activated to proliferate by cross-presentation of OVA antigen in the tumor-draining LN.

Figure 2.

ID8-OVA tumors induce proliferation of adoptively transferred OT-3 CD8 T cells by cross-presentation. A, RIP-mOVA mice bearing 3-week-old ID8-OVA tumors elicited proliferation of adoptively transferred OT-3 CD8+ T cells that were identifiable by the congenic marker CD45.1 and labeled with cell-trace violet (CTV) within the tumor-draining LN 72 hours after transfer, whereas mice lacking tumors did not demonstrate proliferation. B, Adoptively transferred OT-3 CD8+ T cells into tumor-bearing mice produced Granzyme B 72 hours after transfer, consistent with functional activation.

Figure 2.

ID8-OVA tumors induce proliferation of adoptively transferred OT-3 CD8 T cells by cross-presentation. A, RIP-mOVA mice bearing 3-week-old ID8-OVA tumors elicited proliferation of adoptively transferred OT-3 CD8+ T cells that were identifiable by the congenic marker CD45.1 and labeled with cell-trace violet (CTV) within the tumor-draining LN 72 hours after transfer, whereas mice lacking tumors did not demonstrate proliferation. B, Adoptively transferred OT-3 CD8+ T cells into tumor-bearing mice produced Granzyme B 72 hours after transfer, consistent with functional activation.

Close modal

OT-I and OT-3 CTLs have different thresholds of antitumor reactivity versus autoimmune diabetes

Having established that OT-3 T cells cause autoimmune diabetes when systemically activated by a strong inflammatory stimulus (Lm-OVA), and that ID8-OVA tumors engraft in RIP-mOVA mice and are able to cross-prime adoptively transferred OT-3 T cells, we sought to compare whether OT-3 versus OT-I CD8 T cells could mediate control of ID8-OVA tumors without simultaneous off-tumor, on-target autoimmune destruction of pancreatic beta islets. To address this, 5 × 106 OT-I or OT-3 CD8 T cells were transferred to ID8-OVA tumor–bearing RIP-mOVA mice, which were then monitored simultaneously for changes in tumor size by CCD and diabetes induction by blood glucose analysis. As shown in Fig. 3, although the high-affinity OT-I T cells were able to mediate destruction of ID8-OVA tumors, they did so at the expense of autoimmune destruction of the beta islets, as demonstrated by development of diabetes (Fig. 3A). OT-3 cells, in contrast, were able to achieve partial destruction of the incipient tumor, but without concomitant autoimmunity. This effect was transient, however, and despite the partial destruction of the ID8 tumor by OT-3 ACT, the tumors eventually resumed progressive growth in all treated mice, suggesting that the transferred T cells had become ineffective or functionally silenced (Fig. 3B).

Figure 3.

OT-1 and OT-3 CD8 T cells have different thresholds of antitumor reactivity versus autoimmune diabetes. A, Adoptive cell transfer of 5 × 106 naïve OT-I CD8+ T cells, prepared by negative selection from donor transgenic OT-I mice splenocytes and LNs, mediates destruction of ID8-OVA tumors as reflected by decreased tumor bioluminescence. However, all recipient mice develop autoimmune diabetes. ACT of OT-3 CD8+ T cells, in contrast, mediates partial destruction of the incipient tumor without concomitant autoimmunity. B, ID8-OVA tumors that are transiently controlled by ACT of OT-3 CD8+ T cells demonstrate progressive tumor outgrowth by 21 days post cell transfer.

Figure 3.

OT-1 and OT-3 CD8 T cells have different thresholds of antitumor reactivity versus autoimmune diabetes. A, Adoptive cell transfer of 5 × 106 naïve OT-I CD8+ T cells, prepared by negative selection from donor transgenic OT-I mice splenocytes and LNs, mediates destruction of ID8-OVA tumors as reflected by decreased tumor bioluminescence. However, all recipient mice develop autoimmune diabetes. ACT of OT-3 CD8+ T cells, in contrast, mediates partial destruction of the incipient tumor without concomitant autoimmunity. B, ID8-OVA tumors that are transiently controlled by ACT of OT-3 CD8+ T cells demonstrate progressive tumor outgrowth by 21 days post cell transfer.

Close modal

OT-3 CTLs upregulate inhibitory receptors in response to ID8-OVA tumors and become exhausted

We next investigated the functional status of OT-3 T cells from mice whose tumors had reinitiated progressive growth. To accomplish this, OT-3 T cells were isolated from tumor-draining LNs of treated mice and tested for their ability to produce effector cytokines in response to restimulation by their cognate peptide (SIINFEKL) alongside those primed by the same ID8-OVA tumor offered as an irradiated cellular vaccine. As shown in Fig. 4, whereas the OT-3 T cells were clearly detectable in the tumor-draining LN, they were unable to produce IFNγ in response to OVA257-264 (SIINFEKL) peptide, although those primed by the ID8 vaccine were able to do so. The OT-3 cells from tumor-bearing mice were nonetheless able to produce IFNγ following treatment with PMA/ionomycin, indicating that the functional defect was in TCR-mediated activation (Fig. 4A). These findings demonstrate that the growing tumor is able to induce a state of unresponsiveness, termed “exhaustion,” in the responding OT-3 T cells. Accordingly, the OT-3 cells adoptively transferred to mice bearing ID8-OVA tumors versus ID8-WT tumors, or mice vaccinated with irradiated ID8-OVA cells, demonstrated upregulation of the inhibitory coreceptors PD-1, Tim-3, and Lag-3 (Fig. 4B).

Figure 4.

OT-3 CD8 T cells upregulate inhibitory receptors in response to ID8-OVA tumors and become functionally exhausted. A, 5 × 106 OT-3 CD8+ T cells adoptively transferred to mice bearing mature ID8-OVA tumors and harvested from the tumor-draining LN 15 days after transfer are unable to produce IFNγ in response OVA257–264 (SIINFEKL) peptide restimulation, whereas OT-3 CD8+ T cells primed by an ID8 tumor cell vaccine that lacks persistence of the tumor cells in vivo demonstrate successfully SIINFEKL antigen-specific restimulation. B, OT-3 CD8+ T cells adoptively transferred to mice bearing ID8-OVA tumors and harvested from the tumor-draining LN 15 days after transfer demonstrate upregulation of the inhibitory coreceptors PD-1, Tim-3, and Lag-3, whereas mice bearing ID8-WT tumors or mice vaccinated with irradiated ID8-OVA cells did not upregulate exhaustion markers.

Figure 4.

OT-3 CD8 T cells upregulate inhibitory receptors in response to ID8-OVA tumors and become functionally exhausted. A, 5 × 106 OT-3 CD8+ T cells adoptively transferred to mice bearing mature ID8-OVA tumors and harvested from the tumor-draining LN 15 days after transfer are unable to produce IFNγ in response OVA257–264 (SIINFEKL) peptide restimulation, whereas OT-3 CD8+ T cells primed by an ID8 tumor cell vaccine that lacks persistence of the tumor cells in vivo demonstrate successfully SIINFEKL antigen-specific restimulation. B, OT-3 CD8+ T cells adoptively transferred to mice bearing ID8-OVA tumors and harvested from the tumor-draining LN 15 days after transfer demonstrate upregulation of the inhibitory coreceptors PD-1, Tim-3, and Lag-3, whereas mice bearing ID8-WT tumors or mice vaccinated with irradiated ID8-OVA cells did not upregulate exhaustion markers.

Close modal

Immune-checkpoint inhibitors reverse OT-3 CTL exhaustion but induce autoimmune diabetes

Inhibitory receptors on immune cells are pivotal regulators of immune escape in cancer (22). We have shown that adoptively transferred OT-3 cells demonstrate extensive coexpression of PD-1, LAG-3, and TIM-3, and we have documented functional exhaustion in SIINFEKL antigen restimulation experiments following adoptive cell transfer into RIP-mOVA mice bearing ID8-Ova tumors. T-cell exhaustion is a plausible mechanism for the transient tumor control elicited by OT-3 cells in recipient mice. We therefore next sought to determine if coadministration of immune-checkpoint inhibitors along with OT-3 cells could preclude exhaustion and potentiate the antitumor reactivity of the adoptively transferred T cells in RIP-mOVA mice bearing ID8-OVA tumors. In these experiments, 250 μg of anti–PD-1, anti-Lag3, anti-Tim3, or a combination of all 3 immune-checkpoint antibodies, was administered to the contralateral orbital sinus along with adoptive cell transfer of 5 × 106 OT-3 cell into ID8-OVA tumor–bearing RIP-mOVA mice. Coadministration of immune-checkpoint inhibitors did not demonstrate statistically significant changes in tumor killing versus controls (Fig. 5A). However, linear regression analysis of all treated animals from all groups demonstrated that tumor killing did correlate with blood glucose concentration in treated mice (Fig. 5B). The highest blood glucose and tumor killing responses were all observed in the combinatorial immune-checkpoint cohort that received OT-3 cells in addition to PD-1, Lag-3, and Tim-3 inhibitors. In this group, half of the treated mice developed autoimmune diabetes with a trend toward greater tumor killing (Fig. 6). In addition, OVA antigen restimulation of splenocytes harvested from treated mice showed higher responses in the diabetic versus nondiabetic mice in this group. These data suggest that combinatorial ICB can preclude functional exhaustion in adoptively transferred OT-3 cells with a trend toward enhanced tumor killing but at the expense of developing autoimmune diabetes.

Figure 5.

Cotransfer of OT-3 CD8+ T cells with the immune-checkpoint inhibitors anti–PD-1, anti-TIM3, and anti-LAG3 elicits autoimmune diabetes that correlates with antitumor responses. A, Intravenous coadministration of the immune-checkpoint inhibitors anti–PD-1, anti-TIM3, and anti-LAG3 (250 μg) with 5 × 106 OT-3 CD8+ T cells into RIP-mOVA mice bearing 3-week-old ID8-OVA tumors did not demonstrate statistically significant changes in tumor killing by tumor bioluminescence by day 7 after ACT as single agents or in combination versus mice that received only OT-3 CD8+ T cells. B, Linear regression analysis of all treatment conditions demonstrated that tumor killing correlates with blood glucose concentration in treated mice.

Figure 5.

Cotransfer of OT-3 CD8+ T cells with the immune-checkpoint inhibitors anti–PD-1, anti-TIM3, and anti-LAG3 elicits autoimmune diabetes that correlates with antitumor responses. A, Intravenous coadministration of the immune-checkpoint inhibitors anti–PD-1, anti-TIM3, and anti-LAG3 (250 μg) with 5 × 106 OT-3 CD8+ T cells into RIP-mOVA mice bearing 3-week-old ID8-OVA tumors did not demonstrate statistically significant changes in tumor killing by tumor bioluminescence by day 7 after ACT as single agents or in combination versus mice that received only OT-3 CD8+ T cells. B, Linear regression analysis of all treatment conditions demonstrated that tumor killing correlates with blood glucose concentration in treated mice.

Close modal
Figure 6.

Combinatorial immune-checkpoint blockade with anti–PD-1, anti-TIM3, and anti-LAG3 reverses OT-3 CD8+ T-cell exhaustion with a trend to enhanced tumor killing but induces autoimmune diabetes. A, Intravenous coadministration of anti–PD-1, anti-TIM3, and anti-LAG3 (250 μg) with 5 × 106 OT-3 CD8+ T cells into RIP-mOVA mice bearing 3-week-old ID8-OVA tumors induced diabetes in half of the treated mice. B, There is a nonstatistically significant trend toward greater tumor killing in mice that developed autoimmune diabetes. C, SIINFEKL restimulation of splenocytes harvested from treated mice showed higher responses in the diabetic mice versus nondiabetic mice in this group.

Figure 6.

Combinatorial immune-checkpoint blockade with anti–PD-1, anti-TIM3, and anti-LAG3 reverses OT-3 CD8+ T-cell exhaustion with a trend to enhanced tumor killing but induces autoimmune diabetes. A, Intravenous coadministration of anti–PD-1, anti-TIM3, and anti-LAG3 (250 μg) with 5 × 106 OT-3 CD8+ T cells into RIP-mOVA mice bearing 3-week-old ID8-OVA tumors induced diabetes in half of the treated mice. B, There is a nonstatistically significant trend toward greater tumor killing in mice that developed autoimmune diabetes. C, SIINFEKL restimulation of splenocytes harvested from treated mice showed higher responses in the diabetic mice versus nondiabetic mice in this group.

Close modal

The current landscape of clinical cancer immunotherapy involves strategies aimed at both uncoupling a patient's T cells from negative regulation and the adoptive transfer of those which have been expanded in vitro and in some settings, engineered to acquire antitumor specificity (23–25). In each of these settings, the risk of pathologic autoimmunity must be balanced with the therapeutic potential inherent in trying to direct powerful effectors exclusively toward the tumor and its antigens. The model system established and described in this work represents an effort to explore these dichotomies in the context of varying TCR affinity in a controlled preclinical setting to better understand their causes and, thereby, to inform clinical practice.

Our results clearly show that T cells possessing an antigen receptor of sufficiently low functional avidity against a normal self-antigen to escape central tolerance can, nonetheless, be activated when the antigen in question is expressed on a growing tumor. In many key aspects, the OT-3 T cells in RIP-mOVA mice can be considered as a surrogate for the postthymic selection peripheral repertoire of potentially self-reactive T cells, which exist in a state of “immunologic ignorance” with respect to their cognate antigen, undergoing neither activation nor tolerance until some crucial aspect of their regulation is disrupted to enable their full activation. In the present study, this transition could be (unsurprisingly) induced by a strong inflammatory challenge with an OVA-expressing bacterial pathogen, or by a progressively growing tumor, with the latter leading to both proliferation and acquisition of effector functionality, producing a transient reduction in tumor volume. Remarkably, activation of the OT-3 T cells under these conditions did not lead to autoimmune diabetes, despite OVA expression on beta cells, suggesting that a window of opportunity exists for the selective targeting of a tumor by self-reactive T cells without concomitant recognition of a self-tissue sharing expression of the same antigen. These results do not exclude clinically subapparent toxicity, because elevated serum glucose concentration is typically detectable once 90% of insulin-producing cells are destroyed. The basis for this selective recognition of tumor over self is unclear, but could result from differences in the level of antigen presented by the 2 target cell populations or the local regulatory environment which may govern access and functional recognition of the OVA antigen in 2 sites. It is worth noting that, in light of the previously stated ignorance of OVA as an antigen by the OT-3 cells, the OVA257–264/Kb epitope is functionally equivalent to a foreign antigen for the OT-3 T cells, albeit one they recognize with low affinity. In the context of their selective recognition of the ID8-OVA tumor cells, OVA257–264/Kb functions as a tumor-specific neoantigen. In this regard, our findings suggest that such low-affinity T cells, whether induced by vaccination or provided through adoptive transfer, can be an effective therapeutic force for eradicating established tumors.

Although the low-affinity OT-3 T cells could be selectively activated by tumor-derived OVA and mediate some degree of tumor killing, the effect was temporary, and the population eventually underwent functional silencing accompanied by coordinated upregulation of immune-checkpoint molecules PD-1, TIM-3, and LAG-3. In this regard, the low-affinity T cells behaved as numerous reports have demonstrated for the endogenous tumor-reactive T-cell repertoire (26). Attempts to overcome this “exhausted” state through antibody-mediated blockade of the relevant receptors restored antigen-specific production of the effector cytokine IFNγ, by OT-3 T cells, but the ensuing response invariably led to diabetes induction in the majority of treated animals in the current study. These findings demonstrate that this model system can allow an unprecedented degree of insight into the hazards implicit in systemic blockade of immune-checkpoint pathways when a self-reactive T-cell population has been activated, with autoimmune diabetes being the primary immune-mediated adverse event (irAE) observed (27).

A number of studies have demonstrated the existence of optimal TCR “affinity windows” for key parameters of T-cell function, including proliferation, antigen sensitivity, and polyfunctionality (28), above which signaling can render cells hyporesponsive to low amount of antigen and/or mediate dangerous cross-reactivity to self MHC (29–31). These findings add to the growing consensus that TCRs with low to moderate affinities may possess important functional features necessary for sustained signaling to physiologic concentrations of antigen (32, 33). This notion is supported by studies by Sherman and colleagues, who showed that vaccination against an antigen shared between tumor and self-tissue can induce low-avidity antigen-specific CD8+ T cells to reject tumor cells with high expression of target antigen yet remain tolerant of antigen-expressing pancreatic beta cells (34). In adoptive cell transfer experiments conducted by this group, low-avidity antigen-specific T cells that were tolerized by cross-presented tumor antigen could subsequently eradicate tumors if antigen-specific CD4+ T cells were cotransferred into tumor-bearing mice harboring the target antigen, lending further evidence that such “affinity-tuned” cytotoxic T cells can be exploited therapeutically (35, 36).

This study reports that under conditions of steady-state progressive growth, a low-affinity TCR can preferentially be primed to and recognize antigen on tumor without causing autoimmune pathology to a normal self-tissue sharing its expression, whereas a high-affinity TCR was unable to discriminate between the 2 types of target cells. This suggests that, whether due to the amount of antigen produced or the homeostatic state of the tissue in question, the tumor environment is distinct from that of a stable self-tissue in terms of the magnitude and context of antigen (cross-)presentation and that only a low-affinity TCR was capable of responding to this. This finding suggests that further investigation of low-affinity TCRs could further elucidate the factors that govern the functionality in the setting of ACT against shared tumor/self-antigens and in doing so augment the precision and efficacy of ACT.

E.E.W. Cohen is a consultant/advisory board member for Pfizer, Merck, AstraZeneca, Bristol-Myers Squibb, EMD Serono, and Amgen. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A.M. Miller, E.E.W. Cohen, S.P. Schoenberger

Development of methodology: A.M. Miller, M. Bahmanof

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A.M. Miller, M. Bahmanof

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A.M. Miller, E.E.W. Cohen, S.P. Schoenberger

Writing, review, and/or revision of the manuscript: A.M. Miller, D. Zehn, E.E.W. Cohen, S.P. Schoenberger

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A.M. Miller, M. Bahmanof, S.P. Schoenberger

Study supervision: S.P. Schoenberger

This work was supported by 1R21CA179362 and 1R01AI107010 from the NIH to S.P. Schoenberger and from a gift from the Kimmelman Family Foundation.

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.
Hinrichs
CS
,
Rosenberg
SA
. 
Exploiting the curative potential of adoptive T-cell therapy for cancer
.
Immunol Rev
2014
;
257
:
56
71
.
2.
Lizee
G
,
Overqijk
WW
,
Radvanyi
L
,
Gao
J
,
Sharma
P
,
Hwu
P
. 
Harnessing the power of the immune system to target cancer
.
Annu Rev Med
2013
;
64
:
71
90
.
3.
Fesnak
AD
,
June
CH
,
Levine
BL
. 
Engineered T cells: the promise and challenges of cancer immunotherapy
.
Nat Rev Cancer
2016
;
16
:
566
81
.
4.
Lim
WA
,
June
CH
. 
The principles of engineering immune cells to treat cancer
.
Cell
2017
;
168
:
724
40
.
5.
Rosenberg
SA
,
Yang
JC
,
White
DE
,
Steinberg
SM
. 
Durability of complete responses in patients with metastatic cancer treated with high-dose interleukin-2: identification of the antigens mediating response
.
Ann Surg
1998
;
228
:
307
19
.
6.
Rosenberg
SA
,
Yang
JC
,
Sherry
RM
,
Kammula
US
,
Hughes
MS
,
Phan
GQ
, et al
Durable complete responses in heavily pretreated patients with metastatic melanoma
.
Clin Cancer Res
2011
;
17
:
4550
7
.
7.
Stevanovic
S
,
Draper
LM
,
Langhan
MM
,
Campbell
TE
,
Kwong
ML
,
Wunderlich
JR
, et al
Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells
.
J Clin Oncol
2015
;
33
:
1543
50
.
8.
Turcotte
S
,
Gros
A
,
Tran
E
,
Lee
CC
,
Wunderlich
JR
,
Robbins
PF
, et al
Tumor-reactive CD8+ T cells in metastatic gastrointestinal cancer refractory to chemotherapy
.
Clin Cancer Res
2014
;
20
:
331
43
.
9.
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
.
10.
Obenaus
M
,
Leitao
C
,
Leisegang
M
,
Chen
X
,
Gavvovidis
I
,
van der Bruggen
P
, et al
Identification of human T-cell receptors with optimal affinity to cancer antigens using antigen-negative humanized mice
.
Nat Biotechnol
2015
;
33
:
402
7
.
11.
Kunert
A
,
Straetemans
T
,
Govers
C
,
Lamers
C
,
Mathijssen
R
,
Sleijfer
S
, et al
TCR-engineered T cells meet new challenges to treat solid tumors: choice of antigen, T cell fitness, and sensitization of tumor milieu
.
Front Immunol
2013
;
4
:
363
.
12.
Schmitt
TM
,
Stromnes
IM
,
Chapuis
AG
,
Greenberg
PD
. 
New strategies in engineering T-cell receptor gene-modified T cells to more effectively target malignancies
.
Clin Cancer Res
2015
;
21
:
5191
7
.
13.
Draper
LM
,
Kwong
ML
,
Gros
A
,
Stevanovic
S
,
Tran
E
,
Kerkar
S
, et al
Targeting of HPV-16+ epithelial cancer cells by TCR engineered T cells directect against E6
.
Clin Cancer Res
2015
;
21
:
4431
9
.
14.
Morgan
RA
,
Chinnasamy
N
,
Abate-Daga
D
,
Gros
A
,
Robbins
PF
,
Zheng
Z
, et al
Cancer regression and neurologic toxicity following anti-MAGE-A3 TCR gene therapy
.
J Immunother
2014
;
36
:
133
51
.
15.
Parkhurst
MR
,
Yang
JC
,
Langan
RC
,
Dudley
ME
,
Nathan
DA
,
Feldman
SA
, et al
T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis
.
Mol Ther
2011
;
19
:
620
6
.
16.
Linette
GP
,
Stadtmauer
EA
,
Maus
MV
,
Rapoport
AP
,
Levine
BL
,
Emery
L
, et al
Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma
.
Blood
2013
;
122
:
863
71
.
17.
Robbins
PF
,
Kassim
SH
,
Tran
TL
,
Crystal
JS
,
Morgan
RA
,
Feldman
SA
, et al
A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response
.
Clin Cancer Res
2015
;
21
:
1019
27
.
18.
Enouz
S
,
Carrie
L
,
Merkler
D
,
Bevan
MJ
,
Zehn
D
. 
Autoreactive T cells bypass negative selection and respond to self-antigen stimulation during infection
.
J Exp Med
2012
;
209
:
1769
79
.
19.
Ohlen
C
,
Kalos
M
,
Hong
DJ
,
Shur
AC
,
Greenberg
PD
. 
Expression of a tolerizing tumor antigen in peripheral tissue does not preclude recovery of high-affinity CD8+ T cells or CTL immunotherapy of tumors expressing the antigen
.
J Immunol
2001
;
166
:
2863
70
.
20.
Roby
KF
,
Taylor
CC
,
Sweetwood
JP
,
Cheng
Y
,
Pace
JL
,
Tawfik
O
, et al
Development of a syngeneic mouse model for events related to ovarian cancer
.
Carcinogenesis
2000
;
21
:
585
91
.
21.
Sanderson
S
,
Shastri
N
. 
LacZ inducible, antigen/MHC-specific T cell hybrids
.
Int Immunol
1994
;
6
:
369
76
.
22.
Beatty
GL
,
Gladeny
WL
. 
Immune escape mechanisms as a guide for cancer immunotherapy
.
Clin Cancer Res
2015
;
21
:
687
92
.
23.
Callahan
MK
,
Postow
MA
,
Wolchok
JD
. 
Targeting T cell co-receptors for cancer therapy
.
Immunity
2016
;
44
:
1069
78
.
24.
Yang
JC
,
Rosenberg
SA
. 
Adoptive T-cell therapy for cancer
.
Adv Immunol
2016
;
130
:
279
94
.
25.
Lim
WA
,
June
CH
. 
The principles of engineering immune cells to treat cancer
.
Cell
2017
;
168
:
724
40
.
26.
Schietinger
A
,
Greenberg
PD
. 
Tolerance and exhaustion: defining mechanisms of T cell dysfuction
.
Trends Immunol
2014
;
35
:
51
60
.
27.
Postow
MA
,
Sidlow
R
,
Hellman
MD
. 
Immune-related adverse events associated with immune checkpoint blockade
.
N Engl J Med
2018
;
378
:
158
68
.
28.
Hebeisen
M
,
Allard
M
,
Gannon
PO
,
Schmidt
J
,
Speiser
DE
,
Rufer
N
. 
Identifying individual T cell receptors of optimat avidity for tumor antigens
.
Front Immunol
2015
;
6
:
582
.
29.
Tan
MP
,
Gerry
AB
,
Brewer
JE
,
Melchiori
L
,
Bridgeman
JS
,
Bennet
AD
, et al
T cell receptor binding affinity governs the functional profile of cancer-specific CD8+ T cells
.
Clin Exp Immunol
2015
;
180
:
255
70
.
30.
Aleksic
M
,
Liddy
N
,
Molloy
PE
,
Pumphrey
N
,
Vuidepot
A
,
Chang
KM
, et al
Different affinity windows for virus and cancer-specific T-cell receptors: implications for therapeutic strategies
.
Eur J Immunol
2012
;
42
:
3174
79
.
31.
Thomas
S
,
Xue
SA
,
Bangham
CR
,
Jakobsen
BK
,
Morris
EC
,
Stauss
HJ
. 
Human T cells expressing affinity-matured TCR display accelerated responses but fail to recognize low density of MHC-peptide antigen
.
Blood
2011
;
118
:
319
29
.
32.
Martinez
RJ
,
Evavold
BD
. 
Lower affinity T cells are critical components and active participants of the immune response
.
Front Immunol
2015
;
6
:
468
.
33.
Hebeisen
M
,
Baitsch
L
,
Presotto
D
,
Baumgaertner
P
,
Romero
P
,
Michielin
O
, et al
SHP-1 phosphatase activity counteracts increased T cell receptor affinity
.
J Clin Invest
2013
;
123
:
1044
56
.
34.
Morgan
DJ
,
Kreuwel
HT
,
Fleck
S
,
Levitsky
HI
,
Pardoll
DM
,
Sherman
LA
, et al
Activation of low avidity CTL specific for a self epitope results in tumor rejection but not autoimmunity
.
J Immunol
1998
;
160
:
643
.
35.
Lyman
MA
,
Aung
S
,
Biggs
JA
,
Sherman
LA
. 
A spontaneously arising pancreatic tumor does not promote the differentiation of naive CD8+ T lymphocytes into effector CTL
.
J Immunol
2004
;
172
:
6558
.
36.
Wong
SBJ
,
Bos
R
,
Sherman
LA
. 
Tumor-specific CD4+ T cells render the tumor environment permissive for infiltration by low-avidity CD8+ T Cells
.
J Immunol
2008
;
180
:
3122
.