Peptide vaccines can be a successful and cost-effective way of generating T-cell responses against defined tumor antigens, especially when combined with immune adjuvants such as poly-IC. However, strong immune adjuvants can induce a collateral increase in numbers of irrelevant, nonspecific T cells, which limits the effectiveness of the peptide vaccines. Here, we report that providing prolonged IL2 signaling in the form of either IL2/anti-IL2 complexes or pegylated IL2 overcomes the competitive suppressive effect of irrelevant T cells, allowing the preferential expansion of antigen-specific T cells. In addition to increasing the number of tumor-reactive T cells, sustained IL2 enhanced the ability of T cells to resist PD-1–induced negative signals, increasing the therapeutic effectiveness of the vaccines against established tumors. This vaccination strategy using peptides and sustained IL2 could be taken into the clinic for the treatment of cancer. Cancer Immunol Res; 6(5); 617–27. ©2018 AACR.

Cancer therapy has been changing over the past few years. Cancer immunotherapy is becoming the first-line treatment for many cancer types (1, 2). T cells and, in particular, CD8+ cytotoxic T lymphocytes, are the most effective component of the immune system capable of destroying tumor cells. Thus, most cancer immunotherapy strategies focus on unleashing T-cell responses capable of eliminating the malignant cells. However, in many instances, tumor-reactive T cells are too few or lack the necessary functionality to eradicate established tumors (3, 4).

Adoptive cell transfer (ACT) of tumor-reactive T cells is a promising approach to provide a large numbers of tumor-reactive T cells. ACT alongside host lymphodepletion has shown successes in the clinic. However, this approach is patient specific, technically challenging, and not very cost-effective (5). Active immunization (i.e., therapeutic cancer vaccines) is a more practical way of generating tumor-reactive T cells. Although cancer vaccines have shown initial promising results in the clinic and with mouse tumor models, their immunogenicity has been suboptimal with questionable long-term therapeutic benefits (6). These disappointing results can be attributed to the low number of tumor-reactive T cells generated by the vaccines. In addition, the existence of an immunosuppressive tumor microenvironment will likely neutralize the effectiveness of the few T cells generated by suboptimal vaccines. In particular, tumor cells and some tumor-infiltrating cells upregulate T-cell checkpoint inhibitory ligands such as programmed cell death ligand-1 (PD-L1; ref. 7). Indeed, blockade of this checkpoint inhibitory signal with PD-1 or PD-L1 monoclonal antibodies (mAb) has shown therapeutic responses in the clinic (8, 9). A cost-effective vaccine that generates substantial antitumor T-cell responses capable of resisting the immunosuppressive tumor microenvironment could defeat or control cancer.

We have developed peptide-based vaccines in mice capable of generating large T-cell responses that limit growth of established tumors (10–17). However, to obtain desirable therapeutic responses with PD-L1–expressing tumors, the vaccines required concurrent PD1 checkpoint blockade using anti–PD-1 or anti–PD-L1 mAbs. Here, we report that administration of IL2 after peptide vaccination increased the expansion of antigen-specific T cells and rendered them resistance to PD-L1 inhibition, resulting in enhanced antitumor effects. In order to obtain the therapeutic effects of IL2, the cytokine had to be administered either as IL2/anti-IL2 complexes (IL2Cx) or pegylated IL2 (PEG-IL2), both of which prolong the half-life of IL2 and improve T-cell responses by a sustained stimulation of the IL2 receptor complex (18–23). Overall, these results indicate that sustained IL2 administration in the form of IL2Cx or PEG-IL2 enhances the quantity and quality of tumor-reactive T-cell responses following peptide-based vaccines.

Mice and cell lines

C57BL/6 mice (WT), B6-Ly5.1 mice (CD45.1 WT), and BALB/c mice (6-to-8-week-old females and males) were purchased from the National Cancer Institute/Charles River program. B6.129S7-RAG1tm1Mom/J (RAG-KO) and pmel-1 mice were purchased from The Jackson Laboratory. TCR transnuclear mice (TgTR1) that recognize MHC class I–restricted Trp1455-463 peptide in a RAG-KO background were previously described (24) and bred on site at the Georgia Cancer Center animal facility. All experiments followed guidelines of the Committee on the Care of Laboratory Animals Resources, Commission of Life Sciences, and National Research Council. The animal facility at the Georgia Cancer Center is fully accredited by the Association for Assessment and Accreditation of Laboratory Animal Care. Mouse lymphoma cell line EL4 and B16 F10 melanoma both expressing MHC class I (Db/Kb) were purchased from the American Type Culture Collection and maintained as recommended by the vendor. The TUBO (Turin-Bologna) tumor is a cloned cell line established in vitro from a lobular carcinoma that arose spontaneously in a BALB-neuT mouse (25). The rat HER2/neu-transfected mouse mammary breast cancer (A2L2; H-2d) and its parental cell line (66.3; ref. 26) were kind gifts from Drs. J.E. Price and L. Lachman (MD Anderson Cancer Center, Houston, TX). All cells were maintained in culture for less than 4 weeks continuously, and fresh frozen stocks were used at the initiation of each experiment.

Synthetic peptides and reagents

All peptides were purchased from Synthetic Biomolecules. Stock solutions (20 mg/mL) were reconstituted in DMSO with 0.1% trifluoroacetic acid and maintained at −80 C. As determined by high-performance liquid chromatography, the peptides were >80% pure. Peptides and dipalmitoylated (pam2) peptides used in this study were as follows: TAPDNLGYM (Trp1455-463/9M), (pam2)-KMFVTAPDNLGYM (Pam-Trp1), and (pam2)-KMVE-SIINFEKL (Pam-OVA). BALB/c (H2d) CD8 T-cell epitopes include rNEU66-74, TYVPANASL; Pam-rNEU, (pam2)-KMVETYVPANASL; LLO91-99, GYKDGNEYI; and Pam-LLO (pam2)-KMVEGYKDGNEYI). Poly-IC (high molecular weight) was purchased from InvivoGen (cat. #tlrl-pic-5). Poly-ICLC was provided by Dr. Andres Salazar (Oncovir). mAb against mouse CD40 (FGK4.5, cat. #BE0016-2), mouse PD-L1 (10F.9G2, cat. #BE0101), mouse IL2 (JES6-5H4, cat. #BE0042 JES6-1A12, cat. #BE0043), mouse CD4 (GK1.5, cat. #BE0003-1), mouse CD122 (TM-beta 1, cat #BE0298), mouse CD25 (PC-61.5.3, cat #BE00012), and mouse Thy1.1 (19E12, cat. #BE0214) were purchased from BioXcell. Mouse recombinant IL2 was from BioLegend (5 million IU/mg; cat. #575408).

Preparation of IL2/IL2 mAb complexes (IL2Cx)

For each dose of IL2Cx, 2 μg IL2 were mixed with 10 μg IL2 mAb JES6-5H4 (IL2Cx122) or JES6-1A12 (IL2Cx25) and incubated overnight at 10°C. IL2Cx were administered i.p. Human IL2Cx (huIL2Cx) was made from recombinant human IL2 (2 μg/mouse, PeproTech, 10 million IU/mg; cat. #200-02) and human IL2 mAb (10 μg, R&D Systems, cat. #MAB602).

Preparation of pegylated IL2 (PEG-IL2)

Human IL2 was incubated with O-[2-N-Succinimidyloxycarbonyl-ethyl]-O′-methylpolyethylene-glycol (PEG; 500 mw, Sigma-Aldrich, cat. #85969-1G) at a 50:1 molar ratio for 2.5 hours at pH 9.3. The pegylation efficiency was confirmed by gel electrophoresis staining with Blue Code gel stain reagent (Thermo Fisher Scientific, cat. #24590). The efficiency of pegylation was ∼70%. In all experiments, 4 μg PEG-IL2 or the equivalent amount of free IL2/mouse was used.

Immunization protocols

Mice were injected i.v. in a prime–boost protocol with BiVax (120 μg of pam2-peptide and 50 μg of poly-IC) 12 or 5 days apart. Mouse CD40 mAb (100 μg/mouse) was combined with BiVax in the indicated group (TriVax). IL2, IL2Cx25, IL2Cx122, or PEG-IL2 were injected i.p. on days 12, 14, and 16 (or days 5, 7, and 9 for the quick prime–boost protocol). In some experiments, mice received ACT of purified CD8+ T cells from TgTR1 mice, which in some instances were transduced with a constitutively active form of STAT5 (CA-STAT5). Retrovirus (RV) encoding CA-STAT5 coexpressing CD90.1/Thy1.1 (as a marker) was provided by Dr. Gang Zhou (Augusta University) in collaboration with Dr. Susan Kaech (Yale University; ref. 27). For T-cell transduction CD8 T cells isolated from TgTR1 mice (Thy1.2) were activated for 48 hours with mouse dynabeads T-Activator CD3/CD28 beads (Thermo Fisher Scientific, cat. #11456D) and then were transduced with the RV using Retronectin (Takara Bio, cat. #T100B)-coated plates for 10 minutes. The transduction efficiency was ∼50% as evaluated by Thy1.1 expression 48 hours after transduction. CD4 mAb (250 μg/mouse), Thy1.1 mAb (500 μg/mouse) were injected intraperitoneally on days −2, 0, 10, and 12 in the indicated groups to deplete CD4+ T cells or Thy1.1+ cells, respectively.

Flow cytometry and the evaluation of immune responses

For measuring antigen-specific CD8+ T-cell responses, peripheral blood samples or splenocytes were stained with: PE-labeled tetramers Trp1455-463/H-2Db, rNEUp66-74/H2-Kd or LLO91-99/H2-Kd (kindly provided by the NIH Tetramer Core Facility, Emory University, Atlanta, GA). Samples were stained with FITC-labeled MHC class II mAb (BioLegend, cat. #107606) for negative gating and PerCP/Cy5.5 CD8 mAb (BioLegend, cat. #100734) for positive gating. To stain for phospho-STAT5 (pSTAT5), splenocytes were fixed in 1.6% formaldehyde for 10 minutes at room temperature, followed by surface staining in PBS with FITC-labeled anti-CD45.2 (BioLegend, cat. #109805) for 10 minutes. Cells were permeabilized with chilled methanol at −20°C for 30 minutes followed by 3× washing with PBS. The intracellular pSTAT5 staining was performed using PE-labeled anti–phospho-STAT5 (PY694, BD Biosciences, cat. #562077) for 20 minutes at room temperature. All other fluorescent-labeled antibodies were purchased from eBioscience and BioLegend. Flow cytometry was performed using LSRII cytometers (BD Biosciences). Data analysis was performed using FlowJo software (version 8.5, TreeStar).

ELISpot assay

IFNγ ELISpot assays were performed as previously described (10–14). Briefly, CD8+ T cells were purified from spleens of vaccinated mice using CD8+ selection kits (Miltenyi Biotec, cat. #130-049-401). Effector cells were incubated at different numbers together with 1 × 105 stimulator cells. In some experiments, B16F10 were treated with IFNγ (100 ng/mL, PeproTech, cat. #315-05) and pulsed with 1 μg/mL Trp1455-463 for 48 hours. For in vitro PD-L1 blockade, 10 μg/mL of PD-L1 mAb was used. ImmunoSpot System was used to develop and count the cytokine-positive spots (Cellular Technology Ltd.).

Preparation of Trp1455-463/9M artificial antigen-presenting cells (aAPC)

aAPCs were prepared as described previously (28). Briefly, Dynabeads M-450 Epoxy (4 × 108/mL, Thermo Fisher Scientific, cat. #14011) were incubated with 10 μg/mL of Trp1455-463/9M H2Db monomers (NIH Tetramer Core Facility) for 24 hours, and half of the beads (Trp1/PD-L1 aAPCs) were incubated with 10 μg/mL of mouse Fc-Tag PD-L1 protein (AcroBiosystems, cat. #PD1-M5251) and the other half (Trp1/aAPCs) with BSA for an additional 24 hours. Finally, beads were washed twice with PBS and then incubated with 0.2% BSA for 24 hours washed and resuspended in culture media.

TgTR1 CD8+ T cells were labeled with CFSE (1 μmol/L, Thermo Fisher Scientific, cat. #C34554) and cultured in the presence of various concentrations of aAPCs with or without PD-L1 mAb (10 μg/mL) for 72 hours. After the incubation, the CFSE dilution (proliferation) was assessed by flow cytometry.

In vivo tumor model

B16F10 melanomas model.

WT mice (10 mice/group) were inoculated s.c. with B16F10 cells (3 × 105/mouse). Tumor growth was monitored every 2 to 3 days in individual tagged mice by measuring 2 opposing diameters with a set of calipers.

RNEU antitumor model.

For therapeutic vaccination assessment, BALB/c mice were inoculated subcutaneously with A2L2 cells (5 × 105/mouse) 10 days before immunization using BiVax with or without IL2Cx. For prophylactic antitumor assessments BALB/c mice were first immunized with rNEU-BiVax/IL2Cx25 and 50 days later, the immunized or naïve mice were subcutaneously challenged with A2L2 cells (5 × 105/mouse). Nonvaccinated mice were included as controls. Tumor growth was monitored every 2 to 3 days in individual tagged mice by measuring 2 opposing diameters with a set of calipers. Results are presented as the mean tumor size (area in mm2) ± SD for every treatment group at various time points until the termination of the experiment (usually when tumor size reached 20 mm diameter).

Statistical analysis

Statistical significance was determined by unpaired Student's t tests or one-way ANOVA. Tumor sizes between 2 populations throughout time were analyzed for significance using two-way ANOVA. All analyses and graphics were done using Prism 6 software (GraphPad). Results are presented as mean ± SD (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001, and ns: not significant). All experiments were repeated at least twice to ensure reproducibility.

IL2Cx overcomes inhibitory effects of irrelevant CD8+ T cells

A challenge that limits the effectiveness of peptide epitope-based vaccines relates to their capacity to specifically stimulate and expand antigen-specific, tumor-reactive T cells. To generate such responses, peptide vaccines must contain adjuvants (e.g., TLR agonists) and/or costimulatory antibodies (e.g., CD40 mAb). In addition, the vaccine's effective recruitment of the usually low numbers of naïve T cells capable of recognizing the peptide epitope will determine the magnitude of the T-cell expansion. For this reason, we have utilized a systemic (intravenous, i.v.) mode of vaccine administration, which has been proven to be superior to local s.c. immunization for generating T-cell responses (10, 13). However, we have observed that the systemic administration of the vaccine, adjuvant, and costimulatory antibodies can lead to the activation and increase in numbers of nonantigen specific (i.e., irrelevant) T cells (Fig. 1A), which could diminish the effectiveness of the vaccines by competing with antigen-specific T cells for “immune resources” (APCs and cytokines).

Figure 1.

Irrelevant CD8+ T-cell inhibition of antigen-specific CD8+ T-cell responses to peptide vaccines is reversed by IL2Cx. A, WT mice were injected with Trp1-BiVax or Trp1-Trivax. One week later, the total numbers of antigen-specific (tetramer+) and tetramer CD8+ T cells were evaluated in spleens. B, WT or RAG1-KO mice were adoptively transferred with CD8+ T cells from TgTR1 mice (2,000 cells/mouse) with or without WT CD8+ T cells (8 × 106/mouse). One and 12 days later, mice were vaccinated with Trp1-BiVax. On day 19, the total numbers of TgTR1 cells were evaluated in spleens. C, Pmel-1 mice (Thy1.1+, RAG+) received 2,000 TgTR1 cells (Thy1.2+) followed by two Trp1-BiVax vaccinations 12 days apart. In some mice, Thy1.1+ cells (all T cells) or CD4+ T cells were depleted using mAb, and one group of mice was injected with IL2Cx122 on days 12, 14, and 16. The total numbers of TgTR1 cells (Tetramer+ T cells) were evaluated in spleens on day 19. D, WT mice were vaccinated as shown and the percentage of Trp1 tetramer+ T cells was evaluated in the blood 7 days after each vaccination. E, Total numbers of tetramer+ T cells in spleen, 8 days after the boost. F, Purified spleen CD8+ T cells (105) were incubated with B16F10 cells overnight and the numbers of IFNγ producing cells were evaluated using ELISpot. Representative results are shown from at least 3 independent experiments (n = 3 mice/group).

Figure 1.

Irrelevant CD8+ T-cell inhibition of antigen-specific CD8+ T-cell responses to peptide vaccines is reversed by IL2Cx. A, WT mice were injected with Trp1-BiVax or Trp1-Trivax. One week later, the total numbers of antigen-specific (tetramer+) and tetramer CD8+ T cells were evaluated in spleens. B, WT or RAG1-KO mice were adoptively transferred with CD8+ T cells from TgTR1 mice (2,000 cells/mouse) with or without WT CD8+ T cells (8 × 106/mouse). One and 12 days later, mice were vaccinated with Trp1-BiVax. On day 19, the total numbers of TgTR1 cells were evaluated in spleens. C, Pmel-1 mice (Thy1.1+, RAG+) received 2,000 TgTR1 cells (Thy1.2+) followed by two Trp1-BiVax vaccinations 12 days apart. In some mice, Thy1.1+ cells (all T cells) or CD4+ T cells were depleted using mAb, and one group of mice was injected with IL2Cx122 on days 12, 14, and 16. The total numbers of TgTR1 cells (Tetramer+ T cells) were evaluated in spleens on day 19. D, WT mice were vaccinated as shown and the percentage of Trp1 tetramer+ T cells was evaluated in the blood 7 days after each vaccination. E, Total numbers of tetramer+ T cells in spleen, 8 days after the boost. F, Purified spleen CD8+ T cells (105) were incubated with B16F10 cells overnight and the numbers of IFNγ producing cells were evaluated using ELISpot. Representative results are shown from at least 3 independent experiments (n = 3 mice/group).

Close modal

To investigate the negative role of the activation and increase of irrelevant T cells in the responses of antigen-specific T cells to a systemic peptide vaccine (BiVax), we adoptively transferred a low number (2,000 cells) of naïve antigen-specific CD8+ T cells from TCR transnuclear mice (24) specific for the Trp1455-463 epitope (TgTR1) into wild-type C57BL/6 (WT) mice or immune deficient RAG-KO mice (lacking irrelevant T cells). After a prime–boost Trp1-BiVax immunization (12 days apart) the TgTR1 cells expanded ∼500 fold in WT mice and 7,500-fold in the RAG-KO mice (Fig. 1B). Coinjection of a large number (8 million) of naïve CD8+ T cells from WT mice decreased the expansion of the TgTR1 T cells by 50% in the RAG-KO mice (Fig. 1B). When 2,000 TgTR1 T cells were transferred into Pmel-1 TCR transgenic mice containing T cells with a restricted TCR repertoire specific for an irrelevant antigen epitope (gp10025-33, expressing Thy1.1+/RAG+), Trp1-BiVax elicited an expansion of the TgTR1 of a similar magnitude to that observed in RAG-KO mice (Fig. 1C). T-cell depletion by Thy1.1 mAb increased the expansion of the TgTR1 T cells by 300%, but depletion of CD4+ T cells (∼20% of the T-cell repertoire in Pmel-1 mice) using CD4 mAb had no effect. Together, these results indicate that activation and expansion of irrelevant CD8+ T cells are responsible for diminishing the expansion of antigen-specific T cells resulting from systemic peptide vaccination. The negative effect of the irrelevant CD8+ T cells is likely due to competition for cytokines (IL2 and IL15) during the expansion phase of the response, because administration of IL2 in the form of IL2/anti-IL2 complexes (IL2Cx) right after the boost resulted in a roughly 200,000-fold expansion of the TgTR1 T cells (Fig. 1C).

Next, we studied the effect of IL2Cx in the endogenous T-cell response (without adoptive cell transfer) to Trp1455-463 BiVax. We have observed that the efficacy of BiVax is dependent on the formulation of poly-IC used in the vaccine. Although poly-IC formulated with poly-lysine and carboxymethylcellulose (poly-ICLC) can generate sizable immune responses, simple poly-IC was less effective (Supplementary Fig. S1). The activity of poly-IC was enhanced, however, by the addition of IL2Cx.

Thus, for the present studies, we utilized poly-IC to better appreciate the effects of IL2Cx. Two types of IL2Cx were tested: IL2Cx122 uses an IL2 mAb that directs the IL2 to the low-affinity receptor IL2Rβ/CD122, and IL2Cx25 preferentially directs IL2 to the high-affinity receptor IL2Rα/CD25 (18, 19). The addition of either IL2Cx122 or IL2Cx25 in the vaccine booster enhanced the magnitude of the endogenous CD8+ T-cell responses in the blood (Fig. 1D) and the spleen (Fig. 1E). Moreover, the CD8+ T cells generated by these vaccines were effective in recognizing Trp1-expressing tumor cells (B16 melanoma) as ascertained in an ELISpot assay (Fig. 1F). Collectively, these data demonstrate that extra cytokine in the form of IL2Cx enhanced expansion of endogenous or adoptive transferred T cells and overcame competition by irrelevant T cells.

Timing of IL2Cx administration critical for robust CD8+ T-cell responses

In the previous experiments, IL2Cx were administered with the vaccine booster (12 days after the priming immunization). Because rapid tumor growth in the B16 melanoma model would make it challenging to assess the antitumor effect of IL2Cx with peptide vaccines, we evaluated the use of IL2Cx after the priming vaccine. However, providing IL2Cx 1 day after priming with BiVax did not increase the numbers of antigen-specific cells (Fig. 2A). To decrease the time required to attain a large T-cell response, we examined the use of a 5-day interval (quick) prime–boost protocol (10). Administration of IL2Cx122 or IL2Cx25 with the BiVax booster vaccine 5 days after priming generated an endogenous T-cell response (Fig. 2B–D) of a similar magnitude to that observed with a 12-day interval immunization protocol (Fig. 1E). The inability of IL2Cx to enhance the immune response when given soon after the prime (Fig. 2A) could be due to the increase in the total numbers of CD8+ T cells (Fig. 2E), where approximately one half of these expressed CD44 (T-cell memory marker). Tetramer-negative CD8+/CD44+ T cells likely function as irrelevant T cells and compete with antigen-specific T cells for IL2. A similar vaccination protocol was also effective in generating large T-cell responses against the rNEU66-74 and LLO91-99 CTL epitopes in BALB/c mice, in which the rNEU66-74 reactive T cells efficiently recognized rNEU66-74-expressing tumor cells (Supplementary Fig. S2). The combination of recombinant human IL2 (hu-IL2) and anti-hu-IL2 (MAB602; hu-IL2Cx) was as effective as the mouse IL2Cx122 (m-IL2Cx) in expanding antigen-specific cells (Supplementary Fig. S3). Boosting with IL2Cx alone (no peptide) failed to increase the expansion of antigen-specific cells (Fig. 2F). Boosting with peptide and IL2Cx (without poly-IC) increased the numbers of antigen-specific cells. However, poly-IC was required for maximal T-cell expansion. Although IL2Cx administration increased Treg numbers in spleen, this did not appear to interfere with immune responses to the vaccine, perhaps because poly-IC reduced the expansion of Tregs generated by IL2Cx (Supplementary Fig. S4). Collectively, these results suggest that antigen-specific T cells should be previously activated and start to proliferate to properly expand after IL2Cx administration. Moreover, the fast prime–boost (5 days apart) vaccination protocol was sufficient to induce prompt, large, and robust T-cell responses.

Figure 2.

Shortened prime–boost BiVax/IL2Cx vaccine protocol generates large T-cell responses. A, WT mice were vaccinated as shown and the percentage of Trp1 tetramer+ T cells was evaluated in the blood 7 days after vaccination. B, Diagram of the vaccination protocol using WT mice and percentage of Trp1 tetramer+ cells in the blood at day 12. C, Representative flow cytometry dot plots showing the percentage of Trp1 tetramer+ CD8+ T cells in blood. D, Total numbers of Trp1 tetramer+ T cells in spleens on day 12. E, Absolute numbers of CD8+ T cells and activated (CD44+) CD8+ T cells after BiVax with and without IL2Cx. F, WT mice were vaccinated with BiVax prime and 5 days later mice were boosted with pamTrp1/poly-IC (BiVax), IL2Cx25 alone, pamTrp1/IL2Cx25, or pamTrp1/poly-IC/IL2Cx25 and on day 12 the numbers of Trp1 tetramer+ T cells were evaluated in spleens. (n = 3 mice/group). Representative results are shown from at least 3 independent experiments.

Figure 2.

Shortened prime–boost BiVax/IL2Cx vaccine protocol generates large T-cell responses. A, WT mice were vaccinated as shown and the percentage of Trp1 tetramer+ T cells was evaluated in the blood 7 days after vaccination. B, Diagram of the vaccination protocol using WT mice and percentage of Trp1 tetramer+ cells in the blood at day 12. C, Representative flow cytometry dot plots showing the percentage of Trp1 tetramer+ CD8+ T cells in blood. D, Total numbers of Trp1 tetramer+ T cells in spleens on day 12. E, Absolute numbers of CD8+ T cells and activated (CD44+) CD8+ T cells after BiVax with and without IL2Cx. F, WT mice were vaccinated with BiVax prime and 5 days later mice were boosted with pamTrp1/poly-IC (BiVax), IL2Cx25 alone, pamTrp1/IL2Cx25, or pamTrp1/poly-IC/IL2Cx25 and on day 12 the numbers of Trp1 tetramer+ T cells were evaluated in spleens. (n = 3 mice/group). Representative results are shown from at least 3 independent experiments.

Close modal

Antitumor effects of the peptide vaccine/IL2Cx combination

Next, we evaluated the effect of IL2Cx enhancing the antitumor therapeutic effect of Trp1-BiVax in mice with established subcutaneous B16 melanomas. Although Trp1-BiVax alone could reduce the rate of tumor growth and increase survival, the addition of IL2Cx122 or IL2Cx25 augmented the antitumor effects (Fig. 3A and B). The effects of the IL2Cx required an antigen-specific vaccine, because mice receiving an irrelevant peptide (Ova257-264) showed no significant therapeutic effect. Mice receiving Trp1-BiVax and IL2Cx controlled their tumors up to day 25, when all of the control mice (untreated or those receiving irrelevant vaccinations) had already perished due to advanced disease. The recurrence of tumor growth after day 25 in these mice was not the result of the disappearance of antigen-specific, tumor-reactive T cells (Fig. 3C) or due to the expression of the inhibitory receptor PD1 (Fig. 3D). Moreover, antigen-specific cells obtained at day 42 were able to recognize B16F10 in vitro (Fig. 3E and F). Although mice receiving IL2Cx25 had more antigen-specific T cells than did those animals that received IL2Cx122, both types of IL2Cx provided comparable therapeutic effects. These results indicate that IL2Cx122 provides enough T cells to control the tumor and that the additional T cells produced by IL2Cx25 do not provide further benefit. Similar therapeutic antitumor effects were observed in BALB/c mice using the RNEU-expressing A2L2 tumor (Supplementary Fig. S5A and S5B) with more antitumor effect in the prophylactic setting (Supplementary Fig. S5C and S5D). Thus, although the therapeutic benefit of BiVax and IL2Cx did not eliminate the tumors and was transient, the effects did extend overall survival or prevent tumor growth.

Figure 3.

Therapeutic effects of BiVax/IL2Cx. A, Diagram of tumor inoculation and vaccination protocol. B, Tumor size growth curves showing means with SD for each group. C, Percentages of Trp1-tetramer+ T cells in blood (each point = 1 mouse). D, PD-1 expression as mean fluorescence intensity (MFI) on Trp1 tetramer+ T cells on day 31. E, Spleens from surviving mice were harvested on day 42 and purified CD8+ T cells were incubated with B16F10 cells to estimate the number of IFNγ-producing cells by ELISpot. F, Representative ELISpot wells from E (n = 10 mice/group). Representative results are shown from at least 3 independent experiments.

Figure 3.

Therapeutic effects of BiVax/IL2Cx. A, Diagram of tumor inoculation and vaccination protocol. B, Tumor size growth curves showing means with SD for each group. C, Percentages of Trp1-tetramer+ T cells in blood (each point = 1 mouse). D, PD-1 expression as mean fluorescence intensity (MFI) on Trp1 tetramer+ T cells on day 31. E, Spleens from surviving mice were harvested on day 42 and purified CD8+ T cells were incubated with B16F10 cells to estimate the number of IFNγ-producing cells by ELISpot. F, Representative ELISpot wells from E (n = 10 mice/group). Representative results are shown from at least 3 independent experiments.

Close modal

BiVax/IL2Cx vaccine has better antitumor effect than TriVax

The antitumor effect of IL2Cx could be attributed to the higher number of antigen-specific T cells generated by adding IL2Cx or due to reduction in the level of PD-1 expression on the surface of antigen-specific cells. To distinguish between these two possibilities, we used TriVax (BiVax plus CD40 mAb) as a booster vaccine. TriVax generated large T-cell responses similar to those observed with BiVax/IL2Cx using TgTR1 ACT or endogenous responses (Fig. 4A and B) with similar PD-1 expression (Fig. 4C). Even though the two different boosters appeared to be equally immunogenic, the BiVax/IL2Cx boost was more efficient in controlling tumor growth compared with TriVax (Fig. 4D). Nonetheless, coadministration of IL2Cx or PD-L1 blockade with TriVax improved the antitumor effect of TriVax (Fig. 4D and E). Taken together, these data suggest that IL2Cx not only increases the number of antigen-specific T cells but also enhances the quality of the cells, making them more effective in the antitumor response. The reduction of PD1 on BiVax/IL2Cx–generated T cells, compared with those observed on T cells derived from BiVax alone on day 31 (Fig. 3D), could help explain an increased resistance to PD-1 inhibition. However, T-cell responses generated with TriVax had similar expression of PD-1 (Fig. 4C). Thus, the surface expression of PD-1 alone cannot explain these differences in the antitumor effects.

Figure 4.

Comparisons of BiVax/IL2Cx and TriVax for immunogenicity and antitumor efficacy. A, CD45.1 WT mice received 1 × 105 TgTR1 T cells (CD45.2) and were vaccinated as shown in the diagram. Total numbers of TgTR1 cells were evaluated in spleens on day 12. B, WT mice received Trp1-BiVax prime and 5 days later received either a BiVax/IL2Cx25 or a TriVax booster, and on day 12 the numbers of Trp1 tetramer+ T cells were enumerated in spleens. C, Mean fluorescence intensity (MFI) of PD-1 expression on TgTR1 cells in the experiment in A (n = 3 mice/group). D, WT mice were inoculated s.c. with 3 × 105 B16F10 cells and 7 days later mice received Trp1-BiVax vaccine prime followed by boosters with either TriVax, BiVax/IL2Cx25, or TriVax/IL2Cx25. Tumor size growth curves showing means with SD for each group (n = 5 mice/group). E, WT mice were inoculated s.c. with 3 × 105 B16F10 cells, and 7 days later mice received Trp1-BiVax prime followed by booster vaccines with either TriVax, TriVax/PD-L1 (200 μg/mouse), or BiVax/IL2Cx25. Tumor size growth curves showing means with SD for each group (n = 5 mice/group). Representative results are shown from at least 3 independent experiments.

Figure 4.

Comparisons of BiVax/IL2Cx and TriVax for immunogenicity and antitumor efficacy. A, CD45.1 WT mice received 1 × 105 TgTR1 T cells (CD45.2) and were vaccinated as shown in the diagram. Total numbers of TgTR1 cells were evaluated in spleens on day 12. B, WT mice received Trp1-BiVax prime and 5 days later received either a BiVax/IL2Cx25 or a TriVax booster, and on day 12 the numbers of Trp1 tetramer+ T cells were enumerated in spleens. C, Mean fluorescence intensity (MFI) of PD-1 expression on TgTR1 cells in the experiment in A (n = 3 mice/group). D, WT mice were inoculated s.c. with 3 × 105 B16F10 cells and 7 days later mice received Trp1-BiVax vaccine prime followed by boosters with either TriVax, BiVax/IL2Cx25, or TriVax/IL2Cx25. Tumor size growth curves showing means with SD for each group (n = 5 mice/group). E, WT mice were inoculated s.c. with 3 × 105 B16F10 cells, and 7 days later mice received Trp1-BiVax prime followed by booster vaccines with either TriVax, TriVax/PD-L1 (200 μg/mouse), or BiVax/IL2Cx25. Tumor size growth curves showing means with SD for each group (n = 5 mice/group). Representative results are shown from at least 3 independent experiments.

Close modal

We examined the response of CD8+ T cells from each vaccination protocol to antigen stimulation in the presence of PD-1 inhibitory signals. For these experiments, we utilized B16F10 melanoma cells which upregulate expression of PD-L1 in response to IFNγ (12, 29). CD8+ T cells obtained from mice that received TgTR1 ACT, primed with Trp1-BiVax and then boosted either with BiVax, BiVax/IL2Cx, or TriVax recognized B16F10 cells nearly to the same extent (80%–90%) and in an antigen-specific manner (Fig. 5A and B). Because IFNγ treatment of B16 cells reduces the density of Trp1455-463/H2Db cell surface complexes and decreases recognition by Trp1-specific CD8+ T cells (12), the IFNγ-treated B16 cells were pulsed with the Trp1455-463 peptide. The TgTR1 T cells generated by BiVax/IL2Cx recognized B16F10 and IFNγ-treated B16F10 cells (PD-L1 high) to the same extent (Fig. 5A and B). On the other hand, the TgTR1 T cells obtained from mice boosted with BiVax or TriVax were less able to recognize the IFNγ-treated B16 cells (∼80% reduction) as compared with the untreated B16 cells. Blocking PD-1/PD-L1 interactions with PD-L1 mAb restored the capacity of BiVax and TriVax generated T cells to recognize the IFNγ-treated B16F10 cells and had no enhancing effect with the T cells from the BiVax/IL2Cx group (Fig. 5A and B). These results agree with the observation that PD1 blockade improves the antitumor therapeutic effect of TriVax boost (Fig. 4E). To evaluate the capacity of T cells to proliferate in the presence of PD-1/PD-L1 signaling, we utilized aAPCs coated with Trp1455-463/H2Db monomers with or without PD-L1 protein (Fig. 5C). Presence of PD-L1 on the aAPCs reduced the proliferation of BiVax or TriVax generated T cells by 66% and 75%, respectively (Fig. 5D). On the other hand, the proliferation of T cells generated with BiVax/IL2Cx was not affected by the presence of PD-L1. Although IL2 signaling activates both PI3K/AKT and STAT5 pathways, the latter is more important for the proliferation and the effector function of T cells (27, 30–32). Administration of IL2Cx with BiVax increased pSTAT5 expression in the antigen-specific cells in vivo as compared with BiVax alone or TriVax (Fig. 5E). To evaluate the contribution of the STAT5 pathway in mediating resistance to PD-1 inhibition, TgTR1 cells transduced with constitutive active STAT5 (CA-STAT5) showed similar capacity to resist PD-L1/PD-1 signaling (Fig. 5F). The overall results suggest that IL2Cx not only increases the number of antigen-specific cells but also enhances their ability to resist the PD-1 inhibitory signals through the STAT5 pathway.

Figure 5.

CD8+ T cells generated with BiVax/IL2Cx show resistance to PD-L1 inhibition. A, CD45.1 WT mice were vaccinated as described in Fig. 4A, and on day 12 purified TgTR1 CD8+ T cells from spleens were incubated with either B16F10 (PD-L1 low) cells, IFNγ-treated B16F10 (PD-L1 high) cells (pulsed with 1 μg Trp1) with or without PD-L1 mAb (10 μg/mL), and the numbers of IFNγ producing cells were evaluated by ELISpot. B, Representative ELISpot wells from A. C, Diagram illustrating the artificial APCs (aAPC) used in D. D, CFSE-labeled previously activated TgTr1 CD8+ T cells were incubated for 72 hours at a 1:1 ratio with aAPCs containing either Trp1/H2Db monomers or Trp1/H2Db monomers plus PD-L1 protein, in the presence and absence of PD-L1 mAb (10 μg/mL). Percentages of proliferating (CFSE-diluted) TgTR1 cells were evaluated by flow cytometry. E, CD45.1 WT mice were vaccinated as described in Fig. 4A and 1 hour after the last dose of IL2Cx, pSTAT5 expression was evaluated in TgTR1 cells in the spleen. F, TgTR1 cells transduced or not with CA-STAT5 were adoptively transferred into WT mice, followed by a BiVax prime/boost vaccination (5 days apart). Mice receiving nontransduced T cells and treated with IL2Cx were included for comparison. On day 12, CD8+ T cells were purified, and the effects of PD-L1 inhibition were evaluated as described in A. Representative results are shown from at least 3 independent experiments.

Figure 5.

CD8+ T cells generated with BiVax/IL2Cx show resistance to PD-L1 inhibition. A, CD45.1 WT mice were vaccinated as described in Fig. 4A, and on day 12 purified TgTR1 CD8+ T cells from spleens were incubated with either B16F10 (PD-L1 low) cells, IFNγ-treated B16F10 (PD-L1 high) cells (pulsed with 1 μg Trp1) with or without PD-L1 mAb (10 μg/mL), and the numbers of IFNγ producing cells were evaluated by ELISpot. B, Representative ELISpot wells from A. C, Diagram illustrating the artificial APCs (aAPC) used in D. D, CFSE-labeled previously activated TgTr1 CD8+ T cells were incubated for 72 hours at a 1:1 ratio with aAPCs containing either Trp1/H2Db monomers or Trp1/H2Db monomers plus PD-L1 protein, in the presence and absence of PD-L1 mAb (10 μg/mL). Percentages of proliferating (CFSE-diluted) TgTR1 cells were evaluated by flow cytometry. E, CD45.1 WT mice were vaccinated as described in Fig. 4A and 1 hour after the last dose of IL2Cx, pSTAT5 expression was evaluated in TgTR1 cells in the spleen. F, TgTR1 cells transduced or not with CA-STAT5 were adoptively transferred into WT mice, followed by a BiVax prime/boost vaccination (5 days apart). Mice receiving nontransduced T cells and treated with IL2Cx were included for comparison. On day 12, CD8+ T cells were purified, and the effects of PD-L1 inhibition were evaluated as described in A. Representative results are shown from at least 3 independent experiments.

Close modal

IL2 mAb-released free IL2 is responsible for the adjuvant effect of IL2Cx

IL2Cx could enhance the IL2 function either by targeted delivery of IL2 to specific receptors (CD25 or CD122) or by providing sustained IL2 signaling through extending the half-life and availability of free IL2 after dissociating from the anti-IL2. For example, in the case of IL2Cx122 promoting the nonantigen-mediated expansion of memory CD8+ T cells, the antibody–cytokine complex bound directly to CD122 because a 100-fold excess of the mAb (which would neutralize free IL2) did not reduce the effect (18). We observed that administration of either CD25 mAb or CD122 mAb inhibited the expansion of Trp1-specific T cells induced by Trp1-BiVax/IL2Cx122 (Supplementary Fig. S6), suggesting that perhaps IL2 needs to engage both IL2 receptors. However, we could not eliminate the possibility that the mAbs were depleting activated T cells. Thus, to evaluate whether the effects of IL2Cx in the antigen-mediated CD8+ T-cell expansion were driven by the antibody–cytokine complex or free IL2, we examined whether an excess of the same mAb used to make the complex would decrease the strength of T-cell expansion. Indeed, increasing the amount of IL2 mAb used to form the complexes by 10-fold diminished the in vitro antigen-mediated expansion of TgTR1 cells by 50% (Fig. 6A). Furthermore, preparation of IL2Cx25 using 300 μg mAb (IL2Cx25/300μg), which is 30 times greater than the amount previously used (10 μg), did not increase the number of antigen-specific CD8+ T cells as compared with the vaccine without IL2Cx (Fig. 6B). On the other hand, IL2Cx25/10μg, which does not have an excess of the IL2 mAb, increased the numbers of tetramer-positive T cells more than 20-fold. In this experiment, the TgTR1 cells (CD45.2) represented ∼77% of the spleen's cell population in mice vaccinated with IL2Cx25/10μg and only 13%–18% in the mice vaccinated with IL2Cx25/300μg or unvaccinated mice (Fig. 6C). These results indicate that the enhancing effects of IL2Cx in the antigen-induced expansion of T cells requires the free form of IL2, which must dissociate from the mAb to be able to engage both CD25 and CD122.

Figure 6.

Sustained IL2 stimulation is responsible for the adjuvant effect of IL2Cx. A, TgTR1 cells were incubated with Trp1 peptide (1 μg/mL) and IL2Cx122 or IL2Cx25 (100 ng IL2/mL) in the presence of increasing concentrations of homologue IL2 mAb (JES6-5H4 or JES6-1A12, respectively). Seven days later T-cell expansion was evaluated. B, CD45.1 mice were adoptively transferred with 1 × 105 TgTR1 cells followed by BiVax prime. Five days later the mice were boosted with BiVax, BiVax/IL2Cx25(10 μg) (2 μg IL2 + 10 μg IL2 mAb), or BiVax/IL2Cx25(300 μg) (2 μg IL2 + 300 μg IL2 mAb). On day 12, the total numbers of TgTR1 cells were evaluated in spleens. C, Representative flow dot plots from the experiment in B, showing the percentage of TgTR1 cells (MHCIICD45.2+) in spleen gating in total live cells. (n = 3 mice/group). Representative results are shown from at least 3 independent experiments.

Figure 6.

Sustained IL2 stimulation is responsible for the adjuvant effect of IL2Cx. A, TgTR1 cells were incubated with Trp1 peptide (1 μg/mL) and IL2Cx122 or IL2Cx25 (100 ng IL2/mL) in the presence of increasing concentrations of homologue IL2 mAb (JES6-5H4 or JES6-1A12, respectively). Seven days later T-cell expansion was evaluated. B, CD45.1 mice were adoptively transferred with 1 × 105 TgTR1 cells followed by BiVax prime. Five days later the mice were boosted with BiVax, BiVax/IL2Cx25(10 μg) (2 μg IL2 + 10 μg IL2 mAb), or BiVax/IL2Cx25(300 μg) (2 μg IL2 + 300 μg IL2 mAb). On day 12, the total numbers of TgTR1 cells were evaluated in spleens. C, Representative flow dot plots from the experiment in B, showing the percentage of TgTR1 cells (MHCIICD45.2+) in spleen gating in total live cells. (n = 3 mice/group). Representative results are shown from at least 3 independent experiments.

Close modal

PEG-IL2 enhanced expansion of T cells and improved antitumor effects of vaccine

Our results suggest the adjunct effects of IL2Cx during the BiVax boost were mediated by improving the half-life of IL2 and not by directing IL2 to specific receptors. If so, other means of enhancing the half-life of IL2 should have similar adjunct effects. Pegylation of IL2 allows slow release of IL2 and maintains persistent IL2 signaling in vivo (21–23, 33). Pegylated IL2 (PEG-IL2) was as effective as IL2Cx in the expansion of endogenous Trp1455-463 specific T cells induced with BiVax (Fig. 7A). Moreover, BiVax/PEG-IL2 showed antitumor effects (Fig. 7B) similar to those observed with BiVax/IL2Cx (Fig. 3B). The Trp1455-463 specific cells generated by BiVax/PEG-IL2 resisted PD-1 inhibitory signaling in vitro (Fig. 7C) in a similar manner as observed with IL2Cx (Fig. 5A–B). These data show that pegylation of IL2, which allows persistent IL2 signaling, enhanced the antigen-mediated expansion of T cells as did IL2Cx. Twice-daily administration of nonpegylated high-dose human IL2 (HD-IL2), as used in the clinic, had an effect on the immune response to BiVax similar to PEG-IL2, with an increase in the expression pSTAT5 (Supplementary Fig. S7A and S7B). Tumor cells isolated from mice treated with BiVax/PEG-IL2 at the termination of the experiment (day 33) retained surface MHC-I expression and expressed similar amounts of PD-L1 (Supplementary Fig. S8A) and were able to stimulate TgTR1 cells in vitro (Supplementary Fig. S8B).

Figure 7.

PEG-IL2 is effective in expanding BiVax-generated antigen-specific T cells. A, WT mice (3/group) were vaccinated with BiVax prime. Five days later, the mice received boosters with BiVax, BiVax/IL2, BiVax/IL2Cx122, or BiVax/PEG-IL2. Seven days later, the numbers of Trp1 tetramer+ T cells were evaluated in spleens. B, WT mice were inoculated s.c. with B16F10 cells and, 7 days later, received a BiVax prime, followed 5 days later by booster vaccines with BiVax, BiVax/IL2, or BiVax/PEG-IL2. Tumor size growth curves showing means with SD for each group (n = 10 mice/group). C, Purified CD8+ T cells from WT mice receiving BiVax prime–boost (5 days apart) and treated or not with PEG-IL2 were incubated with either B16F10 (PD-L1 low) cells, IFNγ-treated (PD-L1 high) B16F10 cells (pulsed with 1 μg Trp1) with or without PD-L1 mAb (10 μg/mL) and the numbers of IFNγ-producing cells were evaluated by ELISpot. Representative results are shown from at least 3 independent experiments.

Figure 7.

PEG-IL2 is effective in expanding BiVax-generated antigen-specific T cells. A, WT mice (3/group) were vaccinated with BiVax prime. Five days later, the mice received boosters with BiVax, BiVax/IL2, BiVax/IL2Cx122, or BiVax/PEG-IL2. Seven days later, the numbers of Trp1 tetramer+ T cells were evaluated in spleens. B, WT mice were inoculated s.c. with B16F10 cells and, 7 days later, received a BiVax prime, followed 5 days later by booster vaccines with BiVax, BiVax/IL2, or BiVax/PEG-IL2. Tumor size growth curves showing means with SD for each group (n = 10 mice/group). C, Purified CD8+ T cells from WT mice receiving BiVax prime–boost (5 days apart) and treated or not with PEG-IL2 were incubated with either B16F10 (PD-L1 low) cells, IFNγ-treated (PD-L1 high) B16F10 cells (pulsed with 1 μg Trp1) with or without PD-L1 mAb (10 μg/mL) and the numbers of IFNγ-producing cells were evaluated by ELISpot. Representative results are shown from at least 3 independent experiments.

Close modal

In summary, the results presented herein demonstrate that when IL2 persistence was enhanced through use of IL2Cx or pegylation, more antigen-specific T cells could be induced by peptide vaccination. With sustained IL2, the T cells were resistant to inhibitory PD-1 signaling and exerting antitumor effects.

IL2 has been approved for treatment of some cancers, including metastatic melanoma and renal cell carcinoma. In these cases, IL2 has shown objective response rates of 10% to 20%. However, because IL2 is rapidly cleared, high doses are required, causing toxicity. Although IL2 is essential for the expansion of T cells, its use for potentiating cancer vaccines has not been thoroughly investigated. In some instances, the combination of a gp100 peptide vaccine with high doses of IL2 showed little benefit compared with either the vaccine or IL2 by themselves (34, 35). Many cell subsets (effector, memory, regulatory T cells, and NK cells) constitutively express various IL2R levels (CD122 and CD25) and can respond to and expand after IL2 administration. Such cells likely compete with vaccine-induced antigen-specific cells for IL2. We observed that IL2 administration (as IL2Cx) together with the priming vaccine reduced the antigen-specific T cell response as compared with vaccine alone. Verdeil and colleagues reported that IL2Cx enhanced the responses of adoptively transferred CD8+ T cells after a peptide/poly-IC vaccine prime (36). However, this group used a large number of T cells for the ACT (3 × 106) and only observed ∼2-fold expansion. Thus, it appears that to obtain a beneficial effect from IL2, its administration should be delayed so that vaccine-induced T cells can first express IL2Rs and expand, improving their ability to compete with irrelevant cells that might also consume IL2.

Increasing IL2 half-life through use of IL2Cx or PEG-IL2 improved expansion of vaccine-generated T cells. Administration of high doses of IL2 (HD-IL2) during the vaccine boost, in its natural form at 600,000 IU (60 μg)/injection, twice daily for 3 to 5 days (in a similar manner as used in the clinic) also enhanced expansion of antigen-specific T cells and increased pSTAT5 expression. Thus, sustained IL2R stimulation maintained cell division or prevented cell death for vaccine-stimulated T cells. Because of the toxic effects of HD-IL2 therapy, using lower doses of IL2 is more appealing. Both IL2Cx and PEG-IL2 increase IL2 half-life, enabling the use of lower doses. Repeated administration of IL2Cx mediates expansion of various lymphoid cells depending on the IL2 mAb used. IL2Cx122 specifically expands cells expressing CD122, such as CD8+ memory T cells and NK cells, and IL2Cx25 preferentially expands CD4/FoxP3 Tregs through targeting CD25 (18–20). Here, we showed that both forms of IL2Cx had similar ability to mediate antigen-dependent expansion of CD8+ T cells. Although the ability of IL2Cx alone to expand memory T cells, NK cells, or Tregs is mediated by the IL2/mAb complex binding to the IL2 receptors CD122 or CD25, our data suggest that IL2 dissociated from the mAb is responsible for the expansion of the vaccine-stimulated T-cell response. First, we observed that excess of mAb in the IL2Cx, which would decrease the amount of free IL2, prevented both in vitro and in vivo antigen-induced T-cell proliferation. On the other hand, Phelan and colleagues reported that the use of either 50 or 5,000 μg of IL2 mAb in IL2Cx122 had similar effects on the proliferation of CD8+ memory T cells, suggesting that in their case the IL2Cx could bind directly to CD122 (37). Second, the effect of randomly pegylated-IL2 (PEG-IL2) or HD-IL2, which in theory should not target IL2 to a specific IL2R chain, was similar to the use of IL2Cx. These data indicate that IL2 enhances the effect of BiVax by sustaining IL2 signaling, which in the case of IL2Cx and PEG-IL2 is accomplished by decreasing the rate of IL2 clearance from the blood.

Administration of IL2Cx or PEG-IL2 during the vaccine enhanced the antitumor effect of the peptide vaccines, such that in many instances the mice rejected their tumors. Such antitumor effects could be attributed to increased numbers of antigen-specific T cells obtained with IL2Cx. However, TriVax, which induced similar numbers of antigen-specific cells as did BiVax + IL2Cx, did not show antitumor effects. Addition of IL2Cx or PD1 blockade to TriVax enhanced its antitumor effect to a similar level as BiVax + IL2Cx. These findings suggested that IL2Cx enhances the quality of T cells, making them more effective at destroying tumor cells. Expression of PD-L1 by B16 melanoma cells (29) prevents tumor destruction by CTLs; IL2Cx could increase the resistance of T cells to PD-L1 inhibition, perhaps by diminishing the expression of PD-1. Antigen-specific T cells generated with BiVax + IL2Cx expressed less PD-1 than did T cells generated by BiVax alone, but similar amounts as T cells generated with TriVax. Thus, PD-1 cell surface expression may not be solely responsible for enhancing the quality of the T cells generated with IL2Cx (or PEG-IL2). We considered the possibility that sustained IL2 stimulation could decrease PD-1 signaling regardless of PD-1 expression. Indeed, IL2Cx enhanced the ability of the T cells to resist PD-L1 inhibitory signals provided by either tumor cells or aAPCs. The STAT5 pathway regulates proliferation and the effector function of T cells (27, 30–32). Transduction of a constitutively active form of STAT5 into TCR transgenic T cells followed by ACT and BiVax resulted in PD-L1 resistance similar to that observed with IL2Cx. Further studies are warranted to examine the mechanism by which STAT5 signaling confers resistance to PD-L1 inhibition.

BiVax/IL2Cx delayed tumor growth, but in many instances the tumors started to grow by day 25. Numbers of tumor-reactive T cells remained high in these mice. Tumor progression was not due to tumor escape variants. These data suggest that resistance to PD-L1 inhibition conferred by IL2Cx (and PEG-IL2) may not be permanent and that a more continuous IL2Cx (or PEG-IL2) therapy after vaccination may be required to achieve persistent control of tumor growth or tumor rejection. In summary, the present studies provide a strategy for the use of IL2 (in formulations that extend its half-life) for enhancing the therapeutic effects of T cells generated by peptide-based vaccines. Implementation of this strategy into the clinic might be possible in the near future.

No potential conflicts of interest were disclosed.

Conception and design: H. Sultan, T. Kumai, V.I. Fesenkova, A.E. Fan, E. Celis

Development of methodology: H. Sultan, T. Kumai, V.I. Fesenkova, A.E. Fan, H.-II. Cho, E. Celis

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): H. Sultan, T. Kumai, V.I. Fesenkova, A.E. Fan, J. Wu

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): H. Sultan, T. Kumai, V.I. Fesenkova, A.E. Fan, Y. Harabuchi, E. Celis

Writing, review, and/or revision of the manuscript: H. Sultan, T. Kumai, V.I. Fesenkova, A.E. Fan, J. Wu, Y. Harabuchi, E. Celis

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): H. Sultan, H.-II. Cho

Study supervision: H. Kobayashi, E. Celis

This work was supported by a grant from the NCI (R01CA157303) and by start-up funds from Augusta University, Georgia Cancer Center, and the Georgia Research Alliance.

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.
Reck
M
,
Rodriguez-Abreu
D
,
Robinson
AG
,
Hui
R
,
Csoszi
T
,
Fulop
A
, et al
Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer
.
N Engl J Med
2016
;
375
:
1823
33
.
2.
Herbst
RS
,
Baas
P
,
Kim
DW
,
Felip
E
,
Perez-Gracia
JL
,
Han
JY
, et al
Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial
.
Lancet
2016
;
387
:
1540
50
.
3.
Rosenberg
SA
,
Yang
JC
,
Restifo
NP
. 
Cancer immunotherapy: moving beyond current vaccines
.
Nat Med
2004
;
10
:
909
15
.
4.
Klebanoff
CA
,
Rosenberg
SA
,
Restifo
NP
. 
Prospects for gene-engineered T cell immunotherapy for solid cancers
.
Nat Med
2016
;
22
:
26
36
.
5.
Hinrichs
CS
,
Rosenberg
SA
. 
Exploiting the curative potential of adoptive T-cell therapy for cancer
.
Immunol Rev
2014
;
257
:
56
71
.
6.
Melero
I
,
Gaudernack
G
,
Gerritsen
W
,
Huber
C
,
Parmiani
G
,
Scholl
S
, et al
Therapeutic vaccines for cancer: an overview of clinical trials
.
Nat Rev Clin Oncol
2014
;
11
:
509
24
.
7.
Zitvogel
L
,
Kroemer
G
. 
Targeting PD-1/PD-L1 interactions for cancer immunotherapy
.
Oncoimmunology
2012
;
1
:
1223
5
.
8.
Ott
PA
,
Hu
Z
,
Keskin
DB
,
Shukla
SA
,
Sun
J
,
Bozym
DJ
, et al
An immunogenic personal neoantigen vaccine for patients with melanoma
.
Nature
2017
.
doi 10.1038/nature22991
.
9.
Garon
EB
,
Rizvi
NA
,
Hui
R
,
Leighl
N
,
Balmanoukian
AS
,
Eder
JP
, et al
Pembrolizumab for the treatment of non-small-cell lung cancer
.
N Engl J Med
2015
;
372
:
2018
28
.
10.
Cho
HI
,
Barrios
K
,
Lee
YR
,
Linowski
AK
,
Celis
E
. 
BiVax: a peptide/poly-IC subunit vaccine that mimics an acute infection elicits vast and effective anti-tumor CD8 T-cell responses
.
Cancer Immunol Immunother
2013
;
62
:
787
99
.
11.
Cho
HI
,
Celis
E
. 
Optimized peptide vaccines eliciting extensive CD8 T-cell responses with therapeutic antitumor effects
.
Cancer Res
2009
;
69
:
9012
9
.
12.
Cho
HI
,
Lee
YR
,
Celis
E
. 
Interferon gamma limits the effectiveness of melanoma peptide vaccines
.
Blood
2011
;
117
:
135
44
.
13.
Barrios
K
,
Celis
E
. 
TriVax-HPV: an improved peptide-based therapeutic vaccination strategy against human papillomavirus-induced cancers
.
Cancer Immunol Immunother
2012
;
61
:
1307
17
.
14.
Wang
Z
,
Celis
E
. 
STING activator c-di-GMP enhances the anti-tumor effects of peptide vaccines in melanoma-bearing mice
.
Cancer Immunol Immunother
2015
;
64
:
1057
66
.
15.
Sultan
H
,
Fesenkova
VI
,
Addis
D
,
Fan
AE
,
Kumai
T
,
Wu
J
, et al
Designing therapeutic cancer vaccines by mimicking viral infections
.
Cancer Immunol Immunother
2017
;
66
:
203
13
.
16.
Sultan
H
,
Trillo-Tinoco
J
,
Rodriguez
P
,
Celis
E
. 
Effective antitumor peptide vaccines can induce severe autoimmune pathology
.
Oncotarget
2017
;
8
:
70317
31
.
17.
Kumai
T
,
Lee
S
,
Cho
HI
,
Sultan
H
,
Kobayashi
H
,
Harabuchi
Y
, et al
Optimization of peptide vaccines to induce robust antitumor CD4 T-cell responses
.
Cancer Immunol Res
2017
;
5
:
72
83
.
18.
Boyman
O
,
Kovar
M
,
Rubinstein
MP
,
Surh
CD
,
Sprent
J
. 
Selective stimulation of T cell subsets with antibody-cytokine immune complexes
.
Science
2006
;
311
:
1924
7
.
19.
Spangler
JB
,
Tomala
J
,
Luca
VC
,
Jude
KM
,
Dong
S
,
Ring
AM
, et al
Antibodies to interleukin-2 elicit selective T cell subset potentiation through distinct conformational mechanisms
.
Immunity
2015
;
42
:
815
25
.
20.
Letourneau
S
,
van Leeuwen
EM
,
Krieg
C
,
Martin
C
,
Pantaleo
G
,
Sprent
J
, et al
IL-2/anti-IL-2 antibody complexes show strong biological activity by avoiding interaction with IL-2 receptor alpha subunit CD25
.
Proc Natl Acad Sci USA
2010
;
107
:
2171
6
.
21.
Charych
DH
,
Hoch
U
,
Langowski
JL
,
Lee
SR
,
Addepalli
MK
,
Kirk
PB
, et al
NKTR-214, an engineered cytokine with biased IL2 receptor binding, increased tumor exposure, and marked efficacy in mouse tumor models
.
Clin Cancer Res
2016
;
22
:
680
90
.
22.
Turecek
PL
,
Bossard
MJ
,
Schoetens
F
,
Ivens
IA
. 
PEGylation of biopharmaceuticals: a review of chemistry and nonclinical safety information of approved drugs
.
J Pharm Sci
2016
;
105
:
460
75
.
23.
Yang
JC
,
Topalian
SL
,
Schwartzentruber
DJ
,
Parkinson
DR
,
Marincola
FM
,
Weber
JS
, et al
The use of polyethylene glycol-modified interleukin-2 (PEG-IL-2) in the treatment of patients with metastatic renal cell carcinoma and melanoma. A phase I study and a randomized prospective study comparing IL-2 alone versus IL-2 combined with PEG-IL-2
.
Cancer
1995
;
76
:
687
94
.
24.
Dougan
SK
,
Dougan
M
,
Kim
J
,
Turner
JA
,
Ogata
S
,
Cho
HI
, et al
Transnuclear TRP1-specific CD8 T cells with high or low affinity TCRs show equivalent antitumor activity
.
Cancer Immunol Res
2013
;
1
:
99
111
.
25.
Boggio
K
,
Nicoletti
G
,
Di Carlo
E
,
Cavallo
F
,
Landuzzi
L
,
Melani
C
, et al
Interleukin 12-mediated prevention of spontaneous mammary adenocarcinomas in two lines of Her-2/neu transgenic mice
.
J Exp Med
1998
;
188
:
589
96
.
26.
Lachman
LB
,
Rao
XM
,
Kremer
RH
,
Ozpolat
B
,
Kiriakova
G
,
Price
JE
. 
DNA vaccination against neu reduces breast cancer incidence and metastasis in mice
.
Cancer Gene Ther
2001
;
8
:
259
68
.
27.
Hand
TW
,
Cui
W
,
Jung
YW
,
Sefik
E
,
Joshi
NS
,
Chandele
A
, et al
Differential effects of STAT5 and PI3K/AKT signaling on effector and memory CD8 T-cell survival
.
Proc Natl Acad Sci USA
2010
;
107
:
16601
6
.
28.
Oelke
M
,
Maus
MV
,
Didiano
D
,
June
CH
,
Mackensen
A
,
Schneck
JP
. 
Ex vivo induction and expansion of antigen-specific cytotoxic T cells by HLA-Ig-coated artificial antigen-presenting cells
.
Nat Med
2003
;
9
:
619
24
.
29.
Nagato
T
,
Lee
YR
,
Harabuchi
Y
,
Celis
E
. 
Combinatorial immunotherapy of polyinosinic-polycytidylic acid and blockade of programmed death-ligand 1 induce effective CD8 T-cell responses against established tumors
.
Clin Cancer Res
2014
;
20
:
1223
34
.
30.
Gilmour
KC
,
Pine
R
,
Reich
NC
. 
Interleukin 2 activates STAT5 transcription factor (mammary gland factor) and specific gene expression in T lymphocytes
.
Proc Natl Acad Sci USA
1995
;
92
:
10772
6
.
31.
Malek
TR
. 
The biology of interleukin-2
.
Annu Rev Immunol
2008
;
26
:
453
79
.
32.
Moriggl
R
,
Topham
DJ
,
Teglund
S
,
Sexl
V
,
McKay
C
,
Wang
D
, et al
Stat5 is required for IL-2-induced cell cycle progression of peripheral T cells
.
Immunity
1999
;
10
:
249
59
.
33.
Meyers
FJ
,
Paradise
C
,
Scudder
SA
,
Goodman
G
,
Konrad
M
. 
A phase I study including pharmacokinetics of polyethylene glycol conjugated interleukin-2
.
Clin Pharmacol Ther
1991
;
49
:
307
13
.
34.
Schwartzentruber
DJ
,
Lawson
DH
,
Richards
JM
,
Conry
RM
,
Miller
DM
,
Treisman
J
, et al
gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma
.
N Engl J Med
2011
;
364
:
2119
27
.
35.
Smith
FO
,
Downey
SG
,
Klapper
JA
,
Yang
JC
,
Sherry
RM
,
Royal
RE
, et al
Treatment of metastatic melanoma using interleukin-2 alone or in conjunction with vaccines
.
Clin Cancer Res
2008
;
14
:
5610
8
.
36.
Verdeil
G
,
Marquardt
K
,
Surh
CD
,
Sherman
LA
. 
Adjuvants targeting innate and adaptive immunity synergize to enhance tumor immunotherapy
.
Proc Natl Acad Sci USA
2008
;
105
:
16683
8
.
37.
Phelan
JD
,
Orekov
T
,
Finkelman
FD
. 
Cutting edge: mechanism of enhancement of in vivo cytokine effects by anti-cytokine monoclonal antibodies
.
J Immunol
2008
;
180
:
44
8
.