High-dose IL2 immunotherapy can induce long-lasting cancer regression but is toxic and insufficiently efficacious. Improvements are obtained with IL2/anti-IL2 complexes (IL2Cx), which redirect IL2 action to CD8+ T and natural killer (NK) cells. Here, we evaluated the efficacy of combining IL2Cx with blockade of inhibitory immune pathways. In an autochthonous lung adenocarcinoma model, we show that the IL2Cx/anti–PD-1 combination increases CD8+ T-cell infiltration of the lung and controls tumor growth. In the B16-OVA model, which is resistant to checkpoint inhibition, combination of IL2Cx with PD-1 or CTLA-4 pathway blockade reverses that resistance. Both combinations work by reinvigorating exhausted intratumoral CD8+ T cells and by increasing the breadth of tumor-specific T-cell responses. However, only the IL2Cx/anti–CTLA-4 combination is able to rescue NK cell antitumor function by modulating intratumoral regulatory T cells. Overall, association of IL2Cx with PD-1 or CTLA-4 pathway blockade acts by different cellular mechanisms, paving the way for the rational design of combinatorial antitumor therapies.

Administration of high-dose IL2 immunotherapy, which can induce durable cancer regression, is approved by the FDA for treatment of metastatic melanoma and renal carcinoma (1). IL2 therapy can, however, be toxic and has low efficacy (5%–20% of responders) due to its effect on regulatory T cells (Tregs), which express the high-affinity IL2 receptor (composed of subunits IL2-Rα, IL2Rβ, and IL2Rγc) and can block the antitumor immune response (2). Antitumoral activity requires activation of CD8+ T and natural killer (NK) cells, which respond to IL2 through the intermediate affinity IL2 receptor (composed of IL2Rβ and IL2Rγc subunits; ref. 3).

One strategy to avoid unwanted activation of Tregs is to use IL2/anti-IL2 complexes (IL2Cx). IL2Cx can redirect IL2 to Treg or to IL2Rβγc-expressing cells that can mount an antitumoral response (4). IL2Cx has a longer half-life than IL2, resulting in better pharmacodynamics (5). High-dose IL2 therapy can be toxic due to vascular leak syndrome. IL2-associated pulmonary edema seems to be caused by interaction of IL2 with its receptors (IL2-Rα, CD25) on lung endothelial cells (6). This side effect can be abrogated by use of IL2Rβγc-directed IL2Cxs, which should not activate CD25 on the endothelial cells (5). In murine models of cancer, the IL2Rβγc-directed IL2Cx can boost the antitumor response by stimulating NK and CD8+ T cells, encouraging the clinical development of IL2Cx (7). Nevertheless, in experimental models, administration of IL2Cx as monotherapy delays tumor growth only modestly (6–8).

Given the success of anticheckpoint therapies in cancer, combination of IL2Cx with anticheckpoint mAbs that target CTLA-4 or PD-1/PD-L1 inhibitory pathways could improve results. Indeed, single-agent administration of anti–CTLA-4 or anti–PD-L1 shows clinical efficacy in around 20% of patients with tumors (9). Combination of anti–CTLA-4 and anti–PD-1 increases both response rate and toxicities (10), although the majority of cancer patients still do not respond. Acquired resistance to treatment has also been observed (11, 12). Efficient combination therapies are needed to increase the number of responding patients.

In this study, we evaluated whether combining IL2Cx administration with the blockade of inhibitory immune pathways represents an effective antitumor therapy. We tested two experimental models: B16-OVA, which is resistant to anticheckpoint inhibition (13), and the KrasLSL-G12D/+p53flox/flox genetically engineered mouse lung adenocarcinoma model (14). In both models, we observed that combination of IL2Cx with anticheckpoint mAbs was more effective. We found that IL2Cx works in a cell-intrinsic manner, boosting tumoral NK, reinvigorating exhausted CD8+ T cells, and increasing the breadth of tumor-specific T-cell responses. We also found that combination of IL2Cx with blockade of the PD-1 or CTLA-4 pathways increases tumor infiltration by CD8+ T cells. Anti–CTLA-4 contributes to efficacy by a CD8+ T and NK cell–extrinsic mechanism, releasing effector CD8+ T and NK cells from Treg suppression. Consequently, the combined treatment of IL2Cx with CTLA-4 blockade relies on the action of NK cells. These two combinations, which act by different cellular mechanisms, suggest avenues for the design of combinatory immunotherapies adapted to the individual tumor microenvironment of each patient. Our preclinical results show the validity of combining IL2Cx with anticheckpoint mAbs to fight cancer.

Mice

C57BL/6 female mice (5–6 weeks old) were purchased from The Charles River Laboratories. KrasLSL-G12D/+p53flox/flox mice (referred to as KP mice) were kindly given by T. Jacks (ref. 14; NIH) and backcrossed on the C57BL/6 background for more than 10 generations at CNRS Central Animal Facility TAAM (Orléans, France). Experiments with KP mice were performed with females of 8 to 12 weeks old. Experimental animal procedures were approved by the ethics committee of the Institut Curie CEEA-IC #118 (DAP n° 05302.03), in compliance with international guidelines.

Cell lines

B16F10 melanoma cell line expressing OVA (B16-OVA) was kindly given by K. Rock in 1999 and was cultured in RPMI (Gibco) supplemented with 10% heat-inactivated fetal bovine serum (Biosera), 2 mmol/L L-glutamine (Life Technologies), β-mercaptoethanol, and 1% penicillin/streptomycin (Life Technologies). For all experiments, cells were thawed from the same stock generated in 2016. Cells were cultured during maximum 2 weeks before injection into mice. Cells were not authenticated in the past year. Cells were checked for the absence of Mycoplasma by PCR reaction (GAT-Biotech).

HEK293-LTV and 3TZ cells were obtained from ATCC in 2013 and were used upon a second passage. Cells were cultured in Dubeco's modified Eagle medium (DMEM; Gibco) supplemented with 10% heat-inactivated fetal bovine serum (Biosera). Cells were not authenticated in the past year. Cells were checked for the absence of mycoplasma by PCR reaction (GAT-Biotech).

Lentiviral particle production and titration

A second generation of lentivirus (LV) was produced as previously described (15). Briefly, LV particles were produced by transfection of the HEK-293LTV cell line (Cell Biolabs, Inc.) with pAX2 (packaging plasmid; Addgene), pCMV-VSV-G (envelope plasmid; Addgene), and luciferase and recombinase Cre-expressing plasmid. The latter was modified to express SIINFEKL and DBY epitopes fused to the C-terminal end of luciferase protein. Moreover, this luciferase–SIINFEKL–DBY sequence was modified to restrict SIINFEKL and DBY expression to nonhematopoietic cell lineage as previously described (15). Functional particles were quantified using a Cre activity readout system based on the reporter cell line 3TZ expressing β-galactosidase upon Cre-mediated recombination.

Tumor experiments

For the transplantable tumor model, C57BL/6 females (5–6 weeks old) were injected subcutaneously in the flank with 0.5 × 106 B16-OVA cells in 200 μL of PBS. Tumor growth was measured using a metric caliper 2 to 3 times a week.

Lung adenocarcinoma was induced in KP mice (8–12 weeks old) by intratracheal injection of 2 × 104 lentiviral particles expressing the fusion protein luciferase–SIINFEKL–DBY and Cre-recombinase as previously described (15). In vivo tumor growth was measured by bioluminescence. Briefly, lentiviral-injected mice were shaved and anesthetized with Isoflurane (Sigma-Aldrich). D-luciferin substrate (150 mg/kg, Promega) dissolved in PBS was injected intraperitoneally (i.p.) 15 minutes before imaging in IVIS Spectrum (PerkinElmer). Photon fluxes were transformed into pseudocolor images using the Living Image software (PerkinElmer).

In vivo treatments

Recombinant human IL2 (rhIL2, Proleukine, Novartis), anti–CTLA4 (clone 9H10; Bio X Cell), anti–PD-1 (clone RMP1-14; Bio X Cell), anti–PDL-1 (clone 10F9G2; Bio X Cell), anti-human IL2 (clone MAB602; Bio-Techne), and anti-human IL2 (clone 5344.111; BD Biosciences) were purchased. IL2Cx were prepared mixing 15,000 UI of rhIL2 with 4.5 μg of Ab (molar ratio 2:1) and incubated for 30 minutes at 37°C. Treatment of B16-OVA engrafted mice began when tumors became measurable (at approximately day 10). Treatment of KrasLSL-G12D/+p53flox/flox was started when luciferase signal achieved 1 × 105 photons/second (between 12 and 14 weeks after viral inoculation). Where indicated, tumor-bearing mice received i.p. injections of IL2Cx for 5 consecutive days. Anticheckpoint mAbs were injected i.p. at 200 μg/dose 3 times a week, for 7 total doses with the B16-OVA tumor or 8 weekly cycles for KrasLSL-G12D/+p53flox/flox mice.

For depletion experiments, CD8+ cell depletion was performed using 200 μg anti-CD8 (clone 53.6.72; Bio X Cell) and NK cells depletion using 200 μg anti-NK (clone PK136; Bio X Cell). Depleting mAbs were administered i.p. 1 or 2 days prior to therapeutic treatment and 1 or 2 days after. Depletion was maintained by twice-a-week injection of depleting Abs until the end of the experiment. CD8+ T-cell and NK cell depletion was monitored in blood after 4 and 6 doses of depleting mAbs.

Preparation of cell suspensions

Subcutaneous tumors were collected in CO2 independent medium (Gibco). Then, tumors were cut into small pieces and placed into 2.5 mL of CO2 independent medium containing 0.1 mg/mL DNase I (Roche) and 0.1 mg/mL Liberase TL (Roche) in C tubes (Miltenyi Biotec). After mechanical dissociation with gentleMACS Dissociator, samples were incubated with shaking at 37°C for 30 minutes and processed again with gentleMACS. The cell suspension was then filtered with a 100-μm cell strainer for direct tumor cell analysis and further separated on a Percoll gradient (GE Healthcare Life Sciences) from 40% to 75% interface to recover mononuclear cells for immune cell infiltrate analysis.

Inguinal tumor-draining lymph nodes (dLN) were collected in CO2 independent medium. Single-cell suspensions were obtained by mechanical disruption over a 40-μm cell strainer.

Lungs were perfused with 20 mL of cold PBS to remove circulating blood cells prior to preparation of single-cell suspension. Mononuclear cells from lungs were obtained as described above for subcutaneous tumors.

Detection of Ag-specific T cells by ELISPOT

IFNγ-producing antigen-specific CD4+ or CD8+ T cells were measured by ELISPOT. Briefly, microplates (MAIPS4510; Millipore) were coated with anti-murine IFNγ (Diaclone). PBMCs (0.3 × 106) were cultured overnight in the presence of either control medium or class I–restricted OVA-I peptide (257–264, SIINFEKL, 10 mmol/L) or the class II–restricted DBY peptide (NAGFNSNRANSSRSS, 40 mmol/L), or B16-OVA melanoma neoepitopes Pool 1 (10 mmol/L) or melanoma neoepitopes Pool 2 (10 mmol/L). Cells and peptides were resuspended in complete medium RPMI (Gibco) supplemented with 10% heat-inactivated fetal bovine serum (Biosera), 2 mmol/L L-glutamine (Life Technologies), 1% penicillin/streptomycin (Life Technologies), and β-mercaptoethanol. Melanoma neoepitope peptides were kindly given by G. Lynn, NIH. Peptide sequences are: M23 (TNGSFIRLL), M27 (GVELCPGNKYEM), M30 (PSKPSFQEFVDWENVSPELNSTDQPFL), M47 (AAIVGKQVL), M49 (IAMQNTTQL); NB02 (AALTFRRL) and NB08 (KILRMSPL).

Detection of IFNγ-producing cells was performed with biotinylated anti-IFNγ (matched pairs; Diaclone) followed by streptavidin–alkaline phosphatase (Mabtech) and revealed using the appropriate substrate (Bio-Rad). Spots were counted using an ELISPOT Reader System ELR02 (AID). Results were expressed as number of cytokine-producing cells (SFC) per 1 × 106 of total cells.

Flow-cytometric analysis

For flow-cytometry analysis, mAbs specific for mouse CD152 (UC10-4F10-11), CD274 (MIH5), CD4 (RM4-5), CD8 (53-6.7), GITR (DTA-1), Ki67 (B56), KLRG1(2F1), and TCRβ (H57-597) were purchased from BD Biosciences. mAbs against mouse CD19 (6D5), CD25 (3C7), CD27 (LG.3A10), CD4 (GK1.5), CD44 (IM7), CD45.2 (104), Foxp3 (MF-14), GzmB (GB11), ICOS (C398.4A), IFNγ (XMG1.2), NK 1.1 (PK136), PD-1 (29F.1A12), Tbet (4B10), and TCRβ (H57-597) were purchased from BioLegend. mAb against mouse EOMES (Dan11mag) was purchased from eBioscience/Thermo Fisher, unconjugated gp100 (EP4863) was purchased from Abcam, and anti-rabbit IgG (H + L)-A488 secondary Ab was purchased from Thermo Fisher.

Surface staining was performed at 4°C with antibodies resuspended in PBS with 2% FBS and 2 mmol/L EDTA. For intracellular staining of cytokines, cells were restimulated with phorbol 12-myristate 13-acetate (PMA, 20 ng/mL) and 1 μg/mL of ionomycin (Sigma-Aldrich) for 4 hours at 37°C in the presence of GolgiStop and GolgiPlug (BD Biosciences). All intracellular staining was performed using intracellular Fixation/Permeabilization buffer set (eBioscience/Thermo Fisher) according to the manufacturer's instructions. Live-cell detection was performed using the LIVE/DEAD Fixable Aqua Dead Cell Stain Kit (Life Technologies).

FACS data were acquired using an LSRFortessa flow cytometer (BD Biosciences) and analyzed using FlowJo software (version 10x, Tree Star).

Statistical analyses

Statistical analyses were performed in GraphPad Prism v7. P values were calculated with one-way ANOVA and Kruskal–Wallis posttest.

IL2Cx with anti–PD-1 mAb controls established tumors in a lung adenocarcinoma model

In patients with lung cancer, inhibition of the PD-1/PD-L1 pathway has proven to be efficient and safe, leading to the approval of these treatments (16, 17). Nevertheless, not all patients respond, and combination with IL2Cx, which activates NK and CD8+ T cells, could improve treatment efficacy. Thus, we evaluated the therapeutic effect of IL2Cx (made of IL2/a-IL2 mAb MAB602) alone or combined with an anti–PD-1 blocking mAb in an autochthonous lung tumor model. With this aim, we used the spontaneous tumor model described by DuPage and colleagues (14), in which autochthonous lung tumors are induced by lentiviral infection of a limited number of somatic cells in which oncogenesis is stimulated by recombination of LoxP sites, leading to the activation of one oncogene (Kras) and deletion of one suppressor gene (p53; KP mice). This model not only recapitulates the most frequent gene mutations found in human lung tumors, but also mimics features of the human situation, such as vasculature, tumor architecture, and speed of growth. We modified the lentivirus to induce tumor expression of luciferase and one class I (SIINFEKL)– and one class II (DBY)–restricted nominal antigen. To reduce expression of the nominal antigens after virus inoculation and unwanted priming, we used a lentivirus harboring four tandem target sequences of the hematopoietic-specific mir142-3p after the luciferase-Ag cassette, to induce its degradation in hematopoietic cells (ref. 15; see Materials and Methods).

KP mice infected with the lentivirus were monitored for tumor development by whole-body bioluminescence imaging. The first tumors were detected at 13 to 26 weeks after virus inoculation. Once mice reached a bioluminescence signal of 1 × 105 photons/second, they were randomized to receive one of the following treatments: PBS, IL2Cx (made of 15,000 IU of IL2 and anti-IL2; MAB602, 2:1 molar ratio), anti–PD-1 (200 μg twice per week), or IL2Cx plus anti–PD-1, for at least 7 weeks, and tumor growth followed (Fig. 1A and B). Bioluminescence detection usually correlated with tumor burden when evaluated by hematoxylin and eosin staining, which also revealed that IL2Cx-treated mice did not develop lung edema, previously associated with high-dose IL2 therapy (ref. 6; Fig. 1B). Single-agent therapies showed little antitumor effect, whereas the combination of IL2Cx with anti–PD-1 controlled tumors (Fig. 1C). No tumor regression was observed in the PBS group, whereas 33% of the mice showed tumor control or regression in the IL2Cx group, 27% in the anti–PD-1 group, and 42% in the combination group (Fig. 1C). Treatment efficacy was associated with more circulating antitumor CD8+ T cells (specific for the MHC class I–restricted peptide, SIINFEKL) and CD4+ T cells (specific for the MHC class II–restricted DBY epitope; Supplementary Fig. S1A) in treated groups than in the PBS group. At the end of the experiment, no changes were observed on the proportions of CD4+ T effector (Teff) or NK cells in lung tissue (Supplementary Fig. S1B and S1C). IL2Cx administration increased the frequency and the cycling of CD8+ CD44High T cells (Fig. 1D), an effect reinforced by combination with anti–PD-1. The frequency of Tregs also increased in mice receiving combination therapy (Fig. 1E), but the CD8+CD44High/Treg ratio was unaltered (Fig. 1F). Thus, the combination of IL2Cx with anti–PD-1 increases CD8+ T-cell infiltration of the lung and controls cancer growth, rendering autochthonous lung tumors more sensitive to immune attack.

Figure 1.

Combination of IL2Cx with PD-1 mAb limits tumor growth in an inducible spontaneous model of lung cancer. KrasG12D p53flox/flox mice were intratracheally inoculated with a lentivirus coding for luciferase, OVA-SIINFEKL, and DBY peptides. Tumor growth was monitored by recording emitted bioluminescence. When quantification of the average total flux reached 1 × 105 photons/second, mice were randomized for the treatments shown in A. B, Bioluminescence quantification in mice over time for two representative responder (R1 and R2) and two nonresponder (NR1 and NR2) mice treated with IL2Cx plus-PD-1 mAb (right pictures) and hematoxylin and eosin staining of the lower lung lobe of the corresponding mice (left pictures). Black arrows indicate the tumor presence in the lung lobe. C, Quantification of tumor size over time expressed as a percentage of change from baseline. The pie chart indicates the percentage of mice showing tumor regression (sustained or diminution of tumor size) from baseline (thick lines), per treatment group. PBS, n = 11; IL2Cx, n = 7; anti–PD-1, n = 7; IL2Cx/anti–PD-1, n = 11. D, Percentages of lung-infiltrating CD8+ CD44High T cells (left) and Ki67+CD8+CD44High T cells (right) among CD45+ cells. E, Percentages of lung-infiltrating Tregs, defined as TCRβ+CD4+ FoxP3+ cells. F, Ratio of CD8/Tregs among lung hematopoietic cells. CD8/Treg ratio was calculated as a frequency of CD8+CD44High T cells among CD45+ cells/frequency of total Tregs among CD45+ cells. P values were calculated using one-way ANOVA and Kruskal–Wallis test. *, P < 0.05; **, P < 0.01. See also Supplementary Fig. S1.

Figure 1.

Combination of IL2Cx with PD-1 mAb limits tumor growth in an inducible spontaneous model of lung cancer. KrasG12D p53flox/flox mice were intratracheally inoculated with a lentivirus coding for luciferase, OVA-SIINFEKL, and DBY peptides. Tumor growth was monitored by recording emitted bioluminescence. When quantification of the average total flux reached 1 × 105 photons/second, mice were randomized for the treatments shown in A. B, Bioluminescence quantification in mice over time for two representative responder (R1 and R2) and two nonresponder (NR1 and NR2) mice treated with IL2Cx plus-PD-1 mAb (right pictures) and hematoxylin and eosin staining of the lower lung lobe of the corresponding mice (left pictures). Black arrows indicate the tumor presence in the lung lobe. C, Quantification of tumor size over time expressed as a percentage of change from baseline. The pie chart indicates the percentage of mice showing tumor regression (sustained or diminution of tumor size) from baseline (thick lines), per treatment group. PBS, n = 11; IL2Cx, n = 7; anti–PD-1, n = 7; IL2Cx/anti–PD-1, n = 11. D, Percentages of lung-infiltrating CD8+ CD44High T cells (left) and Ki67+CD8+CD44High T cells (right) among CD45+ cells. E, Percentages of lung-infiltrating Tregs, defined as TCRβ+CD4+ FoxP3+ cells. F, Ratio of CD8/Tregs among lung hematopoietic cells. CD8/Treg ratio was calculated as a frequency of CD8+CD44High T cells among CD45+ cells/frequency of total Tregs among CD45+ cells. P values were calculated using one-way ANOVA and Kruskal–Wallis test. *, P < 0.05; **, P < 0.01. See also Supplementary Fig. S1.

Close modal

IL2Cx delays tumor growth in the B16F10-OVA melanoma mouse model

To confirm our results in a different tumor type, we switched to the transplantable syngeneic B16F10-OVA melanoma mouse model. The IL2/a-IL2 mAb Mab602 complex can control B16F10 melanoma growth (4, 5). We used an Ova-expressing tumor that allows us to follow the antitumor-specific T-cell response. For this, mice were inoculated with 0.5 × 106 B16-Ova melanoma tumor cells. When tumors were palpable, mice were randomized to different treatments: high-dose IL2 (200,000 IU), IL2Cx (made of 15,000 IU of IL2 and anti-IL2 Mab602 at 2:1 molar ratio), or PBS (Fig. 2A). As in ref. 6, both high-dose (HD) IL2 and the IL2Cx administered at a 13 times lower dose of IL2 (LD) delayed tumor growth (Fig. 2B). IL2 action was monitored in the blood by measuring the increase of activated and highly cycling CD44High CD8+ T cells (Fig. 2C). ELISPOT analysis at day 6 after the beginning of the treatment showed that administration of both HD IL2 or LD IL2 in the form of IL2Cx induced or amplified tumor-specific T cells, as indicated by the increase of IFNγ-producing T cells upon ex vivo restimulation with an OVA peptide or with peptides containing tumor neoepitopes (Fig. 2D). IL2Cx, at lower doses, thus less toxic than HD IL2, induced a delay in B16 tumor growth and an increase of tumor-specific IFNγ-producing T cells.

Figure 2.

IL2Cx therapy delays melanoma growth and increases tumor-specific CD8+ T-cell responses. A, Schematic representation of mouse treatment strategy. Mice with palpable tumors were (i.p.) treated for five consecutive days with IL2 (200,000 IU), IL2Cx (made of 15,000 IU of IL2 and a 2:1 molar ratio of anti-IL2 MAB602), or PBS. B, Tumor growth kinetics. Data, mean ± SEM of one representative experiment with n = 12 (PBS), n = 10 (HD IL2), and n = 13 (IL2Cx). C, Blood from tumor-bearing mice analyzed at day 6 after beginning of the treatments. Shown are representative dot plots of assessed parameters and numbers are percentages of cells in the indicated gates. D, IFNγ ELISPOT on circulating T cells specific of OVA-SIINFKEL peptide and B16F10 melanoma neoepitope pools (Pool 1: M30 + M23 + M49); Pool 2: NB02 + NB08 + M27 + M47). Each dot represents a pool of two mice. Tables show the number of responder and nonresponder mice (R/NR) according to the threshold value (dashed line; SIINFEKL = 70 SFC; Pool 1 = 1.667 SFC and Pool 2 = 12.5 SFC) defined from ROC curves. ROC curves were calculated using data from all ELISPOT experiments analyzed (n = 67). SFC, spot-forming cells. E–H, Mice harboring B16F10-OVA melanoma tumors were treated as described in E or G. Tumor growth kinetics are shown in F and H. Data, mean ± SEM of n = 3 or 4 per group. P values were calculated using one-way ANOVA and Kruskal–Wallis test. NS, not significant; *, P < 0.05; **, P < 0.01. See also Supplementary Fig. S2.

Figure 2.

IL2Cx therapy delays melanoma growth and increases tumor-specific CD8+ T-cell responses. A, Schematic representation of mouse treatment strategy. Mice with palpable tumors were (i.p.) treated for five consecutive days with IL2 (200,000 IU), IL2Cx (made of 15,000 IU of IL2 and a 2:1 molar ratio of anti-IL2 MAB602), or PBS. B, Tumor growth kinetics. Data, mean ± SEM of one representative experiment with n = 12 (PBS), n = 10 (HD IL2), and n = 13 (IL2Cx). C, Blood from tumor-bearing mice analyzed at day 6 after beginning of the treatments. Shown are representative dot plots of assessed parameters and numbers are percentages of cells in the indicated gates. D, IFNγ ELISPOT on circulating T cells specific of OVA-SIINFKEL peptide and B16F10 melanoma neoepitope pools (Pool 1: M30 + M23 + M49); Pool 2: NB02 + NB08 + M27 + M47). Each dot represents a pool of two mice. Tables show the number of responder and nonresponder mice (R/NR) according to the threshold value (dashed line; SIINFEKL = 70 SFC; Pool 1 = 1.667 SFC and Pool 2 = 12.5 SFC) defined from ROC curves. ROC curves were calculated using data from all ELISPOT experiments analyzed (n = 67). SFC, spot-forming cells. E–H, Mice harboring B16F10-OVA melanoma tumors were treated as described in E or G. Tumor growth kinetics are shown in F and H. Data, mean ± SEM of n = 3 or 4 per group. P values were calculated using one-way ANOVA and Kruskal–Wallis test. NS, not significant; *, P < 0.05; **, P < 0.01. See also Supplementary Fig. S2.

Close modal

We considered the effect that dissociation of IL2 from the IL2Cx in vivo might have on Tregs and cancer growth. Most of the studies evaluating the role of Tregs in cancer have assessed the impact of depleting (18) but not of increasing Tregs in the cancer context (19). Thus, we studied the consequences of administering LD IL2 (15,000 IU of IL2, same amount of IL2 present in the complex), an IL2Cx that redirects IL2 action to Tregs (mAb 5344), and the IL2Cx that redirects IL2 action to CD8+ T and NK cells (mAb MAB602). In tumor-free mice, LD IL2 did not cause changes in circulating Tregs and CD8+ T cells. Both IL2Cx variants induced an increase of Tregs and enhanced their cycling. Only the IL2Cx that redirect IL2 action to CD8+ T and NK cells increased the proportion and the cycling of circulating CD8+ CD44High T cells (Supplementary Fig. S2A–S2C). In tumor-bearing mice, neither LD IL2 nor the IL2/anti-IL2 mAb MAB602 Cx accelerated tumor growth (Fig. 2E–H). Thus, an unwanted boosting effect of free IL2 on Tregs during cancer appears unlikely. These data indicate that although IL2/anti-IL2 MAB602 Cx boosts circulating Tregs (Supplementary Fig. S2C), it also delays tumor growth (Fig. 2H).

IL2Cx enhances therapeutic efficacy of immune-checkpoint-blocking mAbs

IL2Cx and checkpoint-blocking mAbs each alone have limited efficacy (Fig. 2; ref. 13). We observed a synergistic effect when both were used in combination in mice with palpable B16-OVA tumors (Fig. 3A–C). Figure 3D and E, depicting the results of five pooled experiments, show that monotherapy with anti–CTLA-4 was inefficient; blockade of the PD-1 pathway alone and IL2Cx alone both conferred only intermediate control of tumor growth. The combination treatments were more efficient, as reflected by a significant delay in the tumor development (Fig. 3D). The maximum reduction of the mean tumor volume (measured at the end of the experiment, between days 16 and 20) was obtained with the combinatorial treatments, reaching a 6-fold diminution for the IL2Cx with anti–CTLA-4 and 4-fold with PD-1 blockade relative to the PBS group (Fig. 3E). Mice receiving either single or combined treatments showed no signs of toxicity, such as hair or weight loss. Overall, these results show the greater antitumor effect of the combination therapies over the IL2Cx or inhibitory checkpoint blockade monotherapies.

Figure 3.

Combination of IL2Cx with anti-CKP enhances therapeutic efficacy against established B16F10-OVA melanoma tumors. A, Schematic representation of mouse treatment strategy. Mice with palpable tumors were treated (i.p.) with IL2Cx (gray arrows tips) for 5 days, anti–PD-1/anti–PD-L1 or anti–CTLA4 (200 μg, gray thin arrows) three times per week or PBS. IL2Cx were made as described in Fig. 2. Mice received a total of 7 doses of anti–PD-1/anti–PD-L1 or anti–CTLA4. B and C, Tumor growth kinetics and survival (Kaplan–Meier curves) from one representative experiment out of 5 (n = 6 mice per group). D, Tumor growth kinetics from 5 pooled independent experiments. Data are represented as mean ± SEM of n = 5–6 mice per group per experiment including a total of 30 mice. E, Tumor volume at endpoint from experiments shown in D. P values were calculated using one-way ANOVA and Kruskal–Wallis test (D and E). NS, not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. See also Supplementary Fig. S3.

Figure 3.

Combination of IL2Cx with anti-CKP enhances therapeutic efficacy against established B16F10-OVA melanoma tumors. A, Schematic representation of mouse treatment strategy. Mice with palpable tumors were treated (i.p.) with IL2Cx (gray arrows tips) for 5 days, anti–PD-1/anti–PD-L1 or anti–CTLA4 (200 μg, gray thin arrows) three times per week or PBS. IL2Cx were made as described in Fig. 2. Mice received a total of 7 doses of anti–PD-1/anti–PD-L1 or anti–CTLA4. B and C, Tumor growth kinetics and survival (Kaplan–Meier curves) from one representative experiment out of 5 (n = 6 mice per group). D, Tumor growth kinetics from 5 pooled independent experiments. Data are represented as mean ± SEM of n = 5–6 mice per group per experiment including a total of 30 mice. E, Tumor volume at endpoint from experiments shown in D. P values were calculated using one-way ANOVA and Kruskal–Wallis test (D and E). NS, not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. See also Supplementary Fig. S3.

Close modal

IL2Cx together with anti-CKP increases tumor infiltration by CD8+ T cells

Given that IL2Cx, anti–CTLA-4, or anti–PD-1/anti–PD-L1 may be acting by different mechanisms, we explored the contribution of different immune cell subsets to the efficacy of the combined treatments by flow cytometry analysis of tumor infiltrate. To verify the interpretation of our results, we assessed the relative contribution of blood circulating and tumor-resident lymphocytes among cells retrieved from tumor cell suspensions. Regardless of the treatment, more than 98% of the analyzed hematopoietic cells (total CD45+ cells) are tumor tissue-infiltrating cells (Supplementary Fig. S3).

First, we analyzed the effect of the treatments on the tumor microenvironment. Tumors from PBS-treated mice were little infiltrated by hematopoietic cells (mean ± SD of 8.5% ± 2.7% of CD45+ cells among live cells, n = 10), as previously described for this nonimmunogenic tumor (ref. 17; Supplementary Fig. S4A). Compared with the PBS group, treatment with IL2Cx alone or PD-1 pathway blockade alone did not induce a significant increase in the tumor infiltrate [mean ± SD of 12.3% ± 3.4% (n = 10), and 11.2% ± 2.8% (n = 9) of CD45+ cells among live cells, respectively]. Other treatments promoted a significant recruitment of immune cells [mean ± SEM of 13.1% ± 5.2% of CD45+ cells among live cells for anti–CTLA-4, P < 0.05 (n = 10); 17.7% ± 7.0% for IL2Cx/anti–CTLA-4, P < 0.001(n = 10); and 18.6% ± 5.6% for IL2Cx/PD-1 blockade, P < 0.0001 (n = 10)].

Then, we evaluated the impact of the different therapies on CD8+ T cells, which mediate antitumor immunity. Compared with the PBS-treated group, all therapies except PD-1 blockade alone induced a significant increase in the percentage of CD8+ TILs among the CD45+ infiltrate, the vast majority being CD44High, in accordance with the finding that only effector cells reach the tumor (Supplementary Fig. S4A and S4B). Moreover, only mice receiving the combination therapies showed significant higher absolute numbers of CD8+ CD44High TILs (Fig. 4A). To study CD8+ T-cell function, we analyzed T-cell division and IFNγ production. We observed that compared with mice receiving PBS, IL2Cx alone and the combination therapies induced an increase in the division of CD8+ CD44High TILs (Fig. 4B) and in circulating OVA-specific CD8+ T cells, as well as in tumor neoepitope-specific T cells (Fig. 4C).

Figure 4.

Combination of IL2Cx with anti-CKP reinvigorates tumor-infiltrating CD8+ T cells. Mice harboring B16F10-OVA melanoma tumors were treated as described in Fig. 3A. Tumors and blood were analyzed 6 days after the beginning of the treatment. A, Quantification of effector CD8+ TILs expressed as number of CD8+ CD44High cells per gram of tumor. B, Frequency (%) of Ki67+ cells among tumor-infiltrating CD8+ CD44High cells. C, IFNγ ELISPOT of circulating T cells against OVA-SIINFEKL peptide (top) and B16-F10 melanoma neoepitopes Pool1 (M30 + M23 + M49; middle) and Pool 2 (NB02 + NB08 + M27 + M47; bottom), analyzed as in Fig. 2. SFC, spot-forming cells. D, Representative dot plots showing the expression of Eomes and PD-1 among infiltrating CD8+ CD44High T cells (top), and of Ki67 and Granzyme B (GzmB) among Eomes+ PD-1+ cells (bottom). Numbers indicate the frequency (%) of cells in each quadrant. E, Frequency (%) of GzmB+Ki67+ T cells among PD-1+Eomes+CD8+CD44High T cells infiltrating the tumor. F, Frequency (%) of GzmBKi67 T cells, among PD-1+Eomes+CD8+CD44High T cells infiltrating the tumor. Data are a pool of three (A–C) or two (D–F) independent experiments. P values were calculated using one-way ANOVA and Kruskal–Wallis test or Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. See also Supplementary Figs. S3 and S4.

Figure 4.

Combination of IL2Cx with anti-CKP reinvigorates tumor-infiltrating CD8+ T cells. Mice harboring B16F10-OVA melanoma tumors were treated as described in Fig. 3A. Tumors and blood were analyzed 6 days after the beginning of the treatment. A, Quantification of effector CD8+ TILs expressed as number of CD8+ CD44High cells per gram of tumor. B, Frequency (%) of Ki67+ cells among tumor-infiltrating CD8+ CD44High cells. C, IFNγ ELISPOT of circulating T cells against OVA-SIINFEKL peptide (top) and B16-F10 melanoma neoepitopes Pool1 (M30 + M23 + M49; middle) and Pool 2 (NB02 + NB08 + M27 + M47; bottom), analyzed as in Fig. 2. SFC, spot-forming cells. D, Representative dot plots showing the expression of Eomes and PD-1 among infiltrating CD8+ CD44High T cells (top), and of Ki67 and Granzyme B (GzmB) among Eomes+ PD-1+ cells (bottom). Numbers indicate the frequency (%) of cells in each quadrant. E, Frequency (%) of GzmB+Ki67+ T cells among PD-1+Eomes+CD8+CD44High T cells infiltrating the tumor. F, Frequency (%) of GzmBKi67 T cells, among PD-1+Eomes+CD8+CD44High T cells infiltrating the tumor. Data are a pool of three (A–C) or two (D–F) independent experiments. P values were calculated using one-way ANOVA and Kruskal–Wallis test or Student t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. See also Supplementary Figs. S3 and S4.

Close modal

We observed that in the B16-OVA model, a high proportion of CD8+ CD44High TILs expressed PD-1 (Supplementary Fig. S4C) and that the tumor cells expressed its ligand PD-L1, though at different amounts depending on the treatment (Supplementary Fig. S4D). During chronic viral inflammation, increased expression of PD-1 has been associated with a state of T-cell exhaustion, characterized by lower effector cytokine production and lower replication. In the cancer setting, exhausted CD8+ T cells coexpress the transcription factor eomesodermin (Eomes) along with PD-1 (20). Using these markers, we observed that the percentage of TILs with an exhausted phenotype (Eomes+ PD-1+ CD44High CD8+) was not significantly different among the various groups of treatment (Fig. 4D; Supplementary Fig. S4E). Notwithstanding, it has been reported that during treatment with anti–PD-1, exhausted cells may undergo a reinvigoration process marked by an upregulation of Ki67 and an increase in granzyme B (GzmB) production (20, 21). As shown in Fig. 4E, upon ex vivo stimulation, most of the CD8+ TILs from mice treated with IL2 alone or in combinatory treatments significantly increased Ki67 and GzmB expression, whereas CD8+ TILs from mice treated with PBS, anti–CTLA-4 alone, or PD-1 pathway blockade alone maintained a pool of cells unable to divide and produce GzmB (Fig. 4F).

Altogether, these results suggest that tumor growth control during IL2Cx administration is associated with a significant increase of the CD8+ T-cell tumor infiltrate (which is higher in combination with anticheckpoints Abs), an increase in magnitude and breadth of tumor-specific T-cell responses and the reinvigoration of exhausted CD8+ TILs.

Therapy with IL2Cx/anti–CTLA-4 combination is associated with higher CD8+ cell/Treg ratio

The antitumor effect of anti–CTLA-4 in mice may be in part due to Treg depletion (22). On the other hand, IL2 can boost Treg numbers and function (23). We analyzed the effect of the combinations on Tregs. As observed in Fig. 5A and B, IL2Cx administration was not associated with an increase in intratumoral Treg frequencies. Blockade of the PD-1 pathway alone or in combination with IL2Cx did not affect Treg proportions. However, administration of the anti–CTLA-4 alone induced a decrease of intratumoral CD4+ FoxP3+ Treg proportions, which was more pronounced in association with the IL2Cx. The proportion of CD4+ Teff cells was not significantly affected by the different treatments (Supplementary Fig. S5A).

Figure 5.

IL2Cx alone or in combination with anti–CTLA-4 increases the CD8/Treg ratio in the tumor. C57BL/6 mice harboring B16F10-OVA tumors were treated as described in Fig. 3A. Flow-cytometry analysis of tumor-infiltrating T cells was done 6 days after the beginning of the treatment. A, Representative dot plots of Foxp3 and CD4+ expression on CD45+ cells from the tumor and tumor-draining lymph nodes (dLN). Numbers indicate the frequency (%) of Tregs in the indicated gates. B, Frequency (%) of Tregs among CD45+ cells in the tumor (left) and in dLN (right) from A. C, Ratio of CD8+/Tregs in the tumor, calculated as frequency of total CD8+T cells among CD45+ cells/frequency of total Tregs among CD45+ cells. D, Quantification of CTLA-4 median fluorescence intensity (MFI) in the indicated TILs (left). Heat map of the MFI of CTLA4, CD25, ICOS, GITR, and PD-1 on Treg, Teff, and CD8+ T cells in the tumor and dLNs (right). Data are from three (A–C) or one (D) independent experiments. P values were calculated using one-way ANOVA and Kruskal–Wallis test. **, P < 0.01; ****, P < 0.0001. See also Supplementary Fig. S5.

Figure 5.

IL2Cx alone or in combination with anti–CTLA-4 increases the CD8/Treg ratio in the tumor. C57BL/6 mice harboring B16F10-OVA tumors were treated as described in Fig. 3A. Flow-cytometry analysis of tumor-infiltrating T cells was done 6 days after the beginning of the treatment. A, Representative dot plots of Foxp3 and CD4+ expression on CD45+ cells from the tumor and tumor-draining lymph nodes (dLN). Numbers indicate the frequency (%) of Tregs in the indicated gates. B, Frequency (%) of Tregs among CD45+ cells in the tumor (left) and in dLN (right) from A. C, Ratio of CD8+/Tregs in the tumor, calculated as frequency of total CD8+T cells among CD45+ cells/frequency of total Tregs among CD45+ cells. D, Quantification of CTLA-4 median fluorescence intensity (MFI) in the indicated TILs (left). Heat map of the MFI of CTLA4, CD25, ICOS, GITR, and PD-1 on Treg, Teff, and CD8+ T cells in the tumor and dLNs (right). Data are from three (A–C) or one (D) independent experiments. P values were calculated using one-way ANOVA and Kruskal–Wallis test. **, P < 0.01; ****, P < 0.0001. See also Supplementary Fig. S5.

Close modal

Therapeutic efficacy of anticancer immunotherapies is associated with increased CD8/Treg ratios (22, 24). In our model, only administration of anti–CTLA-4 induced an increase of the tumoral CD8+/Treg ratio compared with untreated mice. This increase was greatest when anti–CTLA-4 was combined with IL2Cx (Fig. 5C).

To better understand where CTLA-4–mediated Treg depletion was occurring, we also quantified Treg proportions in the tumor-draining lymph nodes. As observed in Fig. 5A and B, Treg frequencies were only reduced in the tumor. Selective depletion of a specific T-cell population may be due to the differential expression density of the target molecule (25). To explore this hypothesis, we quantified the median fluorescence intensity (MFI) value of CTLA-4 on the T cells in the different organs (Fig. 5D). Tregs from the tumor of PBS-treated mice expressed the highest MFI values of CTLA-4, compared with CD4+ Teff and CD8+ T cells, which expressed less CTLA-4. Upon IL2Cx administration, CTLA-4 MFI was not significantly modified, and anti–CTLA-4 administration induced a reduction on its target expression on Tregs, probably due to internalization or to blockade of the target CTLA-4 molecule. T cells in the draining lymph nodes barely expressed the CTLA-4 molecule, validating our hypothesis that CTLA-4 mAb-selective depletion of tumor-infiltrating Tregs is likely due to the high expression of its target uniquely on Tregs in this location. We extended this analysis to other immune checkpoints and showed (Fig. 5D; quantified in Supplementary Fig. S5B–S5E) that Tregs in the tumor showed the highest MFI not only of CTLA-4, but also of ICOS, and GITR, compared with Treg in lymph nodes, and to CD4+ Teff and CD8+ T cells in all organs. Conversely, CD8+ T cells in the tumor expressed the most PD-1. As previously described (23, 26), CD25 expression on Tregs increased upon IL2 administration; this increase serves as a biomarker of IL2 in vivo activity.

Overall, compared with anti–PD-1 action, additional immune modulation is achieved by anti–CTLA-4, which specifically depletes intratumoral Tregs and tips the effector/regulatory balance in the tumor in favor of CD8+ T cells, an imbalance further amplified when anti–CTLA-4 is combined with IL2Cx.

IL2Cx with anti–CTLA-4 enhances NK cell activation and increases NK cell/Treg ratio

NK cells, which are highly cytotoxic and produce proinflammatory cytokines, are effectors of antitumor immune responses. Thus, we investigated the effect of the combinatorial therapies on tumor-infiltrating NK cells, which are known to be sensitive to IL2 (27). Mice were treated as described in Fig. 3A and sacrificed 6 days after the beginning of the treatment. As observed in Fig. 6A and B (and Supplementary Fig. S6A), the IL2Cx/anti–CTLA-4 combination induced the largest increase in the number of NK cells and in the proportion of IFNγ-producing tumor-infiltrating NK cells. Otherwise, IL2Cx alone and combined with the checkpoint blockers enhanced NK division and activation, as reflected by the significant increase in the proportion of Ki67+ NK cells, as well as by the increment in the expression of KLRG1 and the stimulatory receptor NKG2D by these cells (Fig. 6C and D; Supplementary Fig. S6B–S6D).

Figure 6.

IL2Cx in combination with anti–CTLA-4 boosts NK cells and increases the NK cell/Treg ratio in the tumor. Mice harboring B16F10-OVA melanoma tumors were treated as described in Fig. 3A. Flow-cytometry analysis of tumor-infiltrating NK cells was made 6 days after beginning of the treatment. A, Quantification of NK cells infiltrating the tumor expressed as number of cells per gram of tumor. B, Frequency (%) of IFNγ-producing cells among total NK cells infiltrating the tumor. C, Frequency (%) of Ki67+ cells among tumor-infiltrating NK cells. D, Frequency (%) of KLRG1+ cells among total tumor-infiltrating NK cells. E, Maturation status of tumor-infiltrating NK cells. A representative dot plot of CD27 and CD11b staining on total NK cells is shown (panel). Percentages of NK cells with the indicated maturation status (CD11bCD27+→ DP→ CD11b+CD27) are shown. F, Ratio NK cells/Tregs in the tumor, calculated as the frequency of total NK cells/frequency of total Treg among CD45+ cells. Data are from two independent experiments. P values were calculated using one-way ANOVA and Kruskal–Wallis test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. See also Supplementary Fig. S6.

Figure 6.

IL2Cx in combination with anti–CTLA-4 boosts NK cells and increases the NK cell/Treg ratio in the tumor. Mice harboring B16F10-OVA melanoma tumors were treated as described in Fig. 3A. Flow-cytometry analysis of tumor-infiltrating NK cells was made 6 days after beginning of the treatment. A, Quantification of NK cells infiltrating the tumor expressed as number of cells per gram of tumor. B, Frequency (%) of IFNγ-producing cells among total NK cells infiltrating the tumor. C, Frequency (%) of Ki67+ cells among tumor-infiltrating NK cells. D, Frequency (%) of KLRG1+ cells among total tumor-infiltrating NK cells. E, Maturation status of tumor-infiltrating NK cells. A representative dot plot of CD27 and CD11b staining on total NK cells is shown (panel). Percentages of NK cells with the indicated maturation status (CD11bCD27+→ DP→ CD11b+CD27) are shown. F, Ratio NK cells/Tregs in the tumor, calculated as the frequency of total NK cells/frequency of total Treg among CD45+ cells. Data are from two independent experiments. P values were calculated using one-way ANOVA and Kruskal–Wallis test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. See also Supplementary Fig. S6.

Close modal

We also analyzed the impact of the treatments on NK maturation, which is modulated by IL2 (27, 28). For that, based on the expression of CD27 and CD11b, we subdivided NK cells into three subsets with different maturation status (CD27+CD11b →DP →CD27CD11b+; ref. 29). None of the treatments modified the percentage of immature CD27+ CD11b NK cells, but administration of IL2Cx alone and in both combinations significantly increased the more mature CD27+ CD11b+ NK cells, at the expense of the more terminally differentiated/senescent CD27 CD11b+ NK cells (ref. 30; Fig. 6E). Given that both NK and Tregs can be boosted by IL2 (26), that CTLA-4 mAb can deplete Tregs (22), and that Tregs suppress NK cells (31, 32), we investigated the impact of the treatments on the NK cell/Treg ratio. The only treatment that significantly increased the NK cell/Treg ratio was the combination IL2Cx plus anti–CTLA-4 (Fig. 6F). Overall, these data indicate that intratumoral NK cells are sensitive to IL2Cx, which induces their accumulation, division, maturation, and activation, and it is the combination of IL2Cx with anti–CTLA-4 that maximizes this effect. In view of these results and on our previous observation of the anti–CTLA-4 -mediated depletion on Tregs, the increase in the NK cell/Treg ratio observed with the IL2Cx/anti–CTLA-4 combination can be explained by the fact that IL2-boosting effect on NK cells is potentiated in the absence of Tregs, which both compete for IL2 and suppress NK cells (28, 32, 33).

IL2Cx plus anti–CTLA-4 therapeutic effect depends on NK and CD8+ T cells

Our results suggested that the additive effects of the two combinations could involve different cellular effectors. Thus, to evaluate the relative contribution of NK and CD8+ T cells on the therapeutic efficacy of the combined treatments, we performed Ab-mediated selective depletion (Fig. 7A). We verified the efficacy of depletion by periodic immunomonitoring of CD8+ T and NK cells in the blood (Supplementary Fig. S7). Figure 7B–D illustrates the effect of NK or CD8+ T-cell depletion on the efficacy of the combination treatments. In mice receiving the IL2Cx/anti–CTLA-4 combo, both depletion of CD8+ T or of NK cells abrogated treatment efficacy (Fig. 7B and C). In mice receiving the IL2Cx/anti–PD-1/anti–PD-L1 combo, depletion of CD8+ T cells completely abrogated the therapeutic effect of the treatment. However, although NK cell depletion induced a nonstatistically significant diminution in the efficacy of the combo, the treatment still delayed tumor growth (Fig. 7B–D). Overall, these results indicate that both combinations work through CD8+ T-cell activation, but only the IL2Cx/anti–CTLA-4 combination is dependent on NK cells to be effective.

Figure 7.

Depletion of CD8+ T or NK cells on B16F10-OVA tumor-bearing mice treated with IL2Cx and antibodies to checkpoint pathways. A, Schematic of the treatment strategy for B16F10-OVA tumor–bearing mice. NK cell and CD8 T-cell depletion was started once tumors were palpable and continued during the experiment. B, Tumor growth kinetics of individual mice. Dashed curves represent the tumor growth of mice under NK cell or CD8+ T-cell depletion. C and D, Tumor growth kinetics represented as a mean SEM of n = 7–8 per group from one representative experiment out of two. P values were calculated using one-way ANOVA and Kruskal–Wallis test. NS, not significant; *, P < 0.05; **, P < 0.01; See also Supplementary Fig. S7.

Figure 7.

Depletion of CD8+ T or NK cells on B16F10-OVA tumor-bearing mice treated with IL2Cx and antibodies to checkpoint pathways. A, Schematic of the treatment strategy for B16F10-OVA tumor–bearing mice. NK cell and CD8 T-cell depletion was started once tumors were palpable and continued during the experiment. B, Tumor growth kinetics of individual mice. Dashed curves represent the tumor growth of mice under NK cell or CD8+ T-cell depletion. C and D, Tumor growth kinetics represented as a mean SEM of n = 7–8 per group from one representative experiment out of two. P values were calculated using one-way ANOVA and Kruskal–Wallis test. NS, not significant; *, P < 0.05; **, P < 0.01; See also Supplementary Fig. S7.

Close modal

Successful anticancer immunotherapies aimed at inducing effective antitumor T-cell responses have delivered impressive clinical results, but only in some patients and in some tumor types (34, 35). Combinatory strategies represent powerful therapeutic tools (36). Thus, we evaluated the efficacy of combining a T-cell growth factor, IL2, formulated as an IL2/anti-IL2 complex, with clinically approved immune- checkpoint inhibitors. In an autochthonous lung adenocarcinoma model, we show that the IL2Cx/anti–PD-1 combination durably controls tumor growth and induces lung infiltration by activated CD8+ T cells. Also, in a transplantable melanoma model refractory to monotherapy with immune-checkpoint blockers we found that IL2Cx synergizes with anti–CTLA-4 and anti–PD-1/anti–PDL-1, improving the clinical outcome of the monotherapies. In addition, we show that combination of IL2Cx with PD-1 or CTLA-4 pathway blockade acts by different cellular mechanisms, differentially targeting CD8+ T, NK, and Tregs.

High-dose IL2 was the first immunotherapy assessed for the treatment of human cancer, achieving similar objective responses in metastatic melanoma as anti-CKP treatment (15%; refs. 37, 38). However, fewer than 10% of eligible patients receive this potentially curative treatment, likely due to its associated toxicity and need for hospitalization (3, 39), as well as concern about the expansion of Tregs. Consequently, different approaches have been tried to bring IL2 back to the clinics, trying to bypass Treg activation and to improve IL2 pharmacodynamics; and some of these approaches are under clinical evaluation. These strategies include, among others, a mutated IL2 superkine with increased affinity for IL2Rβγc, a mutein IL2 fused to tumor-specific Abs (40), an IL2 bound to releasable polyethylene glycol chains (41); and IL2Cx (4) that have longer half-life and improved pharmacodynamics. Our results, showing that administration of either the IL2Cx that activates effector cells, low doses of IL2 (that could be released upon dissociation from anti-IL2), or even an IL2Cx designed to preferentially stimulate Tregs (42–44), did not affect tumor growth, casting doubt on a potential unwanted stimulation of Tregs. Consequently, IL2-based therapeutic approaches in general, and IL2Cx, especially in combination with immune-checkpoint blockers, may be useful in addition to the existing arsenal of antitumor immunotherapies.

Few studies have evaluated the efficacy of immunomodulatory treatments in genetic mouse models. In a model of spontaneous skin melanoma (NrasQ61K Ink4a–/–), IL2Cxs have shown some efficacy as monotherapy (7) and greater efficacy when combined with an epigenetic modulator (8). In another model of autochthonous melanoma (Braf/Pten model), antitumor effects required a multidrug therapy including a tumor antigen–targeting antibody, IL2, anti–PD-1, and a T-cell vaccine, which activated innate and adaptive immune responses (45). Finally, another study using a genetic model of lung adenocarcinoma showed that the KP-OVA lung adenocarcinoma model, which was non–T-cell infiltrated, was refractory to an anti–PD-1 plus anti–CTLA-4 combination therapy, and only combination of these two mAbs with immunogenic drugs oxaliplatin and cyclophosphamide induced antitumor CD8+ T cells and controlled tumor progression (46). In light of these results underscoring the elevated resistance of this tumor model to checkpoint blockade therapy, our data showing that IL2Cx increases the sensibility of autochthonous lung tumors to anti–PD-1 provide proof of concept of the efficacy of this combined therapy for lung tumors. The lack of observed toxicity, plus the obtained mechanistic insight on the mechanism of action of this therapeutic combination that increases tumor infiltration by CD8+ T cells and induces systemic tumor-specific CD4+ and CD8+ T-cell responses, provides a rationale for the design of combination therapies that should increase the response rate of patients to immune-checkpoint therapy.

Blockade of the inhibitory PD-1/PD-L1 pathway has been reported to reinvigorate chronically stimulated exhausted CD8+ T cells during lymphocytic choriomeningitis virus (LCMV) infection (47, 48). Similar results were later observed during cancer immunotherapy in mice models and in patients treated with mAbs to PD-1 (49, 50). The benefits of combining IL2 with PD-1/PD-L1 blockade were explored in the LCMV model, in which IL2 synergized with anti–PD-L1 treatment, improving CD8+ T-cell viral-specific responses and decreasing viral load (21). Our results show that IL2 (in the form of IL2Cx) can synergize with immune-checkpoint blockade to rescue CD8+ T cells function in cancer. Indeed, IL2Cx increased the breadth of CD8+ T-cell responses, including reactivities against tumor neoepitopes, boosted CD8+ T-cell division, and reinvigorated exhausted CD8+ TILs. This action was reinforced by an increase in tumor infiltration by CD45+ cells and CD8+ activated T cells when IL2Cx was combined with blockade of immune checkpoints.

CTLA-4 blockade during cancer is associated with increased CD8+ T cell/Treg ratios and tumor rejection. Vaccination of B16-bearing mice with a GM-CSF–transduced tumor cell vaccine (GVAX) induced T-cell activation and infiltration, an effect potentiated by CTLA-4 blockade (24). Additional blockade of the PD-1/PD-L1 pathway allowed further tumor-specific T-cell activation (51). The antitumoral activity of anti–CTLA-4 required binding of the Ab to Fcγ receptors, which mediate the reduction of Tregs at the tumor site (22, 52). In the preclinical cancer setting, one study reported CTLA-4 blockade combined with IL2 administration (53). Although this work used IL2 (and not IL2Cx) and different doses and treatment timing (beginning of treatment at day 3 after tumor implantation) than our study, they observed, as we did, a synergistic effect of the combination on tumor growth delay and on the increase in tumor-infiltrating CD8+ T and NK cells. However, they did not observe Treg depletion when an mAb to a-CTLA-4 was combined with IL2, indicating that CTLA-4 mAb–mediated Treg depletion can be differently modulated when administered with plain IL2 or with IL2 in the form of IL2Cx. Given our results suggesting that CTLA-4 mAb–mediated depletion correlates with the high expression of CTLA-4 on Tregs, it could be interesting to compare CTLA-4 kinetics of expression on intratumoral Tregs during these different therapeutic interventions.

The combination of radiation with CTLA-4 and PD-L1 blockade in both mice and human works by an increase in the CD8+ T cell/Treg ratio induced by CTLA-4 blockade, and by reversion of T-cell exhaustion by PD-L1 blockade; radiation contributed to enhanced diversity of the tumor T-cell repertoire (20). Here, we observed that (i) the IL2Cx/anti–PD-1/PD-L1 combination worked through increasing tumor infiltration by reinvigorated CD8+ T cells; and (ii) that the IL2Cx/anti–CTLA-4 therapy was the only combination dependent upon CD8+ T and NK cells, as depletion of either of these cells disabled the antitumoral effect of this combination. Indeed, we report that only the IL2Cx/anti–CTLA-4 combination induced an increased NK cell/Treg ratio in the tumor, which was due to the diminution of tumor-infiltrating Tregs (likely mediated by anti–CTLA-4), and the accumulation of NK cells (likely mediated by the exogenously administrated IL2Cx and the increased availability of endogenous IL2 secondary to decreased consumption by Tregs). During homeostasis, depletion of Tregs using an anti-CD25 can boost NK cell proliferation and cytotoxicity in vivo (31). Gasteiger and colleagues (27) proposed that Tregs can suppress mature NK cell function via TGFβ and restrain NK cell maturation by IL2 consumption. These two works support our hypothesis that CTLA-4 mAb–mediated Treg depletion in the tumor facilitates NK cell accumulation and maturation.

B16F10 melanoma tumors recruit CD27+CD11b+ NK cells with low proliferative capacity, reduced KLRG1 and NKG2D expression, and low IFNγ and GM-SCF production (54), suggesting reduced effector functions. Our results showed that IL2Cx alone or combined with immune-checkpoint blockers also increased the frequency of CD27+CD11b+ NK cells, which represented the predominant NK subset in the tumor. However, the phenotype of NK cells was modified by the treatments, as KLRG1 and NKG2D expression was increased, and proliferation and IFNγ production was enhanced, likely overcoming the inhibitory phenotype induced by the tumor described by these authors. Overall, these results suggest that depletion of Tregs with concomitant IL2-mediated activation of NK cells could complement immune-checkpoint blockade for cancer therapy.

In conclusion, our data suggest that IL2-Cx acts on T and NK cells by boosting their expansion and effector function. Blockade of the PD-1/PD-L1 pathway reinforces IL2Cx-induced reinvigoration of exhausted CD8+ T cells. Anti–CTLA-4 also acts by depleting Tregs, which in turn affects CD8+ T and NK cells, increasing the CD8+ T cell/Treg and NK cell/Treg ratios and unleashing the effector cells from Treg suppression. Although activation of CD8+ T cells accounts for most of the efficacy of the IL2Cx/PD-1 pathway blockade combination treatment, CD8+ T-cell reinvigoration by the IL2Cx/anti–CTLA-4 combination is not sufficient to control tumor progression, as the latter combination was the only one dependent on NK cell activation. Thus, our results clarify the cellular and molecular consequences that follow from the combination of IL2Cx with CTLA-4 or PD-1/PD-L1 blockade, and should help refine combinatory cancer immunotherapies.

No potential conflicts of interest were disclosed.

Conception and design: P. Caudana, N.G. Núñez, R. Alonso, L.L. Niborski, E. Piaggio

Development of methodology: O. Lantz, C. Sedlik, E. Piaggio

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): N.G. Núñez, P. De La Rochere, A. Pinto, O. Lantz, C. Sedlik, E. Piaggio

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): N.G. Núñez, P. De La Rochere, A. Pinto, J. Denizeau, E. Piaggio

Writing, review, and/or revision of the manuscript: P. Caudana, R Alonso, L.L. Niborski, O. Lantz, C. Sedlik, E. Piaggio

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): P. De La Rochere, J. Denizeau, R. Alonso, E. Piaggio

Study supervision: E. Piaggio

This work was supported by the Fondation Chercher et Trouver-2016, Institute Curie, Institut National de la Santé et de la Recherche Médicale, Association pour la Recherche sur le Cancer (ARC PJA 20131200444); the Agence Nationale pour la Recherche (ANR-16-CE18-0002-02 and ANR-10-IDEX-0001-02 PSL); Labex DCBIOL (ANR-10-IDEX-0001-02 PSL and ANR-11-LABX0043), SIRIC INCa-DGOS-Inserm_12554, Center of Clinical Investigation (CIC IGR-Curie 1428). N.G. Núñez was supported by a fellowship from Ligue Nationale Contre le Cancer and P. Caudana by the International PhD program of Institut Curie.

We thank Virginie Dangles-Marie, Celine Daviaud, Isabelle Grandjean, Mikael Garcial, and Cedrik Pauchard, the mouse facility technicians and flow cytometry core at Institut Curie. We thank Sebastian Amigorena and Clotilde Théry for discussions and critical review of the manuscript and Nathalie Amzallag for technical assistance.

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.
Rosenberg
SA
,
Lotze
MT
,
Muul
LM
,
Leitman
S
,
Chang
AE
,
Ettinghausen
SE
, et al
Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer
.
N Engl J Med
1985
;
313
:
1485
92
.
2.
Sakaguchi
S
,
Sakaguchi
N
,
Shimizu
J
,
Yamazaki
S
,
Sakihama
T
,
Itoh
M
, et al
Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance
.
Immunol Rev
2001
;
182
:
18
32
.
3.
Malek
TR
. 
The biology of interleukin-2
.
Annu Rev Immunol
2008
;
26
:
453
79
.
4.
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
.
5.
Létourneau
S
,
van Leeuwen
EMM
,
Krieg
C
,
Martin
C
,
Pantaleo
G
,
Sprent
J
, et al
IL2/anti-IL2 antibody complexes show strong biological activity by avoiding interaction with IL2 receptor α subunit CD25
.
Proc Natl Acad Sci U S A
2010
;
107
:
2171
6
.
6.
Krieg
C
,
Létourneau
S
,
Pantaleo
G
,
Boyman
O
. 
Improved IL2 immunotherapy by selective stimulation of IL2 receptors on lymphocytes and endothelial cells
.
Proc Natl Acad Sci U S A
2010
;
107
:
11906
11
.
7.
Arenas-Ramirez
N
,
Zou
C
,
Popp
S
,
Zingg
D
,
Brannetti
B
,
Wirth
E
, et al
Improved cancer immunotherapy by a CD25-mimobody conferring selectivity to human interleukin-2
.
Sci Transl Med
2016
;
8
:
367ra166
.
8.
Zingg
D
,
Arenas-Ramirez
N
,
Sahin
D
,
Rosalia
RA
,
Antunes
AT
,
Haeusel
J
, et al
The histone methyltransferase Ezh2 controls mechanisms of adaptive resistance to tumor immunotherapy
.
Cell Rep
2017
;
20
:
854
67
.
9.
Topalian
SL
,
Drake
CG
,
Pardoll
DM
. 
Immune checkpoint blockade: a common denominator approach to cancer therapy
.
Cancer Cell
2015
;
27
:
450
61
.
10.
Wolchok
JD
,
Kluger
H
,
Callahan
MK
,
Postow
MA
,
Rizvi
NA
,
Lesokhin
AM
, et al
Nivolumab plus ipilimumab in advanced melanoma
.
N Engl J Med
2013
;
369
:
122
33
.
11.
Zaretsky
JM
,
Garcia-Diaz
A
,
Shin
DS
,
Escuin-Ordinas
H
,
Hugo
W
,
Hu-Lieskovan
S
, et al
Mutations associated with acquired resistance to PD-1 blockade in melanoma
.
N Engl J Med
2016
;
375
:
819
29
.
12.
Chen
P-L
,
Roh
W
,
Reuben
A
,
Cooper
ZA
,
Spencer
CN
,
Prieto
PA
, et al
Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade
.
Cancer Discov
2016
;
6
:
827
37
.
13.
Mosely
SIS
,
Prime
JE
,
Sainson
RCA
,
Koopmann
J-O
,
Wang
DYQ
,
Greenawalt
DM
, et al
Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery
.
Cancer Immunol Res
2017
;
5
:
29
41
.
14.
DuPage
M
,
Dooley
AL
,
Jacks
T
. 
Conditional mouse lung cancer models using adenoviral or lentiviral delivery of
Cre recombinase
.
Nat Protoc
2009
;
4
:
1064
72
.
15.
Alonso
R
,
Flament
H
,
Lemoine
S
,
Sedlik
C
,
Bottasso
E
,
Péguillet
I
, et al
Induction of anergic or regulatory tumor-specific CD4 + T cells in the tumor-draining lymph node
.
Nat Commun
2018
;
9
:
2113
.
16.
Gettinger
S
,
Horn
L
,
Jackman
D
,
Spigel
D
,
Antonia
S
,
Hellmann
M
, et al
Five-year follow-up of nivolumab in previously treated advanced non–small-cell lung cancer: results from the CA209-003 study
.
J Clin Oncol
2018
;
36
:
1675
84
.
17.
Hellmann
MD
,
Ciuleanu
T-E
,
Pluzanski
A
,
Lee
JS
,
Otterson
GA
,
Audigier-Valette
C
, et al
Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden
.
N Engl J Med
2018
;
378
:
2093
104
.
18.
Arce Vargas
F
,
Furness
AJS
,
Solomon
I
,
Joshi
K
,
Lesko
MH
,
Miranda Rota
E
, et al
Fc-Optimized anti-CD25 depletes tumor-infiltrating regulatory T cells and synergizes with PD-1 blockade to eradicate established tumors
.
Immunity
2017
;
46
:
577
86
.
19.
Churlaud
G
,
Jimenez
V
,
Ruberte
J
,
Amadoudji Zin
M
,
Fourcade
G
,
Gottrand
G
, et al
Sustained stimulation and expansion of Tregs by IL2 control autoimmunity without impairing immune responses to infection, vaccination and cancer
.
Clin Immunol
2014
;
151
:
114
26
.
20.
Twyman-Saint Victor
C
,
Rech
AJ
,
Maity
A
,
Rengan
R
,
Pauken
KE
,
Stelekati
E
, et al
Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer
.
Nature
2015
;
520
:
373
7
.
21.
West
EE
,
Jin
H-T
,
Rasheed
A-U
,
Penaloza-MacMaster
P
,
Ha
S-J
,
Tan
WG
, et al
PD-L1 blockade synergizes with IL2 therapy in reinvigorating exhausted T cells
.
J Clin Invest
2013
;
123
:
2604
15
.
22.
Simpson
TR
,
Li
F
,
Montalvo-Ortiz
W
,
Sepulveda
MA
,
Bergerhoff
K
,
Arce
F
, et al
Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti–CTLA-4 therapy against melanoma
.
J Exp Med
2013
;
210
:
1695
710
.
23.
Grinberg-Bleyer
Y
,
Baeyens
A
,
You
S
,
Elhage
R
,
Fourcade
G
,
Gregoire
S
, et al
IL2 reverses established type 1 diabetes in NOD mice by a local effect on pancreatic regulatory T cells
.
J Exp Med
2010
;
207
:
1871
8
.
24.
Quezada
SA
,
Peggs
KS
,
Curran
MA
,
Allison
JP
. 
CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells
.
J Clin Invest
2006
;
116
:
1935
45
.
25.
Arce Vargas
F
,
Furness
AJS
,
Litchfield
K
,
Joshi
K
,
Rosenthal
R
,
Ghorani
E
, et al
Fc effector function contributes to the activity of human anti-CTLA-4 antibodies
.
Cancer Cell
2018
;
33
:
649
63
.
e4
.
26.
Baeyens
A
,
Pérol
L
,
Fourcade
G
,
Cagnard
N
,
Carpentier
W
,
Woytschak
J
, et al
Limitations of IL2 and rapamycin in immunotherapy of type 1 diabetes
.
Diabetes
2013
;
62
:
3120
31
.
27.
Gasteiger
G
,
Hemmers
S
,
Bos
PD
,
Sun
JC
,
Rudensky
AY
. 
IL2–dependent adaptive control of NK cell homeostasis
.
J Exp Med
2013
;
210
:
1179
87
.
28.
Sitrin
J
,
Ring
A
,
Garcia
KC
,
Benoist
C
,
Mathis
D
. 
Regulatory T cells control NK cells in an insulitic lesion by depriving them of IL2
.
J Exp Med
2013
;
210
:
1153
65
.
29.
Chiossone
L
,
Chaix
J
,
Fuseri
N
,
Roth
C
,
Vivier
E
,
Walzer
T
. 
Maturation of mouse NK cells is a 4-stage developmental program
.
Blood
2009
;
113
:
5488
96
.
30.
Hayakawa
Y
,
Huntington
ND
,
Nutt
SL
,
Smyth
MJ
. 
Functional subsets of mouse natural killer cells
.
Immunol Rev
2006
;
214
:
47
55
.
31.
Ghiringhelli
F
,
Ménard
C
,
Terme
M
,
Flament
C
,
Taieb
J
,
Chaput
N
, et al
CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor–β–dependent manner
.
J Exp Med
2005
;
202
:
1075
85
.
32.
Littwitz-Salomon
E
,
Akhmetzyanova
I
,
Vallet
C
,
Francois
S
,
Dittmer
U
,
Gibbert
K
. 
Activated regulatory T cells suppress effector NK cell responses by an IL2-mediated mechanism during an acute retroviral infection
.
Retrovirology
2015
;
12
:
66
.
33.
Smyth
MJ
,
Teng
MWL
,
Swann
J
,
Kyparissoudis
K
,
Godfrey
DI
,
Hayakawa
Y
. 
CD4+CD25+ T regulatory cells suppress NK cell-mediated immunotherapy of cancer
.
J Immunol
2006
;
176
:
1582
7
.
34.
Jin
C
,
Zhang
X
,
Zhao
K
,
Xu
J
,
Zhao
M
,
Xu
X
. 
The efficacy and safety of nivolumab in the treatment of advanced melanoma: a meta-analysis of clinical trials
.
Onco Targets Ther
2016
;
9
:
1571
8
.
35.
Zhou
G-W
,
Xiong
Y
,
Chen
S
,
Xia
F
,
Li
Q
,
Hu
J
. 
Anti-PD-1/PD-L1 antibody therapy for pretreated advanced nonsmall-cell lung cancer: a meta-analysis of randomized clinical trials
.
Medicine (Baltimore)
2016
;
95
:
e4611
.
36.
Melero
I
,
Berman
DM
,
Aznar
MA
,
Korman
AJ
,
Gracia
JLP
,
Haanen
J
. 
Evolving synergistic combinations of targeted immunotherapies to combat cancer
.
Nat Rev Cancer
2015
;
15
:
457
72
.
37.
Yang
JC
,
Sherry
RM
,
Steinberg
SM
,
Topalian
SL
,
Schwartzentruber
DJ
,
Hwu
P
, et al
Randomized study of high - dose and low - dose inter leukin - 2 in patients with metastatic renal cancer
.
J Clin Oncol
2003
;
21
:
3127
32
.
38.
Klapper
JA
,
Downey
SG
,
Smith
FO
,
Yang
JC
,
Hughes
MS
,
Kammula
US
, et al
High-dose interleukin-2 for the treatment of metastatic renal cell carcinoma
.
Cancer
2008
;
113
:
293
301
.
39.
Rosenberg
SA
. 
Raising the bar: the curative potential of human cancer immunotherapy
.
Sci Transl Med
2012
;
4
:
127ps8
.
40.
Klein
C
,
Waldhauer
I
,
Nicolini
VG
,
Freimoser-Grundschober
A
,
Nayak
T
,
Vugts
DJ
, et al
Cergutuzumab amunaleukin (CEA-IL2v), a CEA-targeted IL2 variant-based immunocytokine for combination cancer immunotherapy: Overcoming limitations of aldesleukin and conventional IL2-based immunocytokines
.
OncoImmunology
2017
;
6
:
e1277306
.
41.
Charych
D
,
Khalili
S
,
Dixit
V
,
Kirk
P
,
Chang
T
,
Langowski
J
, et al
Modeling the receptor pharmacology, pharmacokinetics, and pharmacodynamics of NKTR-214, a kinetically-controlled interleukin-2 (IL2) receptor agonist for cancer immunotherapy
.
PLoS One
2017
;
12
:
e0179431
.
42.
Kim
M-G
,
Koo
TY
,
Yan
J-J
,
Lee
E
,
Han
KH
,
Jeong
JC
, et al
IL2/Anti-IL2 complex attenuates renal ischemia-reperfusion injury through expansion of regulatory T cells
.
J Am Soc Nephrol
2013
;
24
:
1529
36
.
43.
Smaldini
PL
,
Trejo
F
,
Cohen
JL
,
Piaggio
E
,
Docena
GH
. 
Systemic IL2/anti-IL2Ab complex combined with sublingual immunotherapy suppresses experimental food allergy in mice through induction of mucosal regulatory T cells
.
Allergy
2018
;
73
:
885
95
.
44.
Manirarora
JN
,
Wei
C-H
. 
Combination therapy using IL2/IL2 monoclonal antibody complexes, rapamycin, and islet autoantigen peptides increases regulatory T cell frequency and protects against spontaneous and induced type 1 diabetes in nonobese diabetic mice
.
J Immunol
2015
;
195
:
5203
14
.
45.
Moynihan
KD
,
Opel
CF
,
Szeto
GL
,
Tzeng
A
,
Zhu
EF
,
Engreitz
JM
, et al
Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses
.
Nat Med
2016
;
22
:
1402
10
.
46.
Pfirschke
C
,
Engblom
C
,
Rickelt
S
,
Cortez-Retamozo
V
,
Garris
C
,
Pucci
F
, et al
Immunogenic chemotherapy sensitizes tumors to checkpoint blockade therapy
.
Immunity
2016
;
44
:
343
54
.
47.
Barber
DL
,
Wherry
EJ
,
Masopust
D
,
Zhu
B
,
Allison
JP
,
Sharpe
AH
, et al
Restoring function in exhausted CD8 T cells during chronic viral infection
.
Nature
2006
;
439
:
682
7
.
48.
Blackburn
SD
,
Shin
H
,
Freeman
GJ
,
Wherry
EJ
. 
Selective expansion of a subset of exhausted CD8 T cells by αPD-L1 blockade
.
Proc Natl Acad Sci U S A
2008
;
105
:
15016
21
.
49.
Kamphorst
AO
,
Wieland
A
,
Nasti
T
,
Yang
S
,
Zhang
R
,
Barber
DL
, et al
Rescue of exhausted CD8 T cells by PD-1–targeted therapies is CD28-dependent
.
Science
2017
;
355
:
1423
7
.
50.
Huang
AC
,
Postow
MA
,
Orlowski
RJ
,
Mick
R
,
Bengsch
B
,
Manne
S
, et al
T-cell invigoration to tumour burden ratio associated with anti-PD-1 response
.
Nature
2017
;
545
:
60
5
.
51.
Curran
MA
,
Montalvo
W
,
Yagita
H
,
Allison
JP
. 
PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors
.
Proc Natl Acad Sci U S A
2010
;
107
:
4275
80
.
52.
Du
X
,
Tang
F
,
Liu
M
,
Su
J
,
Zhang
Y
,
Wu
W
, et al
A reappraisal of CTLA-4 checkpoint blockade in cancer immunotherapy
.
Cell Res
2018
;
28
:
416
32
.
53.
Kohlhapp
FJ
,
Broucek
JR
,
Hughes
T
,
Huelsmann
EJ
,
Lusciks
J
,
Zayas
JP
, et al
NK cells and CD8+ T cells cooperate to improve therapeutic responses in melanoma treated with interleukin-2 (IL2) and CTLA-4 blockade
.
J Immunother Cancer
2015
;
3
:
18
.
54.
Paul
S
,
Kulkarni
N
,
Shilpi
,
Lal
G
. 
Intratumoral natural killer cells show reduced effector and cytolytic properties and control the differentiation of effector Th1 cells
.
OncoImmunology
2016
;
5
:
e1235106
.

Supplementary data