Activation of RORγ with synthetic small-molecule agonists has been shown to enhance type 17 effector (CD4+ Th17 and CD8+ Tc17 cells) cell functions and decrease immunosuppressive mechanisms, leading to improved antitumor efficacy in adoptive cell transfer and syngeneic murine tumor models. However, whether Tc17 cells possess intrinsic cytotoxicity and the mechanism they use to lyse target cells is controversial. We report here that Tc17 cells were lytic effectors dependent on perforin and granzyme A. In contrast to Tc1 cells, Tc17 cells resisted activation-induced cell death and maintained granzyme A levels, which conferred the ability to lyse target cells in serial encounters. Thus, although the acute lytic capacity of Tc17 cells could be inferior to Tc1 cells, comparable lysis was achieved over time. In addition to direct lytic activity, Tc17 cells infiltrated early into the tumor mass, recruited other CD8+ T cells to the tumor, and enhanced the survival and lytic capability of these cells during repeated target encounters. Synthetic RORγ agonists further augmented Tc17 survival and lytic activity in vitro and in vivo, controlling tumor growth not only through direct cytotoxicity, but also through recruitment and improved function of other effector cells in the tumor microenvironment, which suggests complementary and cooperate activities for effective immunotherapy.

Retinoic acid receptor–related orphan receptor γ (RORγ) is a master transcription factor that controls the differentiation and maintenance of the CD4+ and CD8+ type 17 effector T-cell subsets, Th17 and Tc17, respectively (1). Type 17 T cells participate in regulation of bacterial, viral, and fungal infections; autoimmune disease; and antitumor immunity (2). Targeting the type 17 signature cytokine IL17 or the generation of type 17 T cells through IL23 has been clinically validated in several autoimmune diseases (3–5). On the other hand, type 17 T cells can mediate potent and durable tumor growth inhibition when transferred to tumor-bearing animals (6–8). Although the prognosis can be poor in some situations where IL17 induces angiogenesis, inflammation, and promotes the initiation and early growth of some tumors (9, 10), the presence of RORγ+ type 17 T cells or their hallmark cytokines such as IL17A and GM-CSF is associated with improved antitumor effects in many cancers (11–17). We have previously reported that only a fraction of human T cells coexpress both RORγ and IL17A, suggesting that RORγ and IL17A may play distinct roles in Tc17-mediated antitumor immunity (18).

The antitumor efficacy of type 17 T cells may be mediated by several mechanisms. These include the conversion of Tc17 to Tc1 effectors, the durability of Tc17 cells and formation of memory cells, direct cytotoxic activity, and the ability of type 17 T cells to recruit other immune cells (19). The plasticity of Tc17 cells to transform to cytolytic, IFNγ-secreting Tc1 cells or IFNγ+IL17+ effector cells has been reported in a number of in vitro and in vivo settings, including in tumor-bearing mice (20, 21). Thus, Tc17 cells serve as a reservoir for Tc1 cell development (22). Type 17 cells in cancer patients produce high concentrations of cytokines, such as IL17A and IL17F, GM-CSF, TNFα, IL2, and IFNγ (19), which contribute to the formation of a proimmune microenvironment. IL17A and IL17F bind to IL17 receptors on endothelial cells, stromal cells, and tumor cells (23) and induce the production of various chemokines such as CCL20, CXCL9, and CXCL10, which recruit monocytes, neutrophils, natural killer (NK) cells, and CD8+ T cells into the tumor microenvironment (24, 25).

Tumor antigen–specific type 17 cells eradicate murine tumors better than type 1–polarized clones (7, 8), and the antitumor effect is long-lasting via the enhanced formation of memory cells capable of responding to tumor cell rechallenge (26). However, in vitro type 17 cells had little or no cytotoxic activity (27–29). In contrast to the well-known killing mechanism for Tc1 cells, little is known for Tc17 cells. Type 17 cells express minimal granzyme B, FasL, or TRAIL (27, 28), but increase their expression of TRAIL and FasL after transfer into mice, which is important for their antiviral responses (27, 30). Although human type 17 cells can produce both IFNγ and IL17 in vivo, murine type 17 cells associated with tumor secrete minimal amount of IFNγ (31). Together, these observations challenge the idea of intrinsic cytotoxicity and a direct antitumor effect of type 17 cells.

Here, we showed that Tc17 cells lysed antigen-specific target cells directly. Tc1 cells utilized granzyme B and, to a less extent, TNF superfamily members such as Fas and TRAIL to lysis target cells. Tc17 cells, on the other hand, expressed high amounts of granzyme A and depended on perforin to kill target cells. Tc17 cells had lesser lytic activity compared with Tc1 at initial antigen encounter; however, they could survive longer and maintained their cytotoxicity over a longer time, particularly upon target cell rechallenge. In addition to cell-intrinsic effects, the presence of Tc17 cells increased the survival of cocultured Tc1 cells. In tumors, Tc17 cells infiltrated the tumor microenvironment earlier than Tc1 cells, and the presence of Tc17 cells correlated with the number of CD8+ cells recruited into the tumor. An RORγ agonist further enhanced Tc17 survival, increased their cytotoxicity in vitro and in vivo, and recruited host CD8+ T cells to the tumor, suggesting the utility of RORγ agonists as an anticancer therapy.

Animals

C57BL/6 and BALB/c mice were purchased from Taconic. OT-I (C57BL/6-Tg(TcraTcrb)1100Mjb/J), Thy1.1 (B6.PL-Thy1a/CyJ), Prf1 KO (C57BL/6-Prf1tm1Sdz/J), and RORγt KO (B6.129P2(Cg)-Rorctm2Litt/J) mice were purchased from The Jackson Laboratory. All animal experiments were conducted according to institutional animal care and safety guidelines and with IACUC approval at the University of Michigan (PRO00007524) and Vet Pathology and IND Services (VPI-001-02).

Reagents

All chemicals were purchased from Sigma, Avanti Polar Lipids, Tocris (R&D Systems), or Enzo Life Sciences. RORγ agonists LYC-54143 were described (18) and synthesized by Lycera. E.G7-OVA and MC38 cells were purchased from ATCC, Mycoplasma-free and used less than passage 10. MC38 cells were stably transfected with full-length OVA (pGen-OVA, GenScript) to generate MC38.OVA.

Mouse Tc17 cell differentiation

Splenocytes from OT-I mice were activated for 4 to 5 days with OVA-derived peptide SIINFEKL (50 ng/mL). For polarizing to Tc17 cells, we added cytokines TGFβ (2.5 ng/mL) and IL6 (10 ng/mL), and for Tc1 cells we added IL12 (10 ng/mL). Alternatively, CD8+ T cells were purified from splenocytes from C57BL/6 or Prf1 knockout (KO) mice using an EasySep Mouse CD8+ T-cell isolation kit from STEMCELL Technologies, and were activated with plate-bound anti-CD3 (2.5 mg/mL), soluble CD28 (2.5 mg/mL), and differentiated into Tc17 cells with TGFβ (2.5 ng/mL) and IL6 (10 ng/mL), or Tc1 cells with IL12 (10 ng/mL).

Flow cytometry

Cell-surface expression of coinhibitory receptors was analyzed by flow cytometry. Effector T-cell differentiation was assessed by flow cytometer after 5-hour stimulation with PMA (100 ng/mL), ionomycin (1 mg/mL) in the presence of brefeldin A (eBioscience). Intracellular staining was performed using labeled antibodies after fixation and permeabilization Buffer Set (eBioscience). Antibodies used for flow cytometry were CD8 (PE, clone REA601; Miltenyi), CD73 (eFluor450, clone eBioTY/11.8; eBioscience), IL17a (FITC, clone TC11-18H10.1; BioLegend), IFNγ (PE, clone XMG1.2; eBioscience), Thy1.1 (eFluor450, clone HIS51; eBioscience), FasL (PE, clone MFL3; BioLegend), and TRAIL (APC, clone N2B2; eBioscience). Data were acquired using a BD FACSCanto II flow cytometer and analyzed by FACSDIVA software.

Real-time PCR

For qPCR analysis, RNA was isolated using an RNeasy mini kit (Qiagen), and mRNA expression was analyzed in StepOnePlus (Life Technologies) real-time PCR instrument using housekeeping gene cyclophilin as internal standards. RNA was first reverse transcribed into cDNA using a High Capacity RNA-to-cDNA kit (Applied Bioscience). qPCR was performed using Power SYBR Green PCR Master Mix (Applied Bioscience) and two-cycle amplification (95°C for 15 seconds and 60°C for 60 seconds) for 40 cycles followed by melting curve. The sequence of primers is listed in Supplementary Table S1.

siRNA knockdown in Tc1 and Tc17 cells

Splenocytes from OT-I mice were activated with peptides and polarizing cytokines toward Tc1 or Tc17 cells for 3 days. Effector T cells were nucleofected with granzyme A siRNA oligos (Dharmacon) using a P4 Nucleofector Kit (Lonza) and the Nucleofector program CM137. Transfected cells were resuspended in their original conditioned media for 2 days before analysis.

In vitro cytotoxicity assays

Tc1 and Tc17 cells were differentiated in vitro as indicated above. E.G7-OVA cells and MC38.OVA target cells were labeled with CFSE (10 μmol/L, Thermo Fisher). Various numbers of Tc17 effector (E) cells were mixed with a fixed number of target (T) cells in a 96-round bottom plate, to achieve effector:target (E:T) ratios of 100:1, 30:1, 10:1, 3:1, and 1:1. Mixed cells were incubated for indicated times at 37°C with 5% CO2. Specific lysis was calculated by % of specific lysis = 100×(1 − experimental (CFSElow/(CFSEhigh+ CFSElow)%)/input (CFSElow/(CFSEhigh + CFSElow)%)) (32).

To examine the contribution of FasL/Fas and TRAIL/TRAILR2 pathways to the cytotoxicity of Tc1 and Tc17 cells, blocking antibodies for FasL (clone Kay-10, BioLegend), TRAIL (clone N2B2, BioLegend), or isotype control (clone RTK4530 and clone RTK2758, BioLegend) were added at the beginning of in vitro cytotoxicity assays.

To measure the cytotoxicity of Tc17 cells generated from wild-type (WT) or Prf1 KO mice, allogeneic splenocytes from BALB/c mice were incubated with LPS (10 μg/mL; ref. 33) or ConA (2 μg/mL; refs. 34, 35) for 2 days and used as target cells.

To measure the cytotoxicity of antigen-encountered Tc1, Tc17, or Tc1 cocultured with Tc17 cells, Tc1 cells were differentiated from OT-I splenocytes and Tc17 from Thy1.1 OT-I splenocytes. A combination of Thy1.1-positive/negative selection and CD8+ T cells selection (STEMCELL Technologies) was used to purify antigen-encountered Tc1 and Tc17 cells for recall response.

Adoptive cell therapy tumor models

E.G7-OVA tumor cells (ATCC) were implanted subcutaneously into the flank of C57/BL6 mice and allowed to grow. In parallel, splenocytes from Thy1.1 OT-I mice were isolated and differentiated into Tc17 cells in vitro in the presence/absence of RORγ agonists for 5 days. Once the tumor was measurable (normally between days 7 and 10 after implant), the expanded T cells were washed out of RORγ agonists, and injected intraperitoneally. Antitumor responses were measured 2 to 3 × per week by caliper measurement. Tumor volume calculation was done as follows: 0.5×(length× (width)2). Mice were taken down after tumor volume reached ethical endpoint of 2,000 mm3.

Data analysis and statistics

In vitro experiments were repeated ≥ 3 × unless otherwise noted in figure legends. Most data presented in figures are mean ± standard deviation (SD) of biological replicates indicated in the figure legends. Statistical significance for in vitro data was determined using an unpaired, two-tailed t test (assume Gaussian distribution, with 95% confidence level). Statistics for in vivo data were done using multiple t tests, Mann–Whitney test (fewer assumptions, desired FDR (Q)–1%) in GraphPad Prism 7. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; and n.s., not significant.

Tc17 display a slow onset lytic mechanism in vitro

Type 17 T cells are described as polyfunctional effector cells that can mediate potent antitumor activity in murine syngeneic models. However, in other models and in vitro, type 17 T cells have been shown to have modest, if any, cytolytic activity. These seemingly inconsistent data may be due to differences in the in vivo microenvironments or different polarization conditions used to generate type 17 cells in vitro. In the literature, a variety of different conditions have been used to generate type 17 cells. Here, we differentiated Tc17 from OT-I cells with a minimal cytokine cocktail of TGFβ and IL6 and compared them with Tc1 cells. We confirmed (18, 36) that Tc17 cells expressed more CD73 and IL17 and less IFNγ than Tc1 cells (Fig. 1A).

Figure 1.

Cytotoxicity of Tc17 cells in vitro and in vivo. A, Characterization of Tc17 cells. IL17a, IFNγ, and CD73 levels were measured by flow cytometry. B, Tc1, but not Tc17, cells displayed direct cytotoxic activity against E.G7-OVA target cells in vitro during a 4-hour CTL assay. Specific lysis was calculated based on input controls. C, Low amount of Tc1, but not Tc17, cells (0.2 × 106 cells/mouse) controlled E.G7-OVA tumor in the ACT model. D, Increasing number of transferred Tc17 cells (5 × 106 cells/mouse) shrank established tumor in the ACT model. E, Both Tc17 and Tc1 cells displayed direct cytotoxic activity against target E.G7-OVA cells in vitro during a 16-hour CTL assay. Data are shown as the mean ± SD of a representative experiment (n = 3 or 4 samples per group for the in vitro experiment, n = 10 mice per group for in vivo experiment). *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001, n.s., not significant (unpaired t test for A, B and E; multiple t tests for C and D).

Figure 1.

Cytotoxicity of Tc17 cells in vitro and in vivo. A, Characterization of Tc17 cells. IL17a, IFNγ, and CD73 levels were measured by flow cytometry. B, Tc1, but not Tc17, cells displayed direct cytotoxic activity against E.G7-OVA target cells in vitro during a 4-hour CTL assay. Specific lysis was calculated based on input controls. C, Low amount of Tc1, but not Tc17, cells (0.2 × 106 cells/mouse) controlled E.G7-OVA tumor in the ACT model. D, Increasing number of transferred Tc17 cells (5 × 106 cells/mouse) shrank established tumor in the ACT model. E, Both Tc17 and Tc1 cells displayed direct cytotoxic activity against target E.G7-OVA cells in vitro during a 16-hour CTL assay. Data are shown as the mean ± SD of a representative experiment (n = 3 or 4 samples per group for the in vitro experiment, n = 10 mice per group for in vivo experiment). *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001, n.s., not significant (unpaired t test for A, B and E; multiple t tests for C and D).

Close modal

We measured the direct cytotoxicity of Tc17 effector cells generated from OT-I mice toward antigen-specific E.G7-OVA lymphoma cells in vitro. After 4 hours of coculture, Tc1 cells killed E.G7-OVA target cells in a dose-dependent manner. However, Tc17 cells were ineffective at any E:T ratio tested (Fig. 1B).

We then measured the cytotoxicity of Tc17 cells in vivo using an adoptive cell transfer (ACT) model. Consistent with the in vitro data, when equal numbers of effector T cells (0.2 × 106 per mouse) were transferred into tumor-bearing mice, Tc1 cells decreased the tumor volume, whereas Tc17 cells were not effective (Fig. 1C). However, increasing the number of Tc17 cells (5 × 106 per mouse) transferred displayed good antitumor activity (Fig. 1D), suggesting that these cells are inefficient cytotoxic T lymphocytes (CTL) but not intrinsically incapable of tumor lysis.

One key difference between in vitro CTL assays and in vivo tumor eradication is the time over which lytic activity is assessed. To address this, we measured in vitro target cell lysis at a later time point. When we incubate effector cells with target cells for 16 hours, Tc17 cells started to show substantial CTL activity (Fig. 1E) and in a dose-dependent manner when higher E:T ratio was utilized (Supplementary Fig. S1A), demonstrating that Tc17 cells have a slow onset lytic mechanism that might be different from Tc1 cells, and the timing used to evaluate Tc17 lytic activity, indeed, influenced their cytotoxicity.

Perforin/granzyme pathway is required for Tc17 cytotoxicity

Multiple mechanisms are used by cytotoxic lymphocytes to recognize and lyse target cells, including perforin/granzyme-mediated cleavage of specific intracellular substrates following antigen/MHC or NKG2D recognition, caspase-dependent apoptosis induced by TNF superfamily members such as Fas and TRAIL, as well as antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis. The slow onset of Tc17 cell cytotoxicity in vitro prompted us to examine the involvement of death ligand/death receptor pathways. Compared with the exocytosis of perforin/granzyme granules, FasL and TRAIL are usually expressed on the cell surface at later time points upon CTL activation and sometimes involve a second wave of de novo synthesis (37, 38).

To test the contribution of FasL/Fas and TRAIL/TRAILR2 pathways to the cytotoxicity of Tc17 cells in vitro, blocking antibodies were used in a 16-hour CTL assay against MC38.OVA target cells which expressed both Fas and TRAILR2 on their surface. Tc17 cells expressed less FasL and TRAIL compared with Tc1 cells (Fig. 2A). We found both Tc1 and Tc17 cells lysed target cells (Fig. 2B), as indicated by fewer target cells remaining after incubation with either effector type (isotype vs. input). Blocking of FasL or TRAIL in Tc1, but not in Tc17 cells, slightly increased the number of remaining target cells (from 19296 ±1,285 to 24,022 ± 3,967 or 28,971 ± 1,501), indicating a minor involvement of FasL and TRAIL pathways for Tc1 cytotoxicity.

Figure 2.

Tc17 cells depend on perforin to lyse target cells. A, Tc17 cells express low levels of FasL and TRAIL. Cell-surface expression of FasL and TRAIL were measured by flow cytometry. B, Blocking of FasL or TRAIL slightly enhance the killing of target cells by Tc1 cells. Tc1 and Tc17 cells were incubated with CSFE-labeled target MC38.OVA cells for 16 hours in the presence of isotype control, or blocking antibodies to FasL or TRAIL at E:T = 3:1. At the end of the experiment, remaining viable target cells were counted by flow cytometry. C, Tc17 cells express reduced levels of perforin and granzyme B. Levels of perforin and granzyme B were measured by intracellular staining. D, Perforin knockout Tc17 cells do not have CTL activity. Alloantigen-specific Tc1 or Tc17 effector cells were generated from WT or perforin knockout (Pfr1−/−) mice. In vitro killing activity was measured toward allogeneic ConA-activated splenocyte blasts for 16 hours at E:T = 3:1. Data are shown as the mean ± SD of a representative experiment (n = 3 samples per group). *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; n.s., not significant (unpaired t test).

Figure 2.

Tc17 cells depend on perforin to lyse target cells. A, Tc17 cells express low levels of FasL and TRAIL. Cell-surface expression of FasL and TRAIL were measured by flow cytometry. B, Blocking of FasL or TRAIL slightly enhance the killing of target cells by Tc1 cells. Tc1 and Tc17 cells were incubated with CSFE-labeled target MC38.OVA cells for 16 hours in the presence of isotype control, or blocking antibodies to FasL or TRAIL at E:T = 3:1. At the end of the experiment, remaining viable target cells were counted by flow cytometry. C, Tc17 cells express reduced levels of perforin and granzyme B. Levels of perforin and granzyme B were measured by intracellular staining. D, Perforin knockout Tc17 cells do not have CTL activity. Alloantigen-specific Tc1 or Tc17 effector cells were generated from WT or perforin knockout (Pfr1−/−) mice. In vitro killing activity was measured toward allogeneic ConA-activated splenocyte blasts for 16 hours at E:T = 3:1. Data are shown as the mean ± SD of a representative experiment (n = 3 samples per group). *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; n.s., not significant (unpaired t test).

Close modal

We then revisited the potential contributions of the perforin/granzyme pathway toward the Tc17 lytic activity. Compared with Tc1 cells, fewer perforin+ cells were found in Tc17 cells, and they expressed less perforin mRNA (Fig. 2C; Supplementary Fig. S1B). As reported in the literature (27, 28), we also observed a decreased percentage of granzyme B+ cells and decreased mRNA expression in Tc17 cells (Fig. 2C; Supplementary Fig. S1B). There are 11 different granzymes, and mice deficient in any single granzyme such as granzyme A, B, or M generally display mild cell killing deficiency, highlighting the functional redundancy within the family (39). In contrast, there is only one perforin, and perforin-deficient mice are severely immunodeficient with compromised ability to defend against viral infections and tumors (40). Thus, we utilized perforin-deficient mice to investigate whether the perforin/granzyme pathway is involved in the cytotoxicity of Tc17 cells. WT or perforin KO cells were differentiated into Tc1 or Tc17 cells and tested for their cytotoxicity toward allogeneic ConA- or LPS-activated splenocyte blasts (Fig. 2D; Supplementary Fig. S1C). Loss of perforin completely abolished the killing of Tc17 cells in vitro. These results demonstrated that although their perforin levels are low, this pathway is indispensable for the cytotoxicity of Tc17 cells. Dependence on the perforin/granzyme pathway for Tc17 CTL activity and the low expression of granzyme B suggest that other granzymes are important for Tc17 CTLs.

Tc17 express more granzyme A, and granzyme A is indispensable for Tc17 cytotoxicity

We next investigated which granzymes are expressed in Tc17 cells, and compared expression with Tc1 cells. The major differentially expressed granzyme mRNA in Tc17 compared with Tc1 cells is granzyme A (Fig. 3A), which was confirmed by assessing protein with flow cytometry (Supplementary Fig. S2A). Tc1 and Tc17 also express granzyme C, F, and G at comparable, albeit low levels (Fig. 3A).

Figure 3.

Tc17 cells express high levels of granzyme A, and downregulation of granzyme A decreases Tc17 cytotoxicity. A, The differential expression of granzymes in Tc1 and Tc17 cells. Expressions of various granzymes and perforin were measured by qPCR and normalized to housekeeping gene. The relative expression was calculated to granzyme A levels on Tc1 cells. B, Granzyme A siRNA knockdowns granzyme A expression in both Tc1 and Tc17 cells. Nucleofection with granzyme A or mock siRNA into day 3 in vitro differentiated Tc1 or Tc17 cells, and granzyme A levels were measured by intracellular staining 2 days later. C, Granzyme A siRNA knockdown decreases the cytotoxicity of Tc17 cells at various E:T ratios. In vitro killing activity was measured toward MC38.OVA target cells during a 16-hour CTL assay with various E:T ratios. Data are shown as the mean ± SD of a representative experiment (n = 3 samples per group). *, P < 0.05; **, P < 0.01; n.s., not significant (unpaired t test).

Figure 3.

Tc17 cells express high levels of granzyme A, and downregulation of granzyme A decreases Tc17 cytotoxicity. A, The differential expression of granzymes in Tc1 and Tc17 cells. Expressions of various granzymes and perforin were measured by qPCR and normalized to housekeeping gene. The relative expression was calculated to granzyme A levels on Tc1 cells. B, Granzyme A siRNA knockdowns granzyme A expression in both Tc1 and Tc17 cells. Nucleofection with granzyme A or mock siRNA into day 3 in vitro differentiated Tc1 or Tc17 cells, and granzyme A levels were measured by intracellular staining 2 days later. C, Granzyme A siRNA knockdown decreases the cytotoxicity of Tc17 cells at various E:T ratios. In vitro killing activity was measured toward MC38.OVA target cells during a 16-hour CTL assay with various E:T ratios. Data are shown as the mean ± SD of a representative experiment (n = 3 samples per group). *, P < 0.05; **, P < 0.01; n.s., not significant (unpaired t test).

Close modal

To examine whether granzyme A is involved for Tc17 cytotoxicity, we used granzyme A siRNA to inhibit its production. Nucleofection with granzyme A siRNA did not change the viability or cytokine production profile of Tc17 or Tc1 cells (Supplementary Fig. S2B). We observed reduction of granzyme A mRNA (Supplementary Fig. S2C) and protein (Fig. 3B) in Tc17 (88% and 60% reduction of mRNA and protein, respectively), as well as Tc1 cells (94% and 86% reduction of mRNA and protein, respectively); and no alterations for granzyme B or other granzymes after granzyme A siRNA knockdown (Supplementary Fig. S2D). We then measured the lytic activity of the Tc1 and Tc17 cells with granzyme A knockdown toward target cells (Fig. 3C). Knockdown of granzyme A in Tc17 cells reduced their cytotoxicity, and the effect is greater when a lower E:T ratio of Tc17 cells was used in the CTL assay (22%, 28%, 44%, and 100% reduction at decreasing E:T ratios). There is no difference for Tc1 cells treated with granzyme A siRNA (Supplementary Fig. S2E). These data support the importance of granzyme A for Tc17 cytotoxicity and suggest it is redundant for Tc1 cytotoxicity.

Tc17 have survival advantages and manifest sustained killing capacity

During the course of these experiments, we noted that the number of effector cells remaining at the end of a 16-hour CTL was routinely higher for Tc17 than for Tc1 cells. To determine if this was the result of the coculture with antigen-expressing target cells or intrinsic to the effector T cells, we restimulated Tc1 and Tc17 effector cells with anti-CD3 for 72 hours and found that Tc1 have significantly reduced viability (Fig. 4A). This is consistent with the literature describing a differential sensitivity to activation-induced cell death between Th1 and Th17 cells (41, 42).

Figure 4.

Tc17 cells survive better and maintain their cytotoxicity after antigen encounter. A, Tc17 cells are resistant to AICD. Tc1 and Tc17 cells were differentiated for 5 days, washed, and restimulated with anti-CD3 and IL2 for additional 3 days. Cell viabilities were measured by flow cytometry. B, Tc17 cells survive better and kill target cells equivalently to Tc1 cells over time. In vitro killing activity was measured toward MC38.OVA target cells during a 72-hour CTL assay with E:T = 3:1. Remaining target cells and effector cells at various time points were measured by flow cytometry. Tc17 cells maintain granzyme A expression. C, Effector Tc1 or Tc17 cells were analyzed at various time points for granzyme B or granzyme A expression by intracellular staining. D, Tc17 cells maintain cytotoxicity after the initial antigen encounter. After 48 hours of incubation with MC38.OVA target cells, viable effector cells were purified. An equal number of antigen-experienced effector cells were tested for their lytic activity with fresh MC38.OVA target cells in a 16-hour recall response at E:T=3:1. Fresh effector cells were used as controls. Remaining target cells and effector cells were measured by flow cytometry and E:T ratios were calculated. Data are shown as the mean ± SD of a representative experiment (n = 3–4 samples per group). *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; n.s., not significant (unpaired t test).

Figure 4.

Tc17 cells survive better and maintain their cytotoxicity after antigen encounter. A, Tc17 cells are resistant to AICD. Tc1 and Tc17 cells were differentiated for 5 days, washed, and restimulated with anti-CD3 and IL2 for additional 3 days. Cell viabilities were measured by flow cytometry. B, Tc17 cells survive better and kill target cells equivalently to Tc1 cells over time. In vitro killing activity was measured toward MC38.OVA target cells during a 72-hour CTL assay with E:T = 3:1. Remaining target cells and effector cells at various time points were measured by flow cytometry. Tc17 cells maintain granzyme A expression. C, Effector Tc1 or Tc17 cells were analyzed at various time points for granzyme B or granzyme A expression by intracellular staining. D, Tc17 cells maintain cytotoxicity after the initial antigen encounter. After 48 hours of incubation with MC38.OVA target cells, viable effector cells were purified. An equal number of antigen-experienced effector cells were tested for their lytic activity with fresh MC38.OVA target cells in a 16-hour recall response at E:T=3:1. Fresh effector cells were used as controls. Remaining target cells and effector cells were measured by flow cytometry and E:T ratios were calculated. Data are shown as the mean ± SD of a representative experiment (n = 3–4 samples per group). *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; n.s., not significant (unpaired t test).

Close modal

To assess how improved effector survival affects lytic activity, we measured kinetics of target cell killing during a prolonged, 72-hour CTL assay. As we observed before, Tc1 cells displayed higher CTL activity at early time points, as fewer target cells were left after 4 hours (40% less) or 24 hours (47% less) of culture with Tc1 cells compared with Tc17 cells (Fig. 4B, middle). However, a similar amount of target cells remained at 48 hours or 72 hours of culture (Fig. 4B, middle), indicating that an equivalent sum of total cytotoxicity was achieved with Tc1 and Tc17 cells over 72 hours. When we measured the number of effector cells during the prolonged CTL assay, we found Tc17 had improved viability over time compared with Tc1 cells (Fig. 4B, right). As a result, Tc17 cells maintained higher effector to target ratios at later time points (Supplementary Fig. S3A). These data demonstrate that improved effector survival of Tc17 cells compensates for their slow onset of lytic activity and contributes to the accumulated lysis of target cells over time and are consistent with the in vivo antitumor activity of Tc17 cells.

After synthesis and maturation, granzymes and perforin are packaged and stored within granules. Upon interacting with antigen-expressing target cells, CTLs release stored granzymes within minutes. To maintain their cytotoxicity in a target-rich environment, CTL cells need to replenish their intracellular pool for continuous granule exocytosis. Given the difference between Tc1 and Tc17 lytic activity over time, we examined the expression kinetics of the major granzymes for each subset. A larger fraction of Tc1 cells expressed granzyme B initially, but this decreased after 48 hours (Fig. 4C). Tc17 cells, on the other hand, maintained the same percentage of granzyme A+ cells for as long as we measured (Fig. 4C).

The prolonged cytotoxicity assay in Fig. 4B measured the cumulative effect of target killing over time. We next compared the ability of Tc1 and Tc17 CTLs to serially lyse target cells by determining whether Tc1 or Tc17 effector cells maintained their cytotoxicity after the initial antigen encounter. After 48-hour incubation with target cells, viable effector cells were purified and equal numbers of experienced CTLs or new effector cells were tested for their lytic activity with fresh target cells. Antigen-experienced Tc17 cells lysed target cells in the recall response at a rate comparable with the newly generated Tc17 effector cells, whereas Tc1 cells started to lose their cytotoxicity (20% less; Fig. 4D, middle). Tc17 effector cells had superior viability over Tc1 cells in both the initial antigen encounter (49% increase) and the recall response (309% increase; Supplementary Fig. S3B), and thus maintained a higher E:T ratio in both the initial antigen encounter (52% increase) and the recall response (445% increase; Fig. 4D, right), compared with Tc1 cells. These data indicate that Tc17 effectors have superior survival after antigen encounter and maintain their ability to lyse target cells in serial encounters, which supports their effectiveness in antitumor immune responses.

Synthetic RORγ agonists increase cytotoxicity of Tc17 cells in vitro and in vivo

We first confirmed our previous findings (18) that the RORγ agonist LYC-54143 enhances the direct tumor killing activity of Tc17 cells in vitro and improves the survival of these effectors after encountering antigen-specific target cells. As expected, RORγ agonist added during Tc17 differentiation increased the percentage of IL17+ and RORγ+ cells and decreased CD73+ cells. RORγ agonist treatment increased the lytic activity for Tc17 cells (192% increase; Fig. 5A) and had minimal impact on Tc1 cells (21% increase). Analysis of the effector cells remaining after a 16-hour CTL assay (Fig. 5B) revealed agonist treatment during differentiation further improved the survival of Tc17 effector cells and augments the advantage of Tc17 CTLs over Tc1 cells.

Figure 5.

RORγ agonists increase the cytotoxicity of Tc17 cells both in vitro and in vivo. A, Tc1 and Tc17 cells were differentiated in vitro in the presence or absence of an RORγ agonist for 5 days. Cells were washed and counted, and an equal number of effector cells were used to lyse E.G7-OVA target cells in vitro during a 16-hour CTL assay at E:T = 3:1. B, The agonist increased the survival of Tc17 cells. At the end of the CTL assay, remaining effector cells were measured by flow cytometry. C, The agonist increased antitumor activity of Tc17 cells in the ACT model. Tc17 cells were differentiated in vitro in the presence or absence of the RORγ agonist for 5 days. Cells were washed and counted. Tc17 cells (5 × 106) were used in the ACT experiment. Data are shown as the mean ± SD of a representative experiment (n = 3 or 4 samples per group for an in vitro experiment, n = 10 mice per group for the in vivo experiment). *, P < 0.05; **, P < 0.01; ***, P < 0.001; n.s., not significant (unpaired t test for A and B; multiple t tests for C).

Figure 5.

RORγ agonists increase the cytotoxicity of Tc17 cells both in vitro and in vivo. A, Tc1 and Tc17 cells were differentiated in vitro in the presence or absence of an RORγ agonist for 5 days. Cells were washed and counted, and an equal number of effector cells were used to lyse E.G7-OVA target cells in vitro during a 16-hour CTL assay at E:T = 3:1. B, The agonist increased the survival of Tc17 cells. At the end of the CTL assay, remaining effector cells were measured by flow cytometry. C, The agonist increased antitumor activity of Tc17 cells in the ACT model. Tc17 cells were differentiated in vitro in the presence or absence of the RORγ agonist for 5 days. Cells were washed and counted. Tc17 cells (5 × 106) were used in the ACT experiment. Data are shown as the mean ± SD of a representative experiment (n = 3 or 4 samples per group for an in vitro experiment, n = 10 mice per group for the in vivo experiment). *, P < 0.05; **, P < 0.01; ***, P < 0.001; n.s., not significant (unpaired t test for A and B; multiple t tests for C).

Close modal

We then investigated in perforin-deficient mice as to whether the perforin/granzyme pathway is involved in the efficacy of RORγ agonists on Tc17 cytotoxicity. RORγ agonists increased the cytotoxicity of WT Tc17 cells toward allogeneic ConA-activated splenocyte blasts as expected, but not in Prf1KO Tc17 cells (Supplementary Fig. S4), suggesting the efficacy of RORγ agonists on Tc17 cytotoxicity is dependent on the perforin/granzyme pathway.

The enhanced CTL activity observed after RORγ agonist treatment was assessed for relevance in an in vivo setting using adoptive transfer of equal numbers of Tc17 cells differentiated in vitro with or without LYC-54143 into E.G7-OVA tumor–bearing mice. We observed tumor shrinkage with Tc17 cells, and the agonist further increased their antitumor activity (Fig. 5C), confirming the physiologic relevance of our in vitro findings (Fig. 1D) and our previous in vivo data in a different adoptive transfer system (18).

Tc17 cells recruit Tc1 cells to the tumor microenvironment and improve their function

In vivo, it is likely that mixed populations of Tc1 and Tc17 effectors coexist in tumor microenvironments and other tissues. After adoptive transfer into tumor-bearing animals, Tc17 cells infiltrated tumors as early as 2 days after transfer, but Tc1 cells were not detected (Fig. 6A, left), suggesting that Tc17 cells are better able to traffic to tumors, whereas Tc1 cells may need to be recruited. We observed a concurrent increased percentage of host CD8+ T cells in the tumor-infiltrating lymphocyte (TIL) population the after transfer of Tc17 cells (Fig. 6A, right). This recruitment of host cells by transferred Tc17 cells likely continued over the course of the study, as the percentage of donor cells and host CD8+ T cells 21 days after transfer correlated well (r2 = 0.93; Fig. 6B), indicating Tc17 cells control the tumor growth not only through their direct cytotoxicity, but also through recruiting other effector cells to the tumor microenvironment. The inclusion of the RORγ agonist LYC-54143 during Tc17 differentiation resulted in the generation of Tc17 cells, which survive better in vivo and are more effective at recruiting host effector T cells to the tumor microenvironment (Fig. 6C).

Figure 6.

RORγ agonists enhance Tc17 cells, recruiting effector T cells to the tumor microenvironment and improving their function. A, Donor Tc17 cells migrate to the tumor and recruit host CD8 T cells. Two days after transfer into E.G7-OVA tumor–bearing mice, Thy1.1+ donor cells and Thy1.1 host CD8+ T cells in the tumor were analyzed by flow cytometry. n = 5 mice per group. B, Presence of donor cells correlates with host CD8 T cells in the TIL. Twenty-one days after transfer into E.G7-OVA tumor–bearing mice, Thy1.1+ donor cells and Thy1.1- host CD8+ T cells in the tumor were analyzed by flow cytometry. n = 17 mice per group. C, RORγ agonists increase the number of donor cells and recruit host CD8 T cells. Agonist-treated Tc17 were transferred as in Fig. 5C. The presence of donor and host CD8+ T cells in TIL were analyzed by flow cytometry. n = 10 mice per group. D, Tc17 cells improve Tc1 cytotoxicity in recall response. Tc1 cells were mixed with Tc17 cells during the initial 48 hours of incubation with MC38.OVA target cells. Viable Tc1 effector cells were purified, and an equal number of antigen-experienced effector cells were tested for their lytic activity with fresh MC38.OVA target cells in a 16-hour recall response at E:T=3:1. Fresh effector cells were used as controls. Remaining effector and target cells were measured by flow cytometry, and E:T ratios were calculated. Data are shown as the mean ± SD of a representative experiment (n = 3 samples per group for the in vitro experiment, n = 5–17 mice per group for the in vivo experiment). *, P < 0.05; **, P < 0.01; ****, P < 0.0001; n.s., not significant (unpaired t test).

Figure 6.

RORγ agonists enhance Tc17 cells, recruiting effector T cells to the tumor microenvironment and improving their function. A, Donor Tc17 cells migrate to the tumor and recruit host CD8 T cells. Two days after transfer into E.G7-OVA tumor–bearing mice, Thy1.1+ donor cells and Thy1.1 host CD8+ T cells in the tumor were analyzed by flow cytometry. n = 5 mice per group. B, Presence of donor cells correlates with host CD8 T cells in the TIL. Twenty-one days after transfer into E.G7-OVA tumor–bearing mice, Thy1.1+ donor cells and Thy1.1- host CD8+ T cells in the tumor were analyzed by flow cytometry. n = 17 mice per group. C, RORγ agonists increase the number of donor cells and recruit host CD8 T cells. Agonist-treated Tc17 were transferred as in Fig. 5C. The presence of donor and host CD8+ T cells in TIL were analyzed by flow cytometry. n = 10 mice per group. D, Tc17 cells improve Tc1 cytotoxicity in recall response. Tc1 cells were mixed with Tc17 cells during the initial 48 hours of incubation with MC38.OVA target cells. Viable Tc1 effector cells were purified, and an equal number of antigen-experienced effector cells were tested for their lytic activity with fresh MC38.OVA target cells in a 16-hour recall response at E:T=3:1. Fresh effector cells were used as controls. Remaining effector and target cells were measured by flow cytometry, and E:T ratios were calculated. Data are shown as the mean ± SD of a representative experiment (n = 3 samples per group for the in vitro experiment, n = 5–17 mice per group for the in vivo experiment). *, P < 0.05; **, P < 0.01; ****, P < 0.0001; n.s., not significant (unpaired t test).

Close modal

Tc1 and Tc17 cells express chemokine receptors such as CCR5 or CCR6, which guide these cells to regions of high CCL3/4/5, CXCL9/10/11, or CCL20, respectively (6). Tc17 cells also express chemokines such as CCL3/4/5 and CXCL9/10 themselves, and blocking antibodies to individual chemokine partially decreased their chemotactic activity (6). In our hands, Tc17 cells expressed more chemokine receptors CCR6, CCR7, and CXCR3 compared with Tc1 cells, whereas Tc1 cells expressed more CCR5 (Supplementary Fig. S5A), consistent with literature findings. We also found Tc17 cells expressed high levels of CCL3/4/5, and low levels of CCL20/22, CXCL2/3/9/10. And the levels of CXCL9 and CXCL10 are significantly higher compared with Tc1 cells. RORγ agonists increased the expression of CCR6 and CCL3 in Tc17 cells (Supplementary Fig. S5B), which might contribute to the better migration of Tc17 cells into the tumor mass and recruitment of Tc1 cells.

To investigate whether the persistent Tc17 cells with their sustained lytic activity can benefit the surrounding Tc1 cells directly, we returned to our in vitro system and mixed Tc17 cells with Tc1 cells during the initial target encounter, and then purified Tc1 cells. Tc1 cocultured with Tc17 during the initial target encounter survived better, which improved the E:T ratio of Tc1 cells during the secondary challenge (Fig. 6D). These results suggest that Tc17 cells provide a transferrable factor(s) that enhances the serial killing ability of Tc1 cells.

Type 17 cells, including both Th17 and Tc17, can eradicate large, established murine tumors after adoptive transfer and provide robust protection against repeated tumor challenges (6–8). Despite these observations, little is known about the mechanisms by which Tc17 cells mediate antitumor immunity (43–51). Herein, we demonstrate that Tc17 cells can lyse target cells directly, utilizing four different types of target cells: E.G7-OVA, MC38.OVA or allogeneic ConA- or LPS-activated splenocyte blasts.

Among the nine granzymes we examined, Tc1 expressed more granzyme B, whereas Tc17 expressed more granzyme A. Knockdown of granzyme A specifically decreased the lytic activity of Tc17 cells, but not Tc1 cells, emphasizing the importance of granzyme A to Tc17 cell cytotoxicity. Tc17 cells also expressed granzyme B, C, F, and G at low levels, which might contribute to the residual cytotoxicity in granzyme A knockdown Tc17 cells.

The kinetics and degree to which the individual granzymes and perforin are expressed vary in different T-cell clones in vitro and in vivo, and depend on how they are activated (39). Granzyme B is expressed earlier than granzyme A during initial T-cell activation (52), but the percentage of granzyme B+ cells decreases after 3 days (53). Granzyme A and B induce independent cell death pathways, and cells that overexpress Bcl-2 family antiapoptotic proteins, caspase, or granzyme B protease inhibitors are still sensitive to granzyme A (54). FasL/Fas and granzyme B are involved in AICD of Tc1 and Th1 cells. The low levels of granzyme B, in addition to low FasL and high c-FLIP (41, 42), in Tc17 cells might contribute to their AICD resistance. Consistent with this, granzyme B–deficient mice clear both allogeneic and syngeneic tumor cell lines more efficiently than do WT mice (55). Extracellular granzyme A and granzyme K can also directly release proinflammatory cytokines from monocytes, macrophages, and fibroblasts (56). Thus, both Tc1 and Tc17 cells are CTLs with antitumor activities. Tc1 cells may be better suited for acute release of massive amounts of cytotoxic molecules to control initial tumor expansion with the caveat of collateral damage to nearby T cells and nontarget tissue, whereas Tc17 are better for more measured release of granular contents in a sustained way, and involve more effector cells for the antitumor immunity (57–59).

Once inside the tumor, Tc17 cells can induce chemokine production from tissue-resident cells (60,61) and recruit monocytes, neutrophils, NK cells, and CD8+ T cells into the tumor microenvironment (24, 25). Using RORγt KO mice, Nunez and colleagues found Th17 cells promoted the recruitment of Th1 cells to the tumor and contributed to antitumor immunity (62). Therefore, it is likely chemokines expressed directly by Tc17 cells and indirectly induced through IL17A/F cytokines, paired with chemokine receptors expressed on Tc1 cells, contribute to the recruitment of Tc1 cells. We observed a concurrent increase of host CD8 T cells in TILs 2 days after transfer of Tc17 cells, and the recruitment of host cells was sustained over time. In addition, our data showed that Tc17 could benefit the surrounding Tc1 cells directly, as the in vitro mixture of Tc1 and Tc17 cells during the initial target encounter improved the survival of Tc1 cells during the secondary challenge. Whether direct cell–cell interaction was absolutely needed or which soluble factor(s) mediated this transferrable beneficial effect is currently under investigation.

In summary, we have demonstrated that: (i) Tc17 can be CTLs, and the conditions under which they were differentiated and evaluated can influence their cytotoxicity, (ii) Tc17 CTLs depend on perforin and granzyme A for their lytic activity, (iii) Tc17 CTLs refill/maintain expression of key cytotoxic molecules and survive better, which allows them to kill more targets over time despite lower expression of cytotoxic molecules, (iv) Tc17 cells recruit Tc1 cells and can improve their ability to serially kill target cells. Our characterization of Tc17 cells adds to our understanding of how these cells participate in antitumor immune responses and provides a rationale for the discovery and development of synthetic RORγ agonist drugs which further potentiate Tc17 CTL activity, persistence, and antitumor efficacy. Currently, our RORγ agonist LYC-55716 is being tested in the clinic for locally advanced or metastatic cancer in a phase I/IIa trial (NCT03396497), and in combination with pembrolizumab for non–small cell lung cancer in a phase Ib trial (NCT02929862, www.clincialtrials.gov). Therapeutic strategies that enhance both Tc1 and Tc17 cells may improve the efficacy observed with current immunotherapies.

W. Zou has ownership interest (including stock, patents, etc.) in Lycera Corp. and is a consultant/advisory board member for the same. X. Hu is Executive Director of Lycera Corp. and has ownership interest (including stock, patents, etc.) in the same. L.L. Carter has ownership interest (including stock, patents, etc.) in Lycera Corp. No potential conflicts of interest were disclosed by the other authors.

Conception and design: X. Liu, W. Zou, X. Hu, L.L. Carter

Development of methodology: X. Liu, E.M. Zawidzka, H. Li

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): X. Liu, E.M. Zawidzka, C.A. Lesch, J. Dunbar, D. Bousley

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): X. Liu, E.M. Zawidzka, H. Li, L.L. Carter

Writing, review, and/or revision of the manuscript: X. Liu, E.M. Zawidzka, X. Hu, L.L. Carter

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): X. Liu

Study supervision: X. Liu, X. Hu, L.L. Carter

We thank Peter L. Toogood, Bruce A. Goldsmith, H. Jeffrey Wilkins, and other ECL members for critically reading the manuscript and providing constructive suggestions for revision. We also thank Timothy J. Ley from Washington University and Xuefang Cao from the University of Maryland for the suggestions on the granzyme-deficient mice.

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.
Ivanov
II
,
McKenzie
BS
,
Zhou
L
,
Tadokoro
CE
,
Lepelley
A
,
Lafaille
JJ
, et al
The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells
.
Cell
2006
;
126
:
1121
33
.
2.
Zou
W
,
Restifo
NP
. 
T(H)17 cells in tumour immunity and immunotherapy
.
Nat Rev Immunol
2010
;
10
:
248
56
.
3.
Patel
DD
,
Kuchroo
VK
. 
Th17 cell pathway in human immunity: lessons from genetics and therapeutic interventions
.
Immunity
2015
;
43
:
1040
51
.
4.
Fragoulis
GE
,
Siebert
S
,
McInnes
IB
. 
Therapeutic targeting of IL-17 and IL-23 cytokines in immune-mediated diseases
.
Annu Rev Med
2016
;
67
:
337
53
.
5.
Frieder
J
,
Kivelevitch
D
,
Haugh
I
,
Watson
I
,
Menter
A
. 
Anti-IL-23 and anti-IL-17 biologic agents for the treatment of immune-mediated inflammatory conditions
.
Clin Pharmacol Ther
2018
;
103
:
88
101
.
6.
Garcia-Hernandez Mde
L
,
Hamada
H
,
Reome
JB
,
Misra
SK
,
Tighe
MP
,
Dutton
RW
. 
Adoptive transfer of tumor-specific Tc17 effector T cells controls the growth of B16 melanoma in mice
.
J Immunol
2010
;
184
:
4215
27
.
7.
Muranski
P
,
Borman
ZA
,
Kerkar
SP
,
Klebanoff
CA
,
Ji
Y
,
Sanchez-Perez
L
, et al
Th17 cells are long lived and retain a stem cell-like molecular signature
.
Immunity
2011
;
35
:
972
85
.
8.
Hinrichs
CS
,
Kaiser
A
,
Paulos
CM
,
Cassard
L
,
Sanchez-Perez
L
,
Heemskerk
B
, et al
Type 17 CD8+ T cells display enhanced antitumor immunity
.
Blood
2009
;
114
:
596
9
.
9.
Chen
WC
,
Lai
YH
,
Chen
HY
,
Guo
HR
,
Su
IJ
,
Chen
HH
. 
Interleukin-17-producing cell infiltration in the breast cancer tumour microenvironment is a poor prognostic factor
.
Histopathology
2013
;
63
:
225
33
.
10.
Qian
X
,
Chen
H
,
Wu
X
,
Hu
L
,
Huang
Q
,
Jin
Y
. 
Interleukin-17 acts as double-edged sword in anti-tumor immunity and tumorigenesis
.
Cytokine
2017
;
89
:
34
44
.
11.
Benchetrit
F
,
Ciree
A
,
Vives
V
,
Warnier
G
,
Gey
A
,
Sautes-Fridman
C
, et al
Interleukin-17 inhibits tumor cell growth by means of a T-cell-dependent mechanism
.
Blood
2002
;
99
:
2114
21
.
12.
Derhovanessian
E
,
Adams
V
,
Hahnel
K
,
Groeger
A
,
Pandha
H
,
Ward
S
, et al
Pretreatment frequency of circulating IL-17+ CD4+ T-cells, but not Tregs, correlates with clinical response to whole-cell vaccination in prostate cancer patients
.
Int J Cancer
2009
;
125
:
1372
9
.
13.
Horlock
C
,
Stott
B
,
Dyson
PJ
,
Morishita
M
,
Coombes
RC
,
Savage
P
, et al
The effects of trastuzumab on the CD4+CD25+FoxP3+ and CD4+IL17A+ T-cell axis in patients with breast cancer
.
Br J Cancer
2009
;
100
:
1061
7
.
14.
Kryczek
I
,
Wei
S
,
Szeliga
W
,
Vatan
L
,
Zou
W
. 
Endogenous IL-17 contributes to reduced tumor growth and metastasis
.
Blood
2009
;
114
:
357
9
.
15.
Martin-Orozco
N
,
Muranski
P
,
Chung
Y
,
Yang
XO
,
Yamazaki
T
,
Lu
S
, et al
T helper 17 cells promote cytotoxic T cell activation in tumor immunity
.
Immunity
2009
;
31
:
787
98
.
16.
Sfanos
KS
,
Bruno
TC
,
Maris
CH
,
Xu
L
,
Thoburn
CJ
,
DeMarzo
AM
, et al
Phenotypic analysis of prostate-infiltrating lymphocytes reveals TH17 and Treg skewing
.
Clin Cancer Res
2008
;
14
:
3254
61
.
17.
Yang
ZZ
,
Novak
AJ
,
Ziesmer
SC
,
Witzig
TE
,
Ansell
SM
. 
Malignant B cells skew the balance of regulatory T cells and TH17 cells in B-cell non-Hodgkin's lymphoma
.
Cancer Res
2009
;
69
:
5522
30
.
18.
Hu
X
,
Liu
X
,
Moisan
J
,
Wang
Y
,
Lesch
CA
,
Spooner
C
, et al
Synthetic RORgamma agonists regulate multiple pathways to enhance antitumor immunity
.
Oncoimmunology
2016
;
5
:
e1254854
.
19.
Kryczek
I
,
Banerjee
M
,
Cheng
P
,
Vatan
L
,
Szeliga
W
,
Wei
S
, et al
Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments
.
Blood
2009
;
114
:
1141
9
.
20.
Curtis
MM
,
Way
SS
,
Wilson
CB
. 
IL-23 promotes the production of IL-17 by antigen-specific CD8 T cells in the absence of IL-12 and type-I interferons
.
J Immunol
2009
;
183
:
381
7
.
21.
El-Behi
M
,
Dai
H
,
Magalhaes
JG
,
Hwang
D
,
Zhang
GX
,
Rostami
A
, et al
Committed Tc17 cells are phenotypically and functionally resistant to the effects of IL-27
.
Eur J Immunol
2014
;
44
:
3003
14
.
22.
Flores-Santibanez
F
,
Cuadra
B
,
Fernandez
D
,
Rosemblatt
MV
,
Nunez
S
,
Cruz
P
, et al
In vitro-generated Tc17 cells present a memory phenotype and serve as a reservoir of Tc1 cells in vivo
.
Front Immunol
2018
;
9
:
209
.
23.
Gaffen
SL
. 
Structure and signalling in the IL-17 receptor family
.
Nat Rev Immunol
2009
;
9
:
556
67
.
24.
Codarri
L
,
Gyulveszi
G
,
Tosevski
V
,
Hesske
L
,
Fontana
A
,
Magnenat
L
, et al
RORgammat drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation
.
Nat Immunol
2011
;
12
:
560
7
.
25.
Dong
C
. 
Differentiation and function of pro-inflammatory Th17 cells
.
Microbes Infect
2009
;
11
:
584
8
.
26.
Hu
X
,
Majchrzak
K
,
Liu
X
,
Wyatt
MM
,
Spooner
CJ
,
Moisan
J
, et al
In vitro priming of adoptively transferred T cells with a RORgamma agonist confers durable memory and stemness in vivo
.
Cancer Res
2018
;
78
:
3888
98
.
27.
Yeh
N
,
Glosson
NL
,
Wang
N
,
Guindon
L
,
McKinley
C
,
Hamada
H
, et al
Tc17 cells are capable of mediating immunity to vaccinia virus by acquisition of a cytotoxic phenotype
.
J Immunol
2010
;
185
:
2089
98
.
28.
Tajima
M
,
Wakita
D
,
Satoh
T
,
Kitamura
H
,
Nishimura
T
. 
IL-17/IFN-gamma double producing CD8+ T (Tc17/IFN-gamma) cells: a novel cytotoxic T-cell subset converted from Tc17 cells by IL-12
.
Int Immunol
2011
;
23
:
751
9
.
29.
Yen
HR
,
Harris
TJ
,
Wada
S
,
Grosso
JF
,
Getnet
D
,
Goldberg
MV
, et al
Tc17 CD8 T cells: functional plasticity and subset diversity
.
J Immunol
2009
;
183
:
7161
8
.
30.
Hamada
H
,
Bassity
E
,
Flies
A
,
Strutt
TM
,
Garcia-Hernandez Mde
L
,
McKinstry
KK
, et al
Multiple redundant effector mechanisms of CD8+ T cells protect against influenza infection
.
J Immunol
2013
;
190
:
296
306
.
31.
Sharma
MD
,
Hou
DY
,
Liu
Y
,
Koni
PA
,
Metz
R
,
Chandler
P
, et al
Indoleamine 2,3-dioxygenase controls conversion of Foxp3+ Tregs to TH17-like cells in tumor-draining lymph nodes
.
Blood
2009
;
113
:
6102
11
.
32.
Noto
A
,
Ngauv
P
,
Trautmann
L
. 
Cell-based flow cytometry assay to measure cytotoxic activity
.
J Vis Exp
2013
;
e51105
.
33.
Wonderlich
J
,
Shearer
G
,
Livingstone
A
,
Brooks
A
,
Soloski
MJ
,
Presby
MM
. 
Induction and measurement of cytotoxic T lymphocyte activity
.
Curr Protoc Immunol
2018
;
120
:
3 11 11–13 11 29
.
34.
Kojima
H
,
Shinohara
N
,
Hanaoka
S
,
Someya-Shirota
Y
,
Takagaki
Y
,
Ohno
H
, et al
Two distinct pathways of specific killing revealed by perforin mutant cytotoxic T lymphocytes
.
Immunity
1994
;
1
:
357
64
.
35.
Shresta
S
,
Pham
CT
,
Thomas
DA
,
Graubert
TA
,
Ley
TJ
. 
How do cytotoxic lymphocytes kill their targets?
Curr Opin Immunol
1998
;
10
:
581
7
.
36.
Chatterjee
S
,
Thyagarajan
K
,
Kesarwani
P
,
Song
JH
,
Soloshchenko
M
,
Fu
J
, et al
Reducing CD73 expression by IL1beta-Programmed Th17 cells improves immunotherapeutic control of tumors
.
Cancer Res
2014
;
74
:
6048
59
.
37.
He
JS
,
Ostergaard
HL
. 
CTLs contain and use intracellular stores of FasL distinct from cytolytic granules
.
J Immunol
2007
;
179
:
2339
48
.
38.
Lettau
M
,
Paulsen
M
,
Schmidt
H
,
Janssen
O
. 
Insights into the molecular regulation of FasL (CD178) biology
.
Eur J Cell Biol
2011
;
90
:
456
66
.
39.
Anthony
DA
,
Andrews
DM
,
Watt
SV
,
Trapani
JA
,
Smyth
MJ
. 
Functional dissection of the granzyme family: cell death and inflammation
.
Immunol Rev
2010
;
235
:
73
92
.
40.
Voskoboinik
I
,
Sutton
VR
,
Ciccone
A
,
House
CM
,
Chia
J
,
Darcy
PK
, et al
Perforin activity and immune homeostasis: the common A91V polymorphism in perforin results in both presynaptic and postsynaptic defects in function
.
Blood
2007
;
110
:
1184
90
.
41.
Yu
Y
,
Iclozan
C
,
Yamazaki
T
,
Yang
X
,
Anasetti
C
,
Dong
C
, et al
Abundant c-Fas-associated death domain-like interleukin-1-converting enzyme inhibitory protein expression determines resistance of T helper 17 cells to activation-induced cell death
.
Blood
2009
;
114
:
1026
8
.
42.
Shi
G
,
Ramaswamy
M
,
Vistica
BP
,
Cox
CA
,
Tan
C
,
Wawrousek
EF
, et al
Unlike Th1, Th17 cells mediate sustained autoimmune inflammation and are highly resistant to restimulation-induced cell death
.
J Immunol
2009
;
183
:
7547
56
.
43.
Chang
MR
,
Dharmarajan
V
,
Doebelin
C
,
Garcia-Ordonez
RD
,
Novick
SJ
,
Kuruvilla
DS
, et al
Synthetic RORgammat Agonists Enhance Protective Immunity
.
ACS Chem Biol
2016
;
11
:
1012
8
.
44.
Veldhoen
M
,
Hocking
RJ
,
Atkins
CJ
,
Locksley
RM
,
Stockinger
B
. 
TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells
.
Immunity
2006
;
24
:
179
89
.
45.
Bettelli
E
,
Carrier
Y
,
Gao
W
,
Korn
T
,
Strom
TB
,
Oukka
M
, et al
Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells
.
Nature
2006
;
441
:
235
8
.
46.
Mangan
PR
,
Harrington
LE
,
O'Quinn
DB
,
Helms
WS
,
Bullard
DC
,
Elson
CO
, et al
Transforming growth factor-beta induces development of the T(H)17 lineage
.
Nature
2006
;
441
:
231
4
.
47.
Nelson
MH
,
Kundimi
S
,
Bowers
JS
,
Rogers
CE
,
Huff
LW
,
Schwartz
KM
, et al
The inducible costimulator augments Tc17 cell responses to self and tumor tissue
.
J Immunol
2015
;
194
:
1737
47
.
48.
Gajewski
TF
,
Schreiber
H
,
Fu
YX
. 
Innate and adaptive immune cells in the tumor microenvironment
.
Nat Immunol
2013
;
14
:
1014
22
.
49.
Joyce
JA
,
Fearon
DT
. 
T cell exclusion, immune privilege, and the tumor microenvironment
.
Science
2015
;
348
:
74
80
.
50.
Spranger
S
,
Gajewski
TF
. 
Tumor-intrinsic oncogene pathways mediating immune avoidance
.
Oncoimmunology
2016
;
5
:
e1086862
.
51.
Majchrzak
K
,
Nelson
MH
,
Bailey
SR
,
Bowers
JS
,
Yu
XZ
,
Rubinstein
MP
, et al
Exploiting IL-17-producing CD4+ and CD8+ T cells to improve cancer immunotherapy in the clinic
.
Cancer Immunol Immunother
2016
;
65
:
247
59
.
52.
Kelso
A
,
Costelloe
EO
,
Johnson
BJ
,
Groves
P
,
Buttigieg
K
,
Fitzpatrick
DR
. 
The genes for perforin, granzymes A-C and IFN-gamma are differentially expressed in single CD8(+) T cells during primary activation
.
Int Immunol
2002
;
14
:
605
13
.
53.
Cai
SF
,
Fehniger
TA
,
Cao
X
,
Mayer
JC
,
Brune
JD
,
French
AR
, et al
Differential expression of granzyme B and C in murine cytotoxic lymphocytes
.
J Immunol
2009
;
182
:
6287
97
.
54.
Pardo
J
,
Bosque
A
,
Brehm
R
,
Wallich
R
,
Naval
J
,
Mullbacher
A
, et al
Apoptotic pathways are selectively activated by granzyme A and/or granzyme B in CTL-mediated target cell lysis
.
J Cell Biol
2004
;
167
:
457
68
.
55.
Cao
X
,
Cai
SF
,
Fehniger
TA
,
Song
J
,
Collins
LI
,
Piwnica-Worms
DR
, et al
Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance
.
Immunity
2007
;
27
:
635
46
.
56.
van Eck
JA
,
Shan
L
,
Meeldijk
J
,
Hack
CE
,
Bovenschen
N
. 
A novel proinflammatory role for granzyme A
.
Cell Death Dis
2017
;
8
:
e2630
.
57.
Isaaz
S
,
Baetz
K
,
Olsen
K
,
Podack
E
,
Griffiths
GM
. 
Serial killing by cytotoxic T lymphocytes: T cell receptor triggers degranulation, re-filling of the lytic granules and secretion of lytic proteins via a non-granule pathway
.
Eur J Immunol
1995
;
25
:
1071
9
.
58.
Kryczek
I
,
Zhao
E
,
Liu
Y
,
Wang
Y
,
Vatan
L
,
Szeliga
W
, et al
Human TH17 cells are long-lived effector memory cells
.
Sci Transl Med
2011
;
3
:
104ra100
.
59.
McGeachy
MJ
. 
Th17 memory cells: live long and proliferate
.
J Leukoc Biol
2013
;
94
:
921
6
.
60.
Comerford
I
,
Bunting
M
,
Fenix
K
,
Haylock-Jacobs
S
,
Litchfield
W
,
Harata-Lee
Y
, et al
An immune paradox: how can the same chemokine axis regulate both immune tolerance and activation?: CCR6/CCL20: a chemokine axis balancing immunological tolerance and inflammation in autoimmune disease
.
Bioessays
2010
;
32
:
1067
76
.
61.
Bromley
SK
,
Mempel
TR
,
Luster
AD
. 
Orchestrating the orchestrators: chemokines in control of T cell traffic
.
Nat Immunol
2008
;
9
:
970
80
.
62.
Nunez
S
,
Saez
JJ
,
Fernandez
D
,
Flores-Santibanez
F
,
Alvarez
K
,
Tejon
G
, et al
T helper type 17 cells contribute to anti-tumour immunity and promote the recruitment of T helper type 1 cells to the tumour
.
Immunology
2013
;
139
:
61
71
.