Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2024 Jan 5.
Published in final edited form as: Sci Immunol. 2023 Dec 15;8(90):eabo5558. doi: 10.1126/sciimmunol.abo5558

Acquisition of suppressive function by conventional T cells limits anti-tumor immunity upon Treg depletion

Sarah K Whiteside 1,, Francis M Grant 2, Giorgia Alvisi 3, James Clarke 4, Leqi Tang 1, Charlotte J Imianowski 1, Baojie Zhang 1, Alexander C Evans 1, Alexander J Wesolowski 1, Alberto G Conti 1, Jie Yang 1, Sarah N Lauder 5, Mathew Clement 5, Ian R Humphreys 5, James Dooley 1, Oliver Burton 1, Adrian Liston 1, Marco Alloisio 6,7, Emanuele Voulaz 6,7, Jean Langhorne 8, Klaus Okkenhaug 1, Enrico Lugli 3, Rahul Roychoudhuri 1,
PMCID: PMC7615475  EMSID: EMS192862  PMID: 38100544

Abstract

Regulatory T (Treg) cells contribute to immune homeostasis but suppress immune responses to cancer. Strategies to disrupt Treg cell-mediated cancer immunosuppression have been met with limited clinical success, but the underlying mechanisms for this failure are poorly understood. By modeling Treg cell-targeted immunotherapy in mice, we find that CD4+ Foxp3 conventional T (Tconv) cells acquire suppressive function upon depletion of Foxp3+ Treg cells, limiting therapeutic efficacy. Foxp3 Tconv cells within tumors adopt a Treg cell-like transcriptional profile upon ablation of Treg cells and have a similar capacity to suppress T cell activation and proliferation ex vivo. Suppressive activity is enriched among CD4+ Tconv cells marked by expression of C-C motif receptor 8 (CCR8), which are found in mouse and human tumors. Upon Treg cell depletion, CCR8+ Tconv cells undergo systemic and intratumoral activation and expansion, and mediate IL-10 dependent suppression of anti-tumor immunity. Consequently, conditional deletion of Il10 within T cells augments anti-tumor immunity upon Treg cell-depletion in mice, and antibody blockade of IL-10 signaling synergizes with Treg cell depletion to overcome treatment resistance. These findings reveal a secondary layer of immunosuppression by Tconv cells released upon therapeutic Treg cell depletion and suggest that broader consideration of suppressive function within the T cell lineage is required for development of effective Treg cell-targeted therapies.

Introduction

Immune checkpoint blockade therapies targeting the inhibitory receptors PD-1 and CTLA-4 on Tconv cells have revolutionized the treatment of advanced cancer (15). However, only a minority of patients with a subset of cancers respond to existing therapies (68), necessitating development of mechanistically distinct modes of immunotherapy. Treg cells play a critical role in suppressing endogenous and immunotherapy-driven anti-tumor immunity (914). High relative ratios of Treg cells to CD4+ or CD8+ Tconv cells within tumors are associated with poor prognoses in patients with a variety of cancers, including ovarian cancer (15, 16), breast cancer (17), non-small cell lung carcinoma (18), hepatocellular carcinoma (19), renal cell carcinoma (20), pancreatic cancer (21), gastric cancer (22), cervical cancer (23), intrahepatic cholangiocarcinoma (24) and colorectal carcinoma (25). Foxp3+ Treg cells also powerfully contribute to immunotherapy resistance, including to immune checkpoint inhibitor therapy (18, 2628). There is intense medical interest in therapeutically depleting Treg cells or modulating their immunosuppressive function in cancer patients.

Despite abundant experimental evidence of the immunosuppressive role of Treg cells in cancer, Treg cell-targeted therapies have had limited success in the clinic. Agents developed for depletion of Treg cells in humans have included daclizumab (Zenapax), a monoclonal antibody against CD25 which is expressed highly on the surface of most Treg cells, denikeukin difitox (Ontak), an IL-2:diphtheria toxin fusion protein that targets Treg cells through their ability to bind IL-2, and mogamulizumab, a depleting monoclonal antibody against CCR4, which is expressed by high frequencies of tumor-infiltrating Treg cells (29). Daclizumab therapy failed to enhance the efficacy of a dendritic cell vaccine in metastatic melanoma patients (30), and only modestly increased immune response parameters in patients with glioblastoma (31) and breast cancer (32), while denikeukin difitox treatment failed to induce clinical responses in metastatic melanoma patients (33). Mogamulizumab therapy lacked anti-tumor efficacy in advanced cancer patients (34), likely attributable to concomitant depletion of activated CD4+ and CD8+ Tconv cells expressing CCR4 (35). Lack of clinical efficacy despite depletion of Treg cells in many cases indicates a need to discern the basis for treatment failure of Treg cell-targeted therapies.

In this study, we sought to better understand mechanisms of treatment failure of Treg cell-targeting cancer immunotherapies. We systematically evaluated the consequence of experimental Treg cell ablation on Tconv cells within tumors. Whereas CD4+ and CD8+ Tconv cells were markedly transcriptionally distinct from Treg cells under steady-state conditions, Treg cell ablation caused Tconv cells to adopt a Treg cell-like transcriptional profile, upregulating expression of molecules associated with Treg cell suppressive function. Consistent with acquisition of a Treg cell-like transcriptional profile, Foxp3 Tconv cells from Treg cell-depleted animals acquired a potent ability to suppress Tconv activation and proliferation in vitro, attributable to the expansion and activation of a subset of Tconv cells marked by expression of CCR8. This subset of suppressive Tconv cells was found to be enriched in both murine and human tumors, and its suppressive function was dependent upon IL-10. Consequently, conditional deletion of Il10 specifically within T cells, and blockade of IL-10 receptor (IL-10R) signaling during Treg cell depleting immunotherapy reversed treatment failure and resulted in enhanced tumor clearance. These findings indicate that compensatory suppression by Tconv cells limits efficacy of Treg cell-targeted therapeutic depletion and suggests that broader consideration of suppressive activity within the T cell lineage will be required for development of more effective therapies.

Results

Treg cell ablation causes Tconv cells to adopt a Treg cell-like transcriptional profile

Treg cell depletion has had limited success in cancer patients with advanced disease. To better understand mechanisms underlying treatment failure in the context of therapeutic Treg cell ablation, we utilized Foxp3EGFP-DTR mice, which express human diphtheria toxin receptor (DTR) and enhanced green fluorescent protein (EGFP) under the transcriptional control of the endogenous Foxp3 gene. Administration of diphtheria toxin (DTx) to Foxp3EGFP-DTR mice enables selective depletion of Foxp3+ Treg cells (36). We subcutaneously implanted syngeneic B16-F10 melanoma cells into Foxp3EGFP-DTR mice and ablated Treg cells through administration of DTx. Early Treg cell ablation (before tumors were palpable) resulted in incomplete rejection of primary tumors, whereas Treg cell depletion in mice with established tumors had little discernible effect upon tumor growth (Fig. 1A), despite near complete ablation of Foxp3-expressing Treg cells within the systemic and intratumoral compartments of DTx-treated mice (Fig. 1B).

Figure 1. Treg cell ablation causes Tconv cells to acquire transcriptional features of Treg cells.

Figure 1

(A) Tumor growth of B16-F10 tumors subcutaneously implanted into Foxp3EGFP-DTR mice. Gray shading indicates time period over which PBS or DTx was administered (day 7-13 post-implantation, early disease; day 10-16 post-implantation, established). Dashed lines indicate individual mice. Solid line indicates average tumor area over time. Data representative of 4 individually repeated experiments, n > 5 **P < 0.01; two-tailed Mann–Whitney U-test. (B) Representative frequency of Foxp3+ Treg cells among total CD4+ T cells within spleens (top) or tumors (bottom) of Foxp3EGFP-DTR mice with established tumors administered PBS or DTx. (C) Heatmap showing the relative expression of transcripts upregulated in intratumoral Treg cells compared with CD4+ Tconv cells (q<0.05; FC>4) in the indicated T cell subsets isolated at day 18 after implantation of B16-F10 tumors in Foxp3EGFP-DTR animals administered PBS or DTx. Colors indicate expression normalized to row maxima. x-axis hierarchical clustering of intratumoral Treg cell-expressed transcripts identifies 5 clusters of genes with distinct expression patterns. Gray bars to right of heatmap indicate expression greater than a third of the expression of given transcripts in intratumoral Treg cells. (D) Average expression of genes within the 5 clusters identified in each T cell subset. (E) Heatmap showing pairwise Pearson distances between the global gene expression profiles of the indicated T cell subsets from B16-F10 tumor-bearing Foxp3EGFP-DTR animals administered PBS or DTx. (F) Scatterplot comparing the global differences in gene expression between intratumoral Treg cells and Tconv cells with transcriptional differences between CD4+ Tconv cells isolated from DTx versus PBS-treated animals. A highly significant correlation is observed indicating transcriptional convergence of intratumoral Treg cells with Tconv cells in absence of Treg cells. Data from 2-4 biological replicates isolated on independent days (C-F).

To understand mechanisms of treatment resistance, we first examined the consequence of Treg cell ablation on the transcriptional profiles of CD4+ and CD8+ Tconv cells within tumors. Surprisingly, although intratumoral Foxp3EGFP− CD4+ and CD8+ Tconv cells were markedly transcriptionally distinct from Foxp3EGFP+ Treg cells under steady-state conditions, Treg cell depletion caused Foxp3EGFP− Tconv cells to adopt a Treg cell-like transcriptional profile. We noted that a large proportion of genes specifically enriched within tumor-associated Treg cells compared with CD4+ Tconv cells (|FC| > 4, q < 0.05) under steady-state conditions were induced at high levels within CD4+ or CD8+ Tconv cells upon Treg cell ablation (Fig. 1C and 1D; Data file S1). Clusters A and B comprised intratumoral Treg cell-associated genes upregulated in both CD4+ and CD8+ Tconv cells upon Treg cell ablation; Cluster C contained intratumoral Treg cell-associated genes whose expression was upregulated exclusively in CD8+ Tconv cells; Cluster D included intratumoral Treg cell-associated genes whose expression was upregulated exclusively in CD4+ Tconv cells; Cluster E comprised a limited set of Treg cell-specific transcripts that were not expressed at high relative levels in CD4+ or CD8+ Tconv cells, including Foxp3, Lrrc32, Ikzf2, Runx2, and Ctla4. Similarly, a substantial fraction of transcripts highly expressed within intratumoral Tconv cells compared with Treg cells were downregulated within Tconv cells upon Treg cell ablation. (Fig. S1; Data file S2). Consistent with these observations, hierarchical clustering analysis of Pearson distances between global transcriptional profiles of samples revealed that intratumoral CD4+ and CD8+ Tconv cells from Treg cell-depleted animals clustered more strongly with Treg cells than Tconv cells from Treg cell-sufficient animals (Fig. 1E). Moreover, differences in gene expression between CD4+ Tconv cells in the absence versus presence of Treg cells were significantly positively correlated with differences in gene expression between intratumoral Treg cells and CD4+ Tconv cells from Treg cell-sufficient animals (Fig. 1F). Collectively, these results show that intratumoral Tconv cells adopt a Treg cell-like transcriptional profile upon experimental ablation of Treg cells in vivo.

Ablation of Treg cells promotes the induction of Tconv cell suppression

Treg cells suppress the proliferation of naïve Tconv cells when co-cultured in vitro (3739). Given their acquisition of a Treg cell-like transcriptional profile, we asked whether Tconv cells develop suppressive function upon depletion of Treg cells. To test this, we purified Foxp3EGFP− CD4+ Tconv cells or Foxp3EGFP+ CD4+ Treg cells by FACS from B16-F10-tumor bearing Foxp3EGFP-DTR mice and incubated them with congenically distinct naïve CD4+ Tconv cells in vitro (Fig. 2A, Fig. S2). Strikingly, CD4+ Tconv cells from the tumors of mice whose Treg cells had been ablated by administration of DTx profoundly suppressed the proliferation of naïve CD4+ T cell responders compared with CD4+ Tconv cells from tumors of animals with intact Treg cell populations (Fig. 2B and C). The level of suppression was only marginally less than the level of suppressive activity of a similar number of intratumoral Foxp3+ Treg cells. We observed lower levels of suppressive activity among splenic Tconv cells (Fig. S3A-B), suggesting that suppressive function was enriched in the tumor. In addition to suppressing T cell proliferation, Tconv cells from tumors of Treg cell-depleted animals suppressed stimulation-driven induction of the activation marker CD44 on responder cells in contrast to Tconv cells from tumors of non-Treg cell-depleted animals (Fig. 2D and E). Tconv cells from Treg cell-depleted animals expressed similar levels of the co-inhibitory molecules TIGIT, TIM-3 and GITR to intratumoral Treg cells (Fig. 2F and G). They also expressed higher levels of CTLA-4 and ICOS compared to CD4+ Tconv cells from tumors with intact Treg cell populations. Acquisition of suppressive function by Tconv cells was not an artefact of DTx treatment, since administration of DTx to Foxp3EGFP-DTR and control Foxp3EGFP mice resulted in induction of potent suppressive activity only among CD4+ Tconv cells from Foxp3EGFP-DTR animals, whose Treg cells are sensitive to DTx treatment (Fig. S3C-D). Thus, upon Treg cell ablation, CD4+ Tconv cells within tumors acquire transcriptional and functional characteristics of Treg cells.

Figure 2. Treg cell ablation promotes induction of suppressive function by CD4+ Tconv cells.

Figure 2

(A) Experimental schema. B16-F10 cells were subcutaneously implanted into Foxp3EGFP-DTR CD45.2+ mice and administered PBS or DTx on day 7, 9, 11, and 13. Cells were harvested from tumors of PBS- and DTx-treated animals at day 16 post-implantation and used in suppression assays. (B and C) In vitro suppression assay. Proliferation of naïve CD45.1+ CD4+ Tconv responder (Tresp) cells 4 days cultured at a 4:1 ratio with indicated suppressor cell populations (Foxp3EGFP− Tconv cells from tumors of Treg-replete (PBS) or Treg-depleted (DTx) mice, or GFP+ Treg cells from tumors of Treg-replete mice). Representative histograms and replicate measurements of proliferation dye dilution at day 4 post-stimulation gated on CD45.1+ Tresp cells shown. Tresp cell proliferation in the absence of a suppressor cell population was used as a control. Suppressor cells were co-cultured with responder T (Tresp) cells at a ratio of 1:4, with 2.5×104 suppressor CD4+ Treg or Tconv cells co-cultured with 1×105 Tresp cells in the presence of 5.0×104 antigen-presenting cells (APC). Data are representative of > 4 independently repeated experiments, n > 5 per group; ordinary one-way ANOVA, Tukey’s multiple comparisons. (D and E) Representative histograms and replicate measurements of CD44 expression by CD45.1+ Tresp cells incubated with indicated suppressor cell populations. (F-G) Representative flow cytometry and replicate measurements of the expression of the indicated proteins by intratumoral Treg cells and CD4+ Tconv cells isolated at day 16 after implantation of B16-F10 tumors in Foxp3EGFP-DTR mice treated with PBS or DTx. Data are representative of > 3 independently repeated experiments, n > 7 per group. ****P <0.0001; ns, not significant; one-way ANOVA Kruskal-Wallis, Dunn’s multiple comparisons test. Error bars show standard error of the mean (s.e.m.)

Treg cell depletion results in activation and expansion of CCR8+ Tconv cells

To understand whether changes in the transcriptional and functional properties of bulk populations of Tconv cells in the absence of Treg cells were driven by specific subpopulations, we performed scRNA-Seq of bulk T cell populations sorted by FACS from B16-F10 melanoma tumors of DTx- and PBS-treated Foxp3EGFP-DTR animals at day 16 after tumor implantation. Single-cell gene expression data were clustered using Seurat and global transcriptional differences between cells were visualized in two-dimensional space using Uniform Manifold Approximation and Projection (UMAP). k-means clustering revealed the presence of 8 transcriptionally distinct clusters of cells (Fig. 3A). Clusters 2 and 3 were enriched in control samples, whereas Clusters 0, 1, 5 and 7 were enriched among T cells from tumors of Treg cell-depleted animals (Fig. 3B and C). Enrichment analysis was used to determine which cell cluster was most responsible for the induction of Treg cell-like gene expression within bulk RNA-Seq profiles of Tconv cells from Treg cell-depleted animals. This revealed that Cluster 0 (present at a ~3:1 ratio in the DTx treatment condition) was most enriched in genes specifically upregulated by CD4+ Tconv cells upon DTx treatment (Fig. 3D). To define surface markers which would enable isolation of cells of Cluster 0, we performed an analysis of uniquely upregulated transcripts within each cluster. This analysis revealed that Cluster 0 cells express transcripts associated with T cell activation but which are also highly expressed by Treg cells, including Il2ra, Tigit and Tnfrsf4 (Fig. 3E and Data file S3). Strikingly, Ccr8 mRNA expression was also upregulated in Cluster 0 cells upon depletion of Treg cells, which was notable since we and others have shown that chemokine (C-C motif) receptor 8 (CCR8) marks highly suppressive Treg cells under steady-state conditions within both murine and human tumors (4044). A focused analysis of the CD4+ T cells in Cluster 0 revealed a sub-population of cells (subcluster 5) enriched in expression of transcripts encoding proteins associated with Th2 differentiation, including Ccr8, Gata3, Maf and Il10, and suppressive/co-inhibitory function, including Pdcd1, Tigit, Il10 and Lag3 (Fig. S4, Data file S4). Accordingly, an analysis of the distribution of cells expressing Ccr8, Il2ra, Tigit and Tnfrsf4 revealed that while in Treg cell-replete animals, these markers are largely expressed by intratumoral of Foxp3EGFP+ Treg cells, they became expressed by a subset of Foxp3EGFP− CD4+ Tconv cells upon Treg cell depletion (Fig. 3F and Fig. S5).

Figure 3. Treg cell ablation promotes the expansion of tumor-infiltrating CCR8+ Tconv cells.

Figure 3

(A) Uniform manifold approximation and projection (UMAP) of scRNA-Seq of TCRβ+ cells isolated at day 16 after implantation of B16-F10 tumors in Foxp3EGFP-DTR mice treated with PBS or DTx on days 7, 9, 11 and 13. (B) Density plots showing change in distribution of cells within tumors of Treg-replete (PBS) and Treg-depleted (DTx) animals. (C) Relative frequency of cells within each cluster normalized to their average ratio among PBS animals. N = 3 biological replicates per group. (D) Average enrichment of expression of the genes in Cluster D from Fig. 1C across scRNA-Seq clusters, (n=3, unpaired two-tailed Student’s t-test, *P < 0.05, **P < 0.01). (E) Heatmap showing the expression of differentially upregulated genes in each cluster identified. (F) UMAP plots showing expression of indicated genes within T cells of tumors from tumor-bearing PBS- or DTx-treated Foxp3EGFP-DTR animals. (G) Representative flow cytometry and (H) replicate measurements of total counts (top) and the frequency (bottom) of CCR8+ of CD4+ Tconv cells from spleens, draining lymph nodes (dLN) and tumors of B16-F10 tumor-bearing Foxp3EGFP-DTR mice treated with PBS or DTx. Data are representative of 3 independently repeated experiments (G and H). Numbers in gates show percentages. n > 10. one-way ANOVA Kruskal-Wallis, Dunn’s multiple comparisons test. **P <0.01, ***P < 0.001, ****P < 0.0001; Error bars show standard error of the mean (s.e.m.)

We therefore analyzed the expression of CCR8 on the surface of Treg and Tconv cells in tumors and lymphatics of Foxp3EGFP-DTR animals treated with PBS or DTx (Fig. 3G and H). We found that Treg cell depletion increased the absolute number of CCR8+ Tconv cells within all tissues analyzed, including tumors, draining lymph nodes (dLN) and spleen, whereas the relative frequency of CCR8+ cells among total CD4+ Foxp3EGFP− Tconv cells was increased within the spleens of Treg cell-depleted animals, but not within dLN and tumors due to the absolute expansion of other CD4+ Foxp3EGFP− Tconv cells subsets in these tissues upon Treg cell depletion.

CCR8 expression marks highly suppressive Tconv cells within tumors

To better understand the identity of CCR8+ Tconv cells, we purified CCR8+ and CCR8 CD4+ Tconv cells by FACS from B16-F10 tumors of DTX-treated Foxp3EGFP-DTR mice and subjected them to bulk RNA-Seq. CCR8+ Tconv cells were enriched in transcripts encoding molecules associated with both T cell activation such as Tnfrsf9 (encoding 4-1BB) and Treg cell suppressive function, including Il2ra, Areg, and Il10 (Fig. 4A and Data file S5). Interestingly, CCR8+ Tconv cells were not enriched in transcripts associated with suppressive type 1 regulatory T cells (Tr1) such as Eomes, Gzmk, Itga2 (encoding CD49b), Ccr5, or Cd226, suggesting that they are distinct from Tr1 cells. Gene set enrichment analysis (GSEA) of global gene expression differences between CCR8+ and CCR8 Tconv cells revealed a negative enrichment of genes upregulated in Foxp3 Tconv cells vs Foxp3+ Treg cells among CCR8+ Tconv cells compared with CCR8 Tconv cells (Fig. 4B). Consistently, we observed that global differences in gene expression between CCR8+ and CCR8 Tconv cells were positively correlated with global differences in gene expression between intratumoral Treg and Tconv cells (Fig. 4C), further suggesting that CCR8+ Foxp3 Tconv cells possess a Treg cell-like transcriptional profile.

Figure 4. CCR8 marks highly suppressive Tconv cells.

Figure 4

(A) Heatmap showing the relative expression of differentially expressed genes between intratumoral CCR8+ and CCR8 CD4+ Tconv cells (q<0.05; |FC|>3) isolated at day 16 after subcutaneous implantation of B16-F10 tumors of Treg-depleted Foxp3EGFP-DTR animals treated with DTx at days 7, 9, 11 and 13. Data from 4 biological replicates isolated on the same day. (B) Gene-set enrichment analysis (GSEA) demonstrating a negative enrichment of genes upregulated in Foxp3 Tconv cells vs Foxp3+ Treg cells among CCR8+ Tconv cells compared with CCR8 Tconv cells isolated from tumors of DTx-treated Foxp3EGFP-DTR mice. (C) Scatterplot comparing global changes in gene expression between intratumoral Treg and Tconv cells with transcriptional differences between CCR8+ and CCR8 CD4+ Tconv cells. (D) Representative flow cytometry and (E) replicate measurements of the expression of the indicated proteins from intratumoral Treg cells and CCR8+ and CCR8 CD4+ Tconv cells from tumors of B16-F10 tumor-bearing Foxp3EGFP-DTR mice treated with PBS or DTx. Data are representative of 3 independently repeated experiments n > 4 one-way ANOVA Kruskal-Wallis, Dunn’s multiple comparisons test. *P < 0.05, **P <0.01, ***P < 0.001, ****P < 0.0001, ns, not significant. (F) Representative histograms and (G) replicate measurements of Tresp cells (naïve CD45.1+ CD4+ Tconv cells) incubated with intratumoral CD45.2+ TCRβ+ CD4+ GFP CCR8 or CD45.2+ TCRβ+ CD4+ GFP CCR8+ suppressor Tconv cells isolated at day 16 after implantation of B16-F10 tumors in Foxp3EGFP-DTR mice. Suppressor cells were incubated with responders at a ratio of 1:8, with 1.25×104 suppressor CD4+ Tconv cells co-cultured with 1×105 Tresp cells in the presence of 5×104 APC. Cell proliferation of Tresp cells was analyzed after 4 days. Data are representative of 2 independently repeated experiments n > 3. ordinary one-way ANOVA, Tukey’s multiple comparisons. *P < 0.05; ***P < 0.001, ****P < 0.0001; Error bars show standard error of the mean (s.e.m.)

We compared the phenotype of CCR8 and CCR8+ CD4+ Tconv cells from tumors of mice whose Treg cells had been depleted by DTx with that of Treg cells. Like Treg cells, we found that CCR8+ CD4+ Tconv cells expressed high levels of CD25, OX40, GITR, TIGIT and LAG-3 compared to CCR8 CD4+ Tconv cells (Fig. 4D and E). CCR8+ Tconv cells also expressed increased levels of the transcription factor GATA3, suggesting that they possess a Th2-like differentiation state. To test whether the accumulation of CCR8+ cells within intratumoral Tconv cell populations following Treg cell ablation accounts for their increased suppressive activity, we separately sorted CCR8+ and CCR8 Foxp3EGFP− Tconv cells from the tumors of DTx-treated Foxp3EGFP-DTR mice and assessed their ability to suppress naïve Tconv cell proliferation in vitro. Notably, suppressive function was enriched within the CCR8+ Tconv cell fraction, which were more capable of restricting proliferation of responder cells compared to the CCR8 Tconv cell fraction (Fig. 4F and G). Taken together, these results suggest that CCR8 expression marks a subset of highly activated and suppressive Tconv cells which accumulate systemically and within tumors upon Treg cell depletion.

CD4+ FOXP3 CCR8+ Tconv cells are found within tumors of NSCLC patients

In order to determine if CCR8+ FOXP3 Tconv cells are enriched in human tumors, we analyzed CD4+ T cells from 48 patents with non-small cell lung carcinoma (NSCLC) by flow cytometry (Fig. S6). Similar to our observations in mouse, CCR8+ FOXP3 Tconv cells expressed high levels of CD25 and were significantly enriched in tumor tissue compared to healthy adjacent tissue and blood from the same patients (Fig. 5A and 5B). Moreover, the frequency of CCR8+ CD25+ FOXP3 Tconv cells was inversely correlated with the frequency of cytotoxic CD8+ T cells within tumors (Fig. 5C). They also co-expressed the inhibitory receptors PD-1, TIGIT and TIM-3 (Fig. 5D and E). CD4+ FOXP3 CCR8+ T cells displayed increased expression of the tissue-residency marker CXCR6 and co-stimulatory receptor CD27 compared to CCR8 cells and their phenotype was largely overlapping with that of FOXP3+ Treg cells (Fig. 5E and F). CD4+ FOXP3 CCR8+ T cells lacked expression of EOMES and Granzyme K, providing further evidence that this cell type is distinct from Tr1 cells (Fig. 5E). The presence of CCR8+ FOXP3 Tconv cells in human tumors under steady-state conditions and without Treg cell depletion is consistent with our observations within murine tumors (Fig. 3H and I), which contain a population of CCR8+ CD4+ Tconv cells under basal conditions that undergo substantial expansion upon Treg cell ablation.

Figure 5. CCR8+ Tconv cells expressing high levels of CD25 are found within the tumors of NSCLC patients.

Figure 5

(A) CCR8 and CD25 expression among FOXP3 CD4+ T cells from representative samples from NSCLC patients (n=48). (B) Frequency of FOXP3 CCR8+ CD25bright cells among CD4+ T cells from the indicated patients’ samples. Lines indicate paired samples. (C) Correlation of the frequency of CD8+ T cells (of CD3+ T cells) with FOXP3 CCR8+ CD25bright cells (of CD4+ T cells) in tumors from patient samples. (D) UMAP analysis of concatenated CD4+ FOXP3 Tconv cells. Colors depict cell clusters identified by Phenograph (k=500). Separate UMAP plots of relative marker expression by concatenated CD4+ T cells from tumors. (E) Representative frequency and (F) replicate measurements of indicated markers from patient samples, box plots show median and interquartile range (IQR). Bars indicate standard deviation. Dots depict values of a single tumor sample. **P < 0.01, ***P < 0.001, ****P < 0.0001; two-tailed Mann–Whitney U-test.

IL-10-dependent immune suppression by Tconv cells limits efficacy of Treg cell depletion

We sought to understand how intratumoral CD4+ Tconv cells exert their suppressive function. Informed by the results of our transcriptional analyses, we screened for the involvement of candidate suppressive mechanisms by which Tconv cells from Treg cell-depleted animals suppress T cell activation and proliferation in vitro. We tested whether blocking antibodies directed against CD25, CTLA-4, IL-10 receptor (IL-10R) and CCR8, neutralizing antibodies specific for transforming growth factor (TGF)-β, or pharmacological inhibition of steroid biosynthesis preferentially produced by Th2 cells using aminoglutethimide (AG) (45), are able to reverse the suppressive activity of intratumoral Tconv cells from Treg cell-depleted animals. Importantly, while the proliferation of naïve CD4+ T cells was suppressed by Foxp3 Tconv cells, these differences induced by the presence of suppressive Foxp3 Tconv cells were abolished upon treatment of cells with anti-IL-10R (Fig. 6A and B).

Figure 6. IL-10 production by CD4+ Tconv cells limits anti-tumor immunity upon Treg cell depletion.

Figure 6

(A) Screen to identify mechanisms of suppression by CD4+ Tconv cells from tumors of Treg-depleted animals. Proliferation of CTV-labeled naïve splenic CD4+ Tconv responder cells (Tresp) cultured alone or at a ratio of 8:1 with CD4+ Foxp3EGFP− Tconv suppressor cells (Tsupp) isolated at day 16 from B16-F10 tumors of DTx-treated Foxp3EGFP-DTR animals. Cells were cultured alone (gray) or with suppressors (purple) and with indicated reagents. AG, aminoglutethimide. CD45.2+ GFP Tconv suppressor cells were co-cultured with 1.25×104 suppressor CD4+ Tconv cells in the presence of 5.0×104 antigen-presenting cells (APC). Data are representative of 2 independently repeated experiments n > 3. P values show significance of difference between no suppressor and suppressor (Student t test) and are Bonferroni corrected. (B) Representative frequency of dividing Tresp cells incubated with tumor CD4+ GFP Tconv cells in the presence of anti-IL-10R antibodies or vehicle. Tresp cells without tumor Tconv cells were used as a control. (C) Co-correlation between the expression of indicated genes within single cell gene expression profiles of T cells from tumors of PBS- or DTx-treated B16 tumor-bearing Foxp3EGFP-DTR animals. Pearson correlation co-efficient values are indicated by color scale and genes are hierarchically clustered to identify clusters of co-expressed transcripts within T cell populations. scRNA-Seq data are representative of 3 biological replicates per group. (D) Measurement of Ccr8 and Il10 mRNA expression within CCR8 and CCR8+ Tconv cells from PBS- and DTx-treated animals. Data representative of 3-4 biological replicates per group. ordinary one-way ANOVA, Tukey’s multiple comparisons. ns, not significant. (E) Tumor area of heterotopic B16-F10 melanoma tumors at indicated time-points following implantation into Il10flox/flox Cd4Cre Foxp3EGFP-DTR or Il10+/+ Cd4Cre Foxp3EGFP-DTR control mice administered DTx or PBS from day 10-16 post-implantation. Data are representative of 2 independently repeated experiments, n > 7. ordinary one-way ANOVA, Tukey’s multiple comparisons. (F) Representative histograms (top) and replicate measurements (bottom) of the frequency of CD8+ CD44+ T cells and Foxp3 CD4+ CD44+ Tconv cells from tumors of animals within indicated treatment groups. (G) Representative frequency and (H) replicate measurements of the frequency (top) and total counts (bottom) of CD8+ IFN-γ+ TNF+ T cells within tumors. Data are representative of 2 independently repeated experiments, n > 4, one-way ANOVA, Kruskal-Wallis, Dunn’s multiple comparisons test. *P < 0.05; **P < 0.01; ***P < 0.001, ****P < 0.0001. Error bars show standard error of the mean (s.e.m.)

Given these observations, we asked whether CCR8+ Tconv cells are primary producers of Il10 mRNA upon depletion of Treg cells. We first examined whether the expression of Il10 mRNA is co-correlated with the expression of Ccr8 mRNA, and therefore co-expressed within the same cells, using scRNA-Seq of T cells from tumors of Treg cell-replete and - depleted animals. Since IL-10 is known to be produced by CD4+ TR1 cells which express LAG3 and CD49b (4649), Eomes+ CD4+ Tconv cells (5052), exhausted CD8+ T cells which express PD-1 (53) and CD4+ Th2 cells which express GATA3, IL-4 and IL-13 (54, 55), we included the genes encoding these and other markers in our co-correlation analysis. We found that under steady-state (Treg cell-replete) conditions, Il10 formed a predominant co-correlation cluster with Cd8a, Pdcd1, Ifng, Tnf, and Eomes but also a smaller cluster containing Gata3 and Foxp3, suggesting that CD8+ T cells in differential states of exhaustion, and Th2-like Treg cells are a predominant source of IL-10 (Fig. 6C). However, we found that Treg cell depletion resulted in a striking change in the co-correlation relationship of Il10 mRNA with the other genes examined, forming a predominant cluster of co-correlated genes containing Cd4, Ccr8, Il13, Il14 and Gata3. These results suggested that upon Treg cell ablation, the source of Il10 shifts to the previously identified CD4+ CCR8+ Tconv cell subset with Th2-like characteristics, and that CD4+ CCR8+ Tconv cells undergo activation into Il10-expressing cells. To confirm this, we sorted CCR8 and CCR8+ Tconv cells from tumors of Treg cell-replete and -depleted animals and subjected them to qRT-PCR. We found that Il10 mRNA expression was enriched among CCR8+ Tconv cells from Treg cell-depleted animals compared with both CCR8 cells from Treg cell-depleted animals and CCR8+ or CCR8 cells from Treg cell-replete animals (Fig. 6D). These findings supported the hypothesis that CCR8+ Tconv cells become the predominant source of T cell-expressed IL-10 upon Treg cell depletion.

We therefore asked whether Treg cell depletion triggers induction of IL-10-dependent suppressive activity among Foxp3 Tconv cells, limiting efficacy of Treg cell depletion in vivo. We had observed that Treg cell depletion was ineffective at significantly reducing growth of established B16 tumors, while Treg cell depletion during early disease delayed tumor growth but was ineffective at inducing complete responses (Fig. 1A). To test whether IL-10 production by Tconv cells is responsible for resistance to Treg cell-depleting therapy in vivo, we generated Il10flox/flox Cd4Cre Foxp3EGFP-DTR and littermate Cd4Cre Foxp3EGFP-DTR (IL-10 proficient) control mice. This allowed us to examine the effect of Treg cell depletion in animals whose remaining T cells can or cannot produce IL-10. Since IL-10 ablation has been shown to promote tumor growth under steady-state conditions (56, 57), we subcutaneously implanted with B16-F10 cells into Il10flox/flox Cd4Cre Foxp3EGFP-DTR and littermate Cd4Cre Foxp3EGFP-DTR control animals and selected animals with tumors of similar size (range 12 - 64 mm2) at day 10 after tumor implantation for randomization to treatment groups (PBS or DTx). We found that conditional deletion of IL-10 within T cells resulted in loss of resistance to Treg cell depletion, as indicated by reduced tumor growth when Treg cells were ablated in animals lacking T cell-restricted IL-10 expression, but not when either condition was present alone (Fig. 6E). There was also an increase in expression of the activation marker CD44 on CD4+ Tconv cells and CD8+ T cells from animals bearing a conditional deletion of Il10 and whose Treg cells had been ablated (Fig. 6F). Treg cell ablation in animals bearing a conditional deletion of Il10 within T cells was associated with increased frequencies of CD8+ T cells and CD4+ Tconv cells expressing the cytokines IFN-γ and TNF (Fig. 6G and H). These results demonstrate a critical role for T cell-produced IL-10 in resistance to Treg cell depletion.

Blockade of IL-10 signaling synergizes with Treg cell depletion to induce robust anti-tumor immune responses

We next asked whether antibody blockade of IL-10 signaling synergizes with Treg cell depletion in vivo. We tested whether blockade of IL-10R using anti-IL-10R antibodies reverses resistance to Treg cell-depleting therapy. We found that late Treg cell depletion or IL-10R blockade alone failed to drive significant reduction in tumor growth, whereas their combination resulted in potent tumor regression (Fig. 7A). Moreover, IL-10 blockade synergized with early Treg cell ablation to induce complete responses in a proportion of animals receiving combined therapy (Fig. S7). We found that IL-10R blockade both alone and in combination with Treg cell depletion increased the ratio of CD8+ T cells expressing IFN-γ and TNF within tumors, but the absolute number of IFN-γ- and TNF-expressing CD8+ T cells was markedly increased upon combined Treg cell ablation and IL-10R blockade, reflecting a combination of increased T cell infiltration and cytokine production (Fig. 7B and C). Similarly, the combination of Treg cell depletion and IL-10R blockade resulted in an increase in the frequency and absolute number of IFN-γ- and TNF-expressing CD4+ T cells (Fig. 7D and E). These findings demonstrate that Tconv cells within tumors adopt IL-10 dependent suppressive activity upon therapeutic elimination of Treg cells. This contributes to treatment failure of Treg cell depleting immunotherapies and that combined targeting of Treg cells and compensatory IL-10-dependent suppression invoked upon their depletion may enhance therapy.

Figure 7. Blockade of IL-10 signaling synergizes with Treg cell-depletion to drive potent anti-tumor immune responses.

Figure 7

(A) Tumor area of B16-F10 melanoma tumors at indicated time-points following implantation into Foxp3EGFP-DTR animals administered indicated combinations of DTx and anti-IL-10R or control reagents from days 10-16 after tumor implantation. Data are representative of 2 independently repeated experiments. n > 10, ordinary one-way ANOVA, Tukey’s multiple comparisons. *P < 0.05 **P < 0.01; (B) Representative frequency and (C) replicate measurements of the frequency and total counts of CD8+ IFN-γ+ TNF+ T cells from tumors. n > 3, one-way ANOVA, Kruskal-Wallis, Dunn’s multiple comparisons test., *P < 0.05. (D) Representative frequency and (E) replicate measurements of the frequency and total counts of Foxp3 CD4+ IFN-γ+ TNF+ T cells from tumors. Data from > 2 independent biological replicates. n > 3, Kruskal-Wallis, Dunn’s multiple comparisons test., *P < 0.05; Error bars show standard error of the mean (s.e.m.)

Discussion

Tconv cells and Treg cells share components of their activation programs to meet similar metabolic, proliferative and migratory requirements as they transition from quiescent states to activated states (5860). A substantial effort is now underway to develop therapies that specifically target molecules that distinguish Treg cells within tumors from their Tconv cell counterparts. These efforts have been informed by comparative analyses of the molecular profiles of Treg cells and Tconv cells within tumors under steady-state conditions (61, 62). We compared the transcriptional profiles of Treg cells with Tconv cells not only under steady-state conditions, but upon immune activation provoked by experimental Treg cell ablation. This analysis revealed that Tconv cells adopt a highly similar transcriptional profile to Treg cells upon Treg cell depletion. The extent of this reprograming goes beyond what would be expected as a result of the shared properties of Treg cell and Tconv cell core lymphocyte activation programs and reveals that Tconv cells take on compensatory IL-10-dependent suppressive function when Treg cells are eliminated. Acquisition of a Treg cell-like transcriptional profile by Tconv cells upon Treg cell ablation suggests that in practice there are very few molecules whose targeting will enable highly specific depletion of Treg cells within tumors. Nevertheless, a small cluster of genes was identified in our analyses which has an expression profile limited to Treg cells compared with Tconv cells under both steady-state conditions and upon Treg cell depletion. While this cluster of genes may contain targets for specific depletion of Treg cells within tumors, a question raised by this study is whether specific depletion of Treg cells is indeed desirable, rather than the targeting of molecules shared by Treg cells and cells with compensatory suppressive function induced upon Treg cell depletion.

It is known that CCR8 marks highly suppressive Treg cells found in both mouse and human tumors (4044, 63). There is significant interest in the development of therapies which deplete CCR8+ Treg cells within tumors. Given our observation that CCR8 also marks Tconv cells whose suppressive function is induced upon experimental Treg cell ablation in vivo, it is reasonable to postulate that depletion of CCR8-expressing cells is a superior approach to Treg cell depletion using other Treg cell-expressed markers for induction of anti-tumor immunity, since CCR8-depleting therapies would target both Treg cells and suppressive Tconv cells for destruction. It will be important to consider effects of CCR8-depleting therapies on both Treg cells and CCR8+ Tconv cells within tumors, both in pre-clinical investigations (42, 63), and if robust intratumoral depletion of CCR8+ cells in the human clinical context is achieved. Alternatively, our data suggests that combining Treg cell-targeted immunotherapies with blockade of IL-10 signaling will overcome compensatory suppression by Tconv cells.

It is interesting that the CCR8+ Tconv cell subset observed in human tumors did not phenotypically overlap with previously described Tr1 cells within tumors. Suppressive Tr1 cells have been characterized in several human tumors including head neck and squamous cell carcinoma (HNSCC) (64), colorectal cancer (52, 65), hepatocellular carcinoma (49), Hodgkin lymphoma (66), metastatic melanoma (67) and non-small-cell lung cancer (52), and their presence is often associated with tumor progression. The suppressive subset of CCR8+ Tconv cells we observe does not express EOMES or granzyme K, markers previously reported to be indicative of Tr1 cells. Recent studies have described a contribution of tumor-infiltrating follicular helper T (TFH) cells and follicular regulatory T (TFR) cells to anti-tumor immunity (68, 69). However, CXCR5 was not expressed by CCR8+ Tconv cells suggesting their distinction from TFH cells. We did however observe high levels of GATA3 and Il10 expression among CCR8+ Tconv cells, suggesting that they represent a Th2-like subset expressing high levels of markers associated with T cell activation, including CD25, expanded systemically and within tumors upon Treg cell depletion. Prior work are consistent with these data, showing systemic expansion of Th2 cells expressing either GATA3 or Th2 cytokines upon experimental Treg cell ablation (70, 71), and the intratumoral presence of CD4+ Tconv cells expressing CCR8 (4044, 63). We found that the frequency of CCR8+ CD25+ FOXP3 Tconv cells inversely correlates with the frequency of tumor-infiltrating CD8+ T cells, suggesting that they play an inhibitory role in human tumor immunity. The observation that CCR8+ Tconv cells expand and undergo activation to express IL-10 following depletion of Treg cells provides an explanation of how bulk Tconv cells from tumors of Treg cell-depleted animals acquire IL-10-dependent suppressive function despite lack of a change in the relative frequency of CCR8+ cells within the tumor CD4+ Tconv compartment. While our pre-clinical data from mouse models suggests that CCR8+ Tconv cells expand numerically within tumors upon experimental Treg cell ablation, this is difficult to formally assess in humans due to the lack of specific markers that are differentially expressed in comparison with FOXP3+ Treg cells, enabling their isolation ex vivo, and of clinically approved Treg cell-depleting therapies in widespread use. However, a number of Treg cell-targeted therapies are under development and it will be an important topic of future investigation to determine their effect upon CCR8+ Tconv cells in humans. It will also be important to better define how CCR8+ Tconv cells contribute to immune regulation in other contexts including infection and inflammation, as clones of Foxp3- CD4+ T cells expressing CCR8, CD25 and IL-10 have previously been described in mice with experimental pulmonary granulomata (72), while CD4+ Tconv cells expressing CCR8 are observed upon experimental allergic lung inflammation in mice (73), and infiltrating human skin (74).

Our findings show that in the context of Treg cell depleting immunotherapies, IL-10 production by Tconv cells represents a secondary layer of immune suppression responsible for driving immunotherapy resistance. It is important that this immunosuppressive function of IL-10 in limiting the efficacy of Treg cell targeted therapies is appreciated, since in other contexts, IL-10 has been shown to possess immunostimulatory activity both in pre-clinical models and in clinical trials (7577). Our findings suggest that IL-10 blocking antibodies may be used as a synergistic therapy with Treg cell-targeted immunotherapies to improve patient outcomes.

Preclinical studies suggest that Treg cell depletion can reinvigorate Tconv cell responses, however clinical trials of Treg cell depleting therapies have thus far been met with limited clinical efficacy (35, 78). While our analysis of human tumor-infiltrating Foxp3 Tconv cells revealed a fraction of CCR8+ Tconv cells under steady-state conditions, it will be valuable to examine whether such cells are expanded upon immunotherapy with either novel Treg cell-depleting therapies, or anti-CTLA-4 therapy, the therapeutic efficacy of which is postulated to in part depend upon depletion or blockade of the suppressive function of Treg cells (79, 80), and indeed in the context of non-Treg cell-targeted immunotherapy approaches. The prognostic significance of such cells in determining the outcome of the immunotherapy responses will reveal insights into their broader contribution to immunotherapy resistance.

Materials and Methods

Study design

The objective of this study was to understand how Treg cell depletion affects the function and immunoregulatory capacity of the T cell lineage in the context of tumor immunity. We used the well-established Foxp3EGFP-DTR mouse to experimentally deplete Treg cells in mice with syngeneic B16-F10 melanoma heterotopic tumors. We examined the consequences of Treg cell depletion for tumor progression, as measured by blinded serial caliper measurements and tumor immunity, as assessed by scRNA-Seq and flow cytometry. We found that Tconv cells acquire Treg cell-like suppressive functions upon depletion of Treg cells. Using transcriptional profiling and in vitro suppression assays to better understand the nature of this suppressive activity, we found that this suppressive function was enriched among a Th2-like Tconv cell subset marked by expression of CCR8. Moreover, using antibody blockade and conditional Il10 deletion experiments, we found that the suppressive activity induced upon Treg cell depletion was dependent upon IL-10. The sample size for each experiment is specified in the figure legends. The number of independent experiments performed is stated in the figure legends. Age-matched male and female mice were randomly assigned to each group.

Mice

Foxp3EGFP-DTR mice were originally described by Kim et al. (36), Foxp3IRES-EGFP, Ptprca (CD45.1), and Rag2−/− mice were obtained from the Jackson Laboratory. Il10flox/flox and Cd4Cre mice (81) were obtained from Jean Langhorne (Francis Crick Institute) and crossed with Foxp3EGFP-DTR mice to generate Il10flox/flox Cd4Cre Foxp3EGFP-DTR animals. Experiments were performed using 8-14 week old mice, with male and female mice equally distributed between experimental and control groups. Mice were housed at the University of Cambridge University Biomedical Services (UBS) Gurdon Institute Facility and Babraham Institute Biological Services Unit (BSU). Experiments were conducted in accordance with UK Home Office guidelines and were approved by University of Cambridge Animal Welfare and Ethics Review Board or by the Babraham Research Campus Animal Welfare and Ethics Review Board.

Human primary tissues

Primary tumors and adjacent healthy tissue were acquired from 48 NSCLC patients. Patients gave consent to be included in the study which was approved by the institutional review board of Humanitas Research Hospital (protocols no. 2578). Patients did not receive chemotherapy, radiotherapy or palliative surgery before samples were obtained. Samples were processed using the gentleMACS Dissociator (Miltenyi Biotec) into single cell suspensions as previously described (82), resuspended in dimethylsulfoxide (DMSO) with 10% Fetal Bovine Serum (FBS) and stored in liquid nitrogen.

High-dimensional flow cytometry analysis of human samples and computational processing of flow cytometric data

Samples were prepared for flow cytometry as previously described (82). Panels were developed according to an established protocol (83). Briefly, Flow Cytometry Standard (FCS) 3.0 files were analysed by standard gating in FlowJo version 9 to remove dead cells and cell aggregates, and identify CD4+ FOXP3 T cells. 5,000 CD4+ FOXP3 T cells per tumor sample (n = 48) were subsequently imported into FlowJo (version 10), biexponentially transformed, and exported in order to be analyzed by a custom-made publicly available pipeline of PhenoGraph (https://github.com/luglilab/Cytophenograph). A representative gating strategy is shown in Fig. S6. All samples were converted into comma separated value (CSV) files and concatenated in a single matrix by using the merge function of pandas package. The K value, indicating the number of nearest neighbors identified in the first iteration of the algorithm, was set at 500. Uniform Manifold Approximation and Projection (UMAP) was obtained by UMAP Python package.

Tumor challenge and treatment

Mice were injected subcutaneously in the left flank with 1.25×105 B16-F10 melanoma cells (ATCC). Tumors were measured at serial time points following implantation using digital calipers and tumor area was calculated as the length × width. Tumor measurements were completed by an independent investigator who was not aware of treatment groups or genotypes. For late DTx treatment experiments (starting day 10 post-tumor implantation), mice with tumors between 12 and 64 mm2 at day 10 were randomized to treatment groups to reduce experimental variability. Mice were injected intraperitoneally (i.p) starting at the indicated time point with 1 μg of diphtheria toxin (DTx) every other day for a total of four injections and/or 250 μg of anti-IL-10R (clone 1B1.3A; BioXcell) daily for a total of 10 days. DTx from Corynebacterium diphtheriae (Sigma-Aldrich) was obtained in lyophilized powder form and reconstituted in sterile double-distilled water according to the manufacturer’s instructions.

Suppression assays

The suppressive capacity of tumor Tconv cells and Treg cells was measured in vitro as previously described (84). Briefly, CD45.2+ TCRβ+ CD4+ GFP+ Treg cells or CD45.2+ TCRβ+ CD4+ GFP Tconv cells were isolated from B16-F10 tumors of Foxp3EGFP-DTR mice treated with PBS or DTx using florescence-activated cell sorting (FACS) 16 days post implantation and used as suppressor cells. A representative gating strategy is shown in shown in Fig. S2. Naïve CD4+ Tconv cells (CD25 CD44 CD62L+) were purified from the spleens of WT CD45.1 mice by FACS and stained with CellTrace Violet™ (CTV) according to the manufacturer’s protocol (Thermo Fisher Scientific) and used as responder T (Tresp) cells. 2.5×104 or 1.25×104 (as indicated) suppressor CD4+ Treg cells or Tconv cells were co-cultured with 1×105 naïve responder T cells in the presence of anti-CD3 (BioLegend 1 μg/mL) and 5.0×104 Rag2−/− antigen presenting cells (APCs). Tresp cells cultured in the presence of anti-CD3 and APCs but without tumor Treg cells or Tconv cells were used as a control. Cell division was evaluated by flow cytometry after 4 days of culture. For the screen, cells were cultured alone (gray) or with suppressors (purple) and with Aminoglutethimide (AG) at a final concentration of 125μM or with other indicated reagents at a final concentration of 10μg/mL.

Flow cytometry of murine samples

Tumor samples were digested using collagenase and DNase for 30 minutes at 37 °C. Percoll was used to isolate lymphocytes from tumors. Tumors and spleens were mechanically dissociated over a 40 μm cell strainer. Red blood cells were lysed using ACK Lysing Buffer (Gibco). Cells were stained with the Fixable Viability Dye eFluor™ 780 (Thermo Fisher Scientific), Viakrome 808 (Beckman Coulter), or DAPI (Sigma) to discriminate between live and dead cells and then incubated with FC block (BioXcell, 2.4G2) the following surface antibodies for 30 minutes on ice: anti-TCRβ FITC (H57-597), anti-CD8 BV605 (53-6.7), anti-TIM-3 BV421 (RMT3-23), anti-CCR8 BV421 (SA214G2), anti-OX40 BV711 (OX-86), anti-TIGIT PE (4D4/mTIGIT), anti-ICOS BV750 (C398.4A), anti-LAG-3 BV785 (C9B7W), anti-CD3ε Spark Blue 550 (17A2) from Biolegend, anti-CD25 PE-Cyanine7 (PC61.5), anti-CD44 PerCP-Cyanine5.5 (IM7), anti-CD45.1 APC (A20), anti-CD4 PE-Cy7 (RM4-5) from eBioscience, and anti-GITR BUV805 (DTA-1), anti-CD4 BUV395 or BUV496 (GK1.5) from BD Biosciences. Cells were stimulated with phorbol 12-myristate 13-acetate (PMA) and ionomycin and blocked with brefeldin A (BFA) for 4 hours in RPMI 1640 complete medium. The intracellular antibodies anti-Foxp3 APC (FJK-16S), anti-IFN-γ PerCPCy5.5 (XMG1.2), anti-GATA3 PE-eFluor610 (TWAJ) were purchased from eBioscience, and anti-CTLA-4 BV605 (UC10-4B9), anti-TNF PE-Cy7 (MP6-XT22) were purchased from BioLegend and used with the eBioscience Foxp3/Transcription Factor Staining Buffer Set (Invitrogen, Thermo Fisher Scientific) according to the manufacturer’s protocol. For determination of CCR8 expression by Foxp3 Tconv cells, cells from tumor-bearing Foxp3EGFP-DTR animals were surface-stained and analyzed unfixed using flow cytometry, with EGFP expression used to discriminate Treg cells and Tconv cells. Samples were analyzed using BD Fortessa, Beckman Coulter CytoFLEX, and Cytek® Aurora analyzers. After analysis, data were analyzed using FlowJo software (Tree Star, Inc.).

scRNA Sequencing and analysis

Single cell suspensions of T cells were purified by total pan T cell enrichment (Invitrogen/Thermo Fisher Scientific), and live TCRβ+ cells were sorted from B16 tumors by FACS 16 days post implantation. RNA libraries were prepared for single cell RNA sequencing (scRNA-Seq) using the Chromium Single Cell 5’ Library & Gel Bead Kit v2 (10x Genomics) processed with Chromium (10x Genomics), and sequenced using the HiSeq 4000 System (Illumnia). Raw 10x sequencing data were processed as previously described and mapped to mm10. We confirmed that cells were sequenced to saturation. Data were merged with cell ranger aggr (cellranger-v5.0.0). Merged data were transferred to the R statistical environment for analysis primarily using the package Seurat (v3.2.2) in R v4.0.3. The analysis included only cells expressing between 200 and 2,500 genes, <5% mitochondrial-associated transcripts, and genes expressed in at least three cells. The data were then log-normalized and scaled per cell, and variable genes were detected using the FindVariableFeatures function in Seurat, as per default settings, using 2000 features and further processed as per the ScaleData function. Principal component analysis (PCA) was run on the variable genes, and the first six principal components (PCs) were selected for further analyses, based on the standard deviation of the PCs, as determined by an “elbow plot” in Seurat. Cells were clustered using the FindClusters function in Seurat with default settings, resolution = 0.5, and six PCs. UMAP was calculated using six PCs (RunUMAP function). For broadly defining the transcriptional features of each cluster, the FindAllMarkers function (only.pos = FALSE, min.pct = 0.1, thresh.use = 0.2, test.use = “MAST”) was used, and the associated heatmap was generated using the DoHeatmap function using up to the top 10 transcripts identified per cluster as defined by FindAllMarkers. The transcriptomic score of a particular cluster was calculated using the AddModuleScore function with default settings. Further visualizations of exported normalized data were generated using the Seurat RidgePlot functions and custom R scripts.

RNA Sequencing and analysis

Single-cell suspensions were purified by FACS (as described above) 16 days post tumor implantation, and stored in 40 μl RNAlater™ Stabilization Solution at -80 °C. RNA was extracted from samples using the RNeasy Plus Mini Kit (Qiagen) with optional QIAshredder step according to the manufacturer’s protocol. RNA-Sequencing (RNA-Seq) analyses were performed using ≥ 2 biological replicates. RNA-Seq was performed and analyzed as described previously (59). RNA Libraries were prepared using the Clontech SMARTer Ultra Low-input RNA kit (Takara) and sequenced on an Illumina HiSeq 2500 instrument using Illumina TruSeq v4.0 chemistry. The resulting FastQ files underwent quality control with FastQC, adaptor trimming with Cutadapt and alignment to the NCBIM37 Mus musculus genome annotation with hisat2 using ClusterFlow pipelines. Uniquely mapped reads were used to calculate gene expression and FPM values normalized to total library size with intergenic read normalization were calculated. Differential expression and statistical significance were calculated using the Wald test with adjustment for multiple testing using the Benjamini-Hochberg method using DESeq2 (85). Differentially expressed genes were further analyzed using R. PCA was performed using R plotPCA with count data transformed using variance stabilizing transformation (VST) from fitted dispersion-mean relationships generated using DESeq2 vst. Expression heatmaps were generated using FPM values normalized to row maxima using the R pheatmap package. Hierarchical clustering was performed using the Ward method. Dendrograms were cut at levels sufficient to allow 3-5 clusters to be discriminated.

Statistical Analysis

Statistical analysis was performed using Graphpad Prism software. Two-tailed Student’s t tests or one-way ordinary ANOVAs were used as indicated to calculate statistical significance of the difference in sample means. P values of less than 0.05 were considered statistically significant. Statistical tests used are specified in the figure legends. In all figures, data represent the mean ± the standard error of the mean (SEM) or standard deviation (SD) as indicated. P values correlate with symbols as follows: ns = not significant, * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001.

Supplementary Material

Data file S1
Data file S2
Data file S3
Data file S4
Data file S5
Data file S6
Supplementary Figures

One sentence summary.

CCR8+ Tconv cells undergo expansion and activation upon Treg cell depletion and limit anti-tumor immunity through production of IL-10.

Acknowledgements

We thank members of the University of Cambridge UBS Gurdon Facility and Babraham Institute Biological Services Facility for technical support with animal experiments. The authors gratefully acknowledge the flow cytometry facility from the School of the Biological Sciences for their support and assistance in this work, and Dr. Simone Puccio (Humanitas) for help with computational analyses, and the members of the high-throughput sequencing facilities. We thank staff at the CRUK Cambridge Institute sequencing facility for their assistance with single-cell RNA-Seq. We thank members of the Roychoudhuri laboratory for sharing of reagents, protocols, ideas and discussion. We apologize to those authors whose work we were unable to reference due to space restrictions.

Funding

The research was supported by the UK Medical Research Council (MRC; grant MR/S024468/1), the Wellcome Trust / Royal Society (grant 105663/Z/14/Z), Cancer Research UK (grant C52623/A22597), the Italian Ministry of Health (Grant Giovani Ricercatori GR-2018-12367258 to E.L.). E.L. is a CRI Llyod J. Old STAR (CRI award 3914).

Footnotes

Author contributions

S.K.W. and R.R. conceived and designed experiments; S.K.W., F.M.G., G.A., J.C., L.T., C.J.I., B.Z., A.C.E., A.C., A.W., S.N.L., M.C., J.D. conducted experiments, S.K.W., G.A., J.C., A.C.E., E.L., and R.R. performed analyses and data visualization. S.K.W., I.H.R., E.L., and R.R. provided funding; S.K.W. O.B., J.C., J.Y., I.R.H., J.L., K.O., and R.R., developed the methodology used; S.K.W., O.B., A.L., E.L., and R.R. provided supervision; M.A., E.V., G.A., and E.L., coordinated and obtained he clinical samples, S.K.W. and R.R wrote the original draft, S.K.W., C.J.I., E.L., and R.R. reviewed and edited the paper.

Competing interests

R.R. holds or has held paid consultancies with Lyell Immunopharma, Achilles Therapeutics and Enhanc3D Genomics; and is a principal investigator of research projects funded by AstraZeneca and F-star Therapeutics on unrelated topics that do not constitute competing interests. E.L. served as a consltant for BD Biosciences on a topic unrelated to this work. All other authors declare no competing interests.

Data and materials availability

All bulk RNA-Seq and scRNA-Seq data will be made publicly available under NCBI Gene Expression Omnibus (GEO) accession number GSE236825. All other data needed to support the conclusions of the paper are present in the paper or the Supplementary Materials.

References

  • 1.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–723. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Robert C, Thomas L, Bondarenko I, O’Day S, Weber J, Garbe C, Lebbe C, Baurain JF, Testori A, Grob JJ, Davidson N, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–2526. doi: 10.1056/NEJMoa1104621. [DOI] [PubMed] [Google Scholar]
  • 3.Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, Ivanova Y, Hundal J, Arthur CD, Krebber WJ, Mulder GE, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature. 2014;515:577–581. doi: 10.1038/nature13988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Larkin J, Lao CD, Urba WJ, McDermott DF, Horak C, Jiang J, Wolchok JD. Efficacy and Safety of Nivolumab in Patients With BRAF V600 Mutant and BRAF Wild-Type Advanced Melanoma: A Pooled Analysis of 4 Clinical Trials. JAMA Oncol. 2015;1:433–440. doi: 10.1001/jamaoncol.2015.1184. [DOI] [PubMed] [Google Scholar]
  • 5.Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, Ferrucci PF, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med. 2015;373:23–34. doi: 10.1056/NEJMoa1504030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell. 2017;168:707–723. doi: 10.1016/j.cell.2017.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, Gottfried M, Peled N, Tafreshi A, Cuffe S, O’Brien M, Rao S, et al. K.-. Investigators, Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl JMed. 2016;375:1823–1833. doi: 10.1056/NEJMoa1606774. [DOI] [PubMed] [Google Scholar]
  • 8.Hamid O, Molinero L, Bolen CR, Sosman JA, Munoz-Couselo E, Kluger HM, McDermott DF, Powderly JD, Sarkar I, Ballinger M, Fasso M, et al. Safety, Clinical Activity, and Biological Correlates of Response in Patients with Metastatic Melanoma: Results from a Phase I Trial of Atezolizumab. Clin Cancer Res. 2019;25:6061–6072. doi: 10.1158/1078-0432.CCR-18-3488. [DOI] [PubMed] [Google Scholar]
  • 9.Plitas G, Rudensky AY. Regulatory T Cells in Cancer. Annual Review of Cancer Biology. 2020;4:459–477. [Google Scholar]
  • 10.De Peaux K, Delgoffe GM. Metabolic barriers to cancer immunotherapy. Nature review Immunologys. 2021 doi: 10.1038/s41577-021-00541-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Sakaguchi S, Mikami N, Wing JB, Tanaka A, Ichiyama K, Ohkura N. Regulatory T Cells and Human Disease. Annu Rev Immunol. 2020;38:541–566. doi: 10.1146/annurev-immunol-042718-041717. [DOI] [PubMed] [Google Scholar]
  • 12.Gallimore A, Quezada SA, Roychoudhuri R. Regulatory T cells in cancer: where are we now? Immunology. 2019;157:187–189. doi: 10.1111/imm.13088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Stockis J, Roychoudhuri R, Halim TYF. Regulation of regulatory T cells in cancer. Immunology. 2019;157:219–231. doi: 10.1111/imm.13064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Roychoudhuri R, Eil RL, Restifo NP. The interplay of effector and regulatory T cells in cancer. Curr Opin Immunol. 2015;33:101–111. doi: 10.1016/j.coi.2015.02.003. [DOI] [PubMed] [Google Scholar]
  • 15.Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, Jungbluth AA, Frosina D, Gnjatic S, Ambrosone C, Kepner J, et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A. 2005;102:18538–18543. doi: 10.1073/pnas.0509182102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, Evdemon-Hogan M, Conejo-Garcia JR, Zhang L, Burow M, Zhu Y, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–949. doi: 10.1038/nm1093. [DOI] [PubMed] [Google Scholar]
  • 17.Bates GJ, Fox SB, Han C, Leek RD, Garcia JF, Harris AL, Banham AH. Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. J Clin Oncol. 2006;24:5373–5380. doi: 10.1200/JCO.2006.05.9584. [DOI] [PubMed] [Google Scholar]
  • 18.Petersen RP, Campa MJ, Sperlazza J, Conlon D, Joshi MB, Harpole DH, Jr, Patz EF., Jr Tumor infiltrating Foxp3+ regulatory T-cells are associated with recurrence in pathologic stage I NSCLC patients. Cancer. 2006;107:2866–2872. doi: 10.1002/cncr.22282. [DOI] [PubMed] [Google Scholar]
  • 19.Gao Q, Qiu SJ, Fan J, Zhou J, Wang XY, Xiao YS, Xu Y, Li YW, Tang ZY. Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. J Clin Oncol. 2007;25:2586–2593. doi: 10.1200/JCO.2006.09.4565. [DOI] [PubMed] [Google Scholar]
  • 20.Griffiths RW, Elkord E, Gilham DE, Ramani V, Clarke N, Stern PL, Hawkins RE. Frequency of regulatory T cells in renal cell carcinoma patients and investigation of correlation with survival. Cancer Immunol Immunother. 2007;56:1743–1753. doi: 10.1007/s00262-007-0318-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hiraoka N, Onozato K, Kosuge T, Hirohashi S. Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions. Clin Cancer Res. 2006;12:5423–5434. doi: 10.1158/1078-0432.CCR-06-0369. [DOI] [PubMed] [Google Scholar]
  • 22.Perrone G, Ruffini PA, Catalano V, Spino C, Santini D, Muretto P, Spoto C, Zingaretti C, Sisti V, Alessandroni P, Giordani P, et al. Intratumoural FOXP3-positive regulatory T cells are associated with adverse prognosis in radically resected gastric cancer. Eur J Cancer. 2008;44:1875–1882. doi: 10.1016/j.ejca.2008.05.017. [DOI] [PubMed] [Google Scholar]
  • 23.Jordanova ES, Gorter A, Ayachi O, Prins F, Durrant LG, Kenter GG, van der Burg SH, Fleuren GJ. Human leukocyte antigen class I, MHC class I chain-related molecule A, and CD8+/regulatory T-cell ratio: which variable determines survival of cervical cancer patients? Clin Cancer Res. 2008;14:2028–2035. doi: 10.1158/1078-0432.CCR-07-4554. [DOI] [PubMed] [Google Scholar]
  • 24.Alvisi G, Termanini A, Soldani C, Portale F, Carriero R, Pilipow K, Costa G, Polidoro M, Franceschini B, Malenica I, Puccio S, et al. Multimodal single-cell profiling of intrahepatic cholangiocarcinoma defines hyperactivated Tregs as a potential therapeutic target. J Hepatol. 2022;77:1359–1372. doi: 10.1016/j.jhep.2022.05.043. [DOI] [PubMed] [Google Scholar]
  • 25.Sinicrope FA, Rego RL, Ansell SM, Knutson KL, Foster NR, Sargent DJ. Intraepithelial effector (CD3+)/regulatory (FoxP3+) T-cell ratio predicts a clinical outcome of human colon carcinoma. Gastroenterology. 2009;137:1270–1279. doi: 10.1053/j.gastro.2009.06.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Maj T, Wang W, Crespo J, Zhang H, Wang W, Wei S, Zhao L, Vatan L, Shao I, Szeliga W, Lyssiotis C, et al. Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat Immunol. 2017;18:1332–1341. doi: 10.1038/ni.3868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Simpson TR, Li F, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F, Roddie C, Henry JY, Yagita H, Wolchok JD, Peggs KS, 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;210:1695–1710. doi: 10.1084/jem.20130579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Yu P, Lee Y, Liu W, Krausz T, Chong A, Schreiber H, Fu YX. Intratumor depletion of CD4+ cells unmasks tumor immunogenicity leading to the rejection of late-stage tumors. J Exp Med. 2005;201:779–791. doi: 10.1084/jem.20041684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Liu C, Workman CJ, Vignali DA. Targeting regulatory T cells in tumors. FEBS J. 2016;283:2731–2748. doi: 10.1111/febs.13656. [DOI] [PubMed] [Google Scholar]
  • 30.Jacobs JF, Punt CJ, Lesterhuis WJ, Sutmuller RP, Brouwer HM, Scharenborg NM, Klasen IS, Hilbrands LB, Figdor CG, de Vries IJ, Adema GJ. Dendritic cell vaccination in combination with anti-CD25 monoclonal antibody treatment: a phase I/II study in metastatic melanoma patients. Clin Cancer Res. 2010;16:5067–5078. doi: 10.1158/1078-0432.CCR-10-1757. [DOI] [PubMed] [Google Scholar]
  • 31.Sampson JH, Schmittling RJ, Archer GE, Congdon KL, Nair SK, Reap EA, Desjardins A, Friedman AH, Friedman HS, Herndon JE, 2nd, Coan A, et al. A pilot study of IL-2Ralpha blockade during lymphopenia depletes regulatory T-cells and correlates with enhanced immunity in patients with glioblastoma. PLoS One. 2012;7:e31046. doi: 10.1371/journal.pone.0031046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Rech AJ, Mick R, Martin S, Recio A, Aqui NA, Powell DJ, Jr, Colligon TA, Trosko JA, Leinbach LI, Pletcher CH, Tweed CK, et al. CD25 blockade depletes and selectively reprograms regulatory T cells in concert with immunotherapy in cancer patients. Sci Transl Med. 2012;4:134ra162. doi: 10.1126/scitranslmed.3003330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Attia P, Maker AV, Haworth LR, Rogers-Freezer L, Rosenberg SA. Inability of a fusion protein of IL-2 and diphtheria toxin (Denileukin Diftitox, DAB389IL-2, ONTAK) to eliminate regulatory T lymphocytes in patients with melanoma. J Immunother. 2005;28:582–592. doi: 10.1097/01.cji.0000175468.19742.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zamarin D, Hamid O, Nayak-Kapoor A, Sahebjam S, Sznol M, Collaku A, Fox FE, Marshall MA, Hong DS. Mogamulizumab in Combination with Durvalumab or Tremelimumab in Patients with Advanced Solid Tumors: A Phase I Study. Clin Cancer Res. 2020;26:4531–4541. doi: 10.1158/1078-0432.CCR-20-0328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kurose K, Ohue Y, Wada H, Iida S, Ishida T, Kojima T, Doi T, Suzuki S, Isobe M, Funakoshi T, Kakimi K, et al. Phase Ia Study of FoxP3+ CD4 Treg Depletion by Infusion of a Humanized Anti-CCR4 Antibody, KW-0761, in Cancer Patients. Clin Cancer Res. 2015;21:4327–4336. doi: 10.1158/1078-0432.CCR-15-0357. [DOI] [PubMed] [Google Scholar]
  • 36.Kim JM, Rasmussen JP, Rudensky AY. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol. 2007;8:191–197. doi: 10.1038/ni1428. [DOI] [PubMed] [Google Scholar]
  • 37.Takahashi T, Kuniyasu Y, Toda M, Sakaguchi N, Itoh M, Iwata M, Shimizu J, Sakaguchi S. Immunologic self-tolerance maintained by CD25+CD4+ naturally anergic and suppressive T cells: induction of autoimmune disease by breaking their anergic/suppressive state. Int Immunol. 1998;10:1969–1980. doi: 10.1093/intimm/10.12.1969. [DOI] [PubMed] [Google Scholar]
  • 38.Collison LW, Pillai MR, Chaturvedi V, Vignali DA. Regulatory T cell suppression is potentiated by target T cells in a cell contact, IL-35- and IL-10-dependent manner. J Immunol. 2009;182:6121–6128. doi: 10.4049/jimmunol.0803646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Thornton AM, Shevach EM. CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J Exp Med. 1998;188:287–296. doi: 10.1084/jem.188.2.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Villarreal DO, L’Huillier A, Armington S, Mottershead C, Filippova EV, Coder BD, Petit RG, Princiotta MF. Targeting CCR8 Induces Protective Antitumor Immunity and Enhances Vaccine-Induced Responses in Colon Cancer. Cancer Res. 2018;78:5340–5348. doi: 10.1158/0008-5472.CAN-18-1119. [DOI] [PubMed] [Google Scholar]
  • 41.Whiteside SK, Grant FM, Gyori DS, Conti AG, Imianowski CJ, Kuo P, Nasrallah R, Sadiyah F, Lira SA, Tacke F, Eil RL, et al. CCR8 marks highly suppressive Treg cells within tumours but is dispensable for their accumulation and suppressive function. Immunology. 2021;163:512–520. doi: 10.1111/imm.13337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Van Damme H, Dombrecht B, Kiss M, Roose H, Allen E, Van Overmeire E, Kancheva D, Martens L, Murgaski A, Bardet PMR, Blancke G, et al. Therapeutic depletion of CCR8(+) tumor-infiltrating regulatory T cells elicits antitumor immunity and synergizes with anti-PD-1 therapy. J Immunother Cancer. 2021;9 doi: 10.1136/jitc-2020-001749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Campbell JR, McDonald BR, Mesko PB, Siemers NO, Singh PB, Selby M, Sproul TW, Korman AJ, Vlach LM, Houser J, Sambanthamoorthy S, et al. Fc-Optimized Anti-CCR8 Antibody Depletes Regulatory T Cells in Human Tumor Models. Cancer Res. 2021;81:2983–2994. doi: 10.1158/0008-5472.CAN-20-3585. [DOI] [PubMed] [Google Scholar]
  • 44.Alvisi G, Brummelman J, Puccio S, Mazza EM, Tomada EP, Losurdo A, Zanon V, Peano C, Colombo FS, Scarpa A, Alloisio M, et al. IRF4 instructs effector Treg differentiation and immune suppression in human cancer. J Clin Invest. 2020;130:3137–3150. doi: 10.1172/JCI130426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mahata B, Pramanik J, van der Weyden L, Polanski K, Kar G, Riedel A, Chen X, Fonseca NA, Kundu K, Campos LS, Ryder E, et al. Tumors induce de novo steroid biosynthesis in T cells to evade immunity. Nat Commun. 2020;11:3588. doi: 10.1038/s41467-020-17339-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Groux H, O’Garra A, Bigler M, Rouleau M, Antonenko S, de Vries JE, Roncarolo MG. A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature. 1997;389:737–742. doi: 10.1038/39614. [DOI] [PubMed] [Google Scholar]
  • 47.Roncarolo MG, Yssel H, Touraine JL, Betuel H, De Vries JE, Spits H. Autoreactive T cell clones specific for class I and class II HLA antigens isolated from a human chimera. J Exp Med. 1988;167:1523–1534. doi: 10.1084/jem.167.5.1523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gagliani N, Magnani CF, Huber S, Gianolini ME, Pala M, Licona-Limon P, Guo B, Herbert DR, Bulfone A, Trentini F, Di Serio C, et al. Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells. Nat Med. 2013;19:739–746. doi: 10.1038/nm.3179. [DOI] [PubMed] [Google Scholar]
  • 49.Pedroza-Gonzalez A, Zhou G, Vargas-Mendez E, Boor PP, Mancham S, Verhoef C, Polak WG, Grunhagen D, Pan Q, Janssen H, Garcia-Romo GS, Biermann K, et al. Tumor-infiltrating plasmacytoid dendritic cells promote immunosuppression by Tr1 cells in human liver tumors. Oncoimmunology. 2015;4:e1008355. doi: 10.1080/2162402X.2015.1008355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Zhang P, Lee JS, Gartlan KH, Schuster IS, Comerford I, Varelias A, Ullah MA, Vuckovic S, Koyama M, Kuns RD, Locke KR, et al. Eomesodermin promotes the development of type 1 regulatory T (T(R)1) cells. Sci Immunol. 2017;2 doi: 10.1126/sciimmunol.aah7152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gruarin P, Maglie S, De Simone M, Haringer B, Vasco C, Ranzani V, Bosotti R, Noddings JS, Larghi P, Facciotti F, Sarnicola ML, et al. Eomesodermin controls a unique differentiation program in human IL-10 and IFN-gamma coproducing regulatory T cells. Eur J Immunol. 2019;49:96–111. doi: 10.1002/eji.201847722. [DOI] [PubMed] [Google Scholar]
  • 52.Bonnal RJP, Rossetti G, Lugli E, De Simone M, Gruarin P, Brummelman J, Drufuca L, Passaro M, Bason R, Gervasoni F, Della Chiara G, et al. Clonally expanded EOMES(+) Tr1-like cells in primary and metastatic tumors are associated with disease progression. Nat Immunol. 2021;22:735–745. doi: 10.1038/s41590-021-00930-4. [DOI] [PubMed] [Google Scholar]
  • 53.Jin HT, Anderson AC, Tan WG, West EE, Ha SJ, Araki K, Freeman GJ, Kuchroo VK, Ahmed R. Cooperation of Tim-3 and PD-1 in CD8 T-cell exhaustion during chronic viral infection. Proc Natl Acad Sci U S A. 2010;107:14733–14738. doi: 10.1073/pnas.1009731107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Fiorentino DF, Bond MW, Mosmann TR. Two types of mouse T helper cell. IV. Th2 clones secrete a factor that inhibits cytokine production by Th1 clones. J Exp Med. 1989;170:2081–2095. doi: 10.1084/jem.170.6.2081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Moore KW, de Waal Malefyt R, Coffman RL, O’Garra A. Interleukin-10 and the interleukin-10 receptor. Annu Rev Immunol. 2001;19:683–765. doi: 10.1146/annurev.immunol.19.1.683. [DOI] [PubMed] [Google Scholar]
  • 56.Tanikawa T, Wilke CM, Kryczek I, Chen GY, Kao J, Nunez G, Zou W. Interleukin-10 ablation promotes tumor development, growth, and metastasis. Cancer Res. 2012;72:420–429. doi: 10.1158/0008-5472.CAN-10-4627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Pastille E, Bardini K, Fleissner D, Adamczyk A, Frede A, Wadwa M, von Smolinski D, Kasper S, Sparwasser T, Gruber AD, Schuler M, et al. Transient ablation of regulatory T cells improves antitumor immunity in colitis-associated colon cancer. Cancer Res. 2014;74:4258–4269. doi: 10.1158/0008-5472.CAN-13-3065. [DOI] [PubMed] [Google Scholar]
  • 58.Grant FM, Yang J, Nasrallah R, Clarke J, Sadiyah F, Whiteside SK, Imianowski CJ, Kuo P, Vardaka P, Todorov T, Zandhuis N, et al. BACH2 drives quiescence and maintenance of resting Treg cells to promote homeostasis and cancer immunosuppression. J Exp Med. 2020;217 doi: 10.1084/jem.20190711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Roychoudhuri R, Clever D, Li P, Wakabayashi Y, Quinn KM, Klebanoff CA, Ji Y, Sukumar M, Eil RL, Yu Z, Spolski R, et al. BACH2 regulates CD8(+) T cell differentiation by controlling access of AP-1 factors to enhancers. Nat Immunol. 2016;17:851–860. doi: 10.1038/ni.3441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Igarashi K, Kurosaki T, Roychoudhuri R. BACH transcription factors in innate and adaptive immunity. Nat Rev Immunol. 2017;17:437–450. doi: 10.1038/nri.2017.26. [DOI] [PubMed] [Google Scholar]
  • 61.De Simone M, Arrigoni A, Rossetti G, Gruarin P, Ranzani V, Politano C, Bonnal RJP, Provasi E, Sarnicola ML, Panzeri I, Moro M, et al. Transcriptional Landscape of Human Tissue Lymphocytes Unveils Uniqueness of Tumor-Infiltrating T Regulatory Cells. Immunity. 2016;45:1135–1147. doi: 10.1016/j.immuni.2016.10.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Plitas G, Konopacki C, Wu K, Bos PD, Morrow M, Putintseva EV, Chudakov DM, Rudensky AY. Regulatory T Cells Exhibit Distinct Features in Human Breast Cancer. Immunity. 2016;45:1122–1134. doi: 10.1016/j.immuni.2016.10.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kidani Y, Nogami W, Yasumizu Y, Kawashima A, Tanaka A, Sonoda Y, Tona Y, Nashiki K, Matsumoto R, Hagiwara M, Osaki M, et al. CCR8-targeted specific depletion of clonally expanded Treg cells in tumor tissues evokes potent tumor immunity with long-lasting memory. Proc Natl Acad Sci U S A. 2022;119 doi: 10.1073/pnas.2114282119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Bergmann C, Strauss L, Wang Y, Szczepanski MJ, Lang S, Johnson JT, Whiteside TL. T regulatory type 1 cells in squamous cell carcinoma of the head and neck: mechanisms of suppression and expansion in advanced disease. Clin Cancer Res. 2008;14:3706–3715. doi: 10.1158/1078-0432.CCR-07-5126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Scurr M, Ladell K, Besneux M, Christian A, Hockey T, Smart K, Bridgeman H, Hargest R, Phillips S, Davies M, Price D, et al. Highly prevalent colorectal cancer-infiltrating LAP(+) Foxp3(-) T cells exhibit more potent immunosuppressive activity than Foxp3(+) regulatory T cells. Mucosal Immunol. 2014;7:428–439. doi: 10.1038/mi.2013.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Marshall NA, Christie LE, Munro LR, Culligan DJ, Johnston PW, Barker RN, Vickers MA. Immunosuppressive regulatory T cells are abundant in the reactive lymphocytes of Hodgkin lymphoma. Blood. 2004;103:1755–1762. doi: 10.1182/blood-2003-07-2594. [DOI] [PubMed] [Google Scholar]
  • 67.Yan H, Zhang P, Kong X, Hou X, Zhao L, Li T, Yuan X, Fu H. Primary Tr1 cells from metastatic melanoma eliminate tumor-promoting macrophages through granzyme B- and perforin-dependent mechanisms. Tumour Biol. 2017;39:1010428317697554. doi: 10.1177/1010428317697554. [DOI] [PubMed] [Google Scholar]
  • 68.Zappasodi R, Budhu S, Hellmann MD, Postow MA, Senbabaoglu Y, Manne S, Gasmi B, Liu C, Zhong H, Li Y, Huang AC, et al. Non-conventional Inhibitory CD4(+)Foxp3(-)PD-1(hi) T Cells as a Biomarker of Immune Checkpoint Blockade Activity. Cancer Cell. 2018;33:1017–1032.:e1017. doi: 10.1016/j.ccell.2018.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Eschweiler S, Clarke J, Ramirez-Suastegui C, Panwar B, Madrigal A, Chee SJ, Karydis I, Woo E, Alzetani A, Elsheikh S, Hanley CJ, et al. Intratumoral follicular regulatory T cells curtail anti-PD-1 treatment efficacy. Nat Immunol. 2021;22:1052–1063. doi: 10.1038/s41590-021-00958-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lahl K, Mayer CT, Bopp T, Huehn J, Loddenkemper C, Eberl G, Wirnsberger G, Dornmair K, Geffers R, Schmitt E, Buer J, et al. Nonfunctional regulatory T cells and defective control of Th2 cytokine production in natural scurfy mutant mice. J Immunol. 2009;183:5662–5672. doi: 10.4049/jimmunol.0803762. [DOI] [PubMed] [Google Scholar]
  • 71.Tian L, Altin JA, Makaroff LE, Franckaert D, Cook MC, Goodnow CC, Dooley J, Liston A. Foxp3(+) regulatory T cells exert asymmetric control over murine helper responses by inducing Th2 cell apoptosis. Blood. 2011;118:1845–1853. doi: 10.1182/blood-2011-04-346056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Freeman CM, Chiu BC, Stolberg VR, Hu J, Zeibecoglou K, Lukacs NW, Lira SA, Kunkel SL, Chensue SW. CCR8 is expressed by antigen-elicited, IL-10-producing CD4+CD25+ T cells, which regulate Th2-mediated granuloma formation in mice. J Immunol. 2005;174:1962–1970. doi: 10.4049/jimmunol.174.4.1962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Mikhak Z, Fukui M, Farsidjani A, Medoff BD, Tager AM, Luster AD. Contribution of CCR4 and CCR8 to antigen-specific T(H)2 cell trafficking in allergic pulmonary inflammation. J Allergy Clin Immunol. 2009;123:67–73.:e63. doi: 10.1016/j.jaci.2008.09.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.McCully ML, Ladell K, Andrews R, Jones RE, Miners KL, Roger L, Baird DM, Cameron MJ, Jessop ZM, Whitaker IS, Davies EL, et al. CCR8 Expression Defines Tissue-Resident Memory T Cells in Human Skin. J Immunol. 2018;200:1639–1650. doi: 10.4049/jimmunol.1701377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Guo Y, Xie YQ, Gao M, Zhao Y, Franco F, Wenes M, Siddiqui I, Bevilacqua A, Wang H, Yang H, Feng B, et al. Metabolic reprogramming of terminally exhausted CD8(+) T cells by IL-10 enhances anti-tumor immunity. Nat Immunol. 2021;22:746–756. doi: 10.1038/s41590-021-00940-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Naing A, Infante JR, Papadopoulos KP, Chan IH, Shen C, Ratti NP, Rojo B, Autio KA, Wong DJ, Patel MR, Ott PA, et al. PEGylated IL-10 (Pegilodecakin) Induces Systemic Immune Activation, CD8(+) T Cell Invigoration and Polyclonal T Cell Expansion in Cancer Patients. Cancer Cell. 2018;34:775–791.:e773. doi: 10.1016/j.ccell.2018.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Tannir NM, Papadopoulos KP, Wong DJ, Aljumaily R, Hung A, Afable M, Kim JS, Ferry D, Drakaki A, Bendell J, Naing A. Pegilodecakin as monotherapy or in combination with anti-PD-1 or tyrosine kinase inhibitor in heavily pretreated patients with advanced renal cell carcinoma: Final results of cohorts A, G, H and I of IVY Phase I study. Int J Cancer. 2021;149:403–408. doi: 10.1002/ijc.33556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Glisson BS, Leidner RS, Ferris RL, Powderly J, Rizvi NA, Keam B, Schneider R, Goel S, Ohr JP, Burton J, Zheng Y, et al. Safety and Clinical Activity of MEDI0562, a Humanized OX40 Agonist Monoclonal Antibody, in Adult Patients with Advanced Solid Tumors. Clin Cancer Res. 2020;26:5358–5367. doi: 10.1158/1078-0432.CCR-19-3070. [DOI] [PubMed] [Google Scholar]
  • 79.Romano E, Kusio-Kobialka M, Foukas PG, Baumgaertner P, Meyer C, Ballabeni P, Michielin O, Weide B, Romero P, Speiser DE. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc Natl Acad Sci U S A. 2015;112:6140–6145. doi: 10.1073/pnas.1417320112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Arce Vargas F, Furness AJS, Litchfield K, Joshi K, Rosenthal R, Ghorani E, Solomon I, Lesko MH, Ruef N, Roddie C, Henry JY, et al. Fc Effector Function Contributes to the Activity of Human Anti-CTLA-4 Antibodies. Cancer Cell. 2018;33:649–663.:e644. doi: 10.1016/j.ccell.2018.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Freitas do Rosario AP, Lamb T, Spence P, Stephens R, Lang A, Roers A, Muller W, O’Garra A, Langhorne J. IL-27 promotes IL-10 production by effector Th1 CD4+ T cells: a critical mechanism for protection from severe immunopathology during malaria infection. J Immunol. 2012;188:1178–1190. doi: 10.4049/jimmunol.1102755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Brummelman J, Mazza EMC, Alvisi G, Colombo FS, Grilli A, Mikulak J, Mavilio D, Alloisio M, Ferrari F, Lopci E, Novellis P, et al. High-dimensional single cell analysis identifies stem-like cytotoxic CD8(+) T cells infiltrating human tumors. J Exp Med. 2018;215:2520–2535. doi: 10.1084/jem.20180684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Brummelman J, Haftmann C, Nunez NG, Alvisi G, Mazza EMC, Becher B, Lugli E. Development, application and computational analysis of high-dimensional fluorescent antibody panels for single-cell flow cytometry. Nat Protoc. 2019;14:1946–1969. doi: 10.1038/s41596-019-0166-2. [DOI] [PubMed] [Google Scholar]
  • 84.Collison LW, Vignali DA. In vitro Treg suppression assays. Methods Mol Biol. 2011;707:21–37. doi: 10.1007/978-1-61737-979-6_2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Data file S1
Data file S2
Data file S3
Data file S4
Data file S5
Data file S6
Supplementary Figures

Data Availability Statement

All bulk RNA-Seq and scRNA-Seq data will be made publicly available under NCBI Gene Expression Omnibus (GEO) accession number GSE236825. All other data needed to support the conclusions of the paper are present in the paper or the Supplementary Materials.

RESOURCES