Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 Jan 24;15(1):682.
doi: 10.1038/s41467-024-44787-1.

Tumour-retained activated CCR7+ dendritic cells are heterogeneous and regulate local anti-tumour cytolytic activity

Affiliations

Tumour-retained activated CCR7+ dendritic cells are heterogeneous and regulate local anti-tumour cytolytic activity

Colin Y C Lee et al. Nat Commun. .

Abstract

Tumour dendritic cells (DCs) internalise antigen and upregulate CCR7, which directs their migration to tumour-draining lymph nodes (dLN). CCR7 expression is coupled to an activation programme enriched in regulatory molecule expression, including PD-L1. However, the spatio-temporal dynamics of CCR7+ DCs in anti-tumour immune responses remain unclear. Here, we use photoconvertible mice to precisely track DC migration. We report that CCR7+ DCs are the dominant DC population that migrate to the dLN, but a subset remains tumour-resident despite CCR7 expression. These tumour-retained CCR7+ DCs are phenotypically and transcriptionally distinct from their dLN counterparts and heterogeneous. Moreover, they progressively downregulate the expression of antigen presentation and pro-inflammatory transcripts with more prolonged tumour dwell-time. Tumour-residing CCR7+ DCs co-localise with PD-1+CD8+ T cells in human and murine solid tumours, and following anti-PD-L1 treatment, upregulate stimulatory molecules including OX40L, thereby augmenting anti-tumour cytolytic activity. Altogether, these data uncover previously unappreciated heterogeneity in CCR7+ DCs that may underpin a variable capacity to support intratumoural cytotoxic T cells.

PubMed Disclaimer

Conflict of interest statement

AstraZeneca provided therapeutic anti-PD-L1 antibodies, isotype control antibodies, and MC38-Ova cells. GC, SAH and SJD are full employees and shareholders in AstraZeneca. No other disclosures or conflicts of interest are reported.

Figures

Fig. 1
Fig. 1. Landscape and temporal dynamics of DCs in murine tumours.
a Experiment design. Tumours from Kaede transgenic mice were harvested 5, 24, 48, or 72 h after photoconversion. b UMAP of myeloid cells from scRNA-seq of FACS-sorted CD45+TER119- Kaede-green+/Kaede-red+ cells 48 h after photoconversion of subcutaneous MC38-Ova tumours. DC, dendritic cell; LC, Langerhans cell; Mac, macrophage; Mast, mast cell; pDC, plasmacytoid DC. c UMAP of DCs from (b) and canonical marker gene expression in DC clusters (d). e Kernel density embedding of DCs by Kaede fluorescence and treatment group. f, g Flow cytometry of Kaede fluorescence in DCs subsets/states from MC38-Ova tumours: f Representative plots 48 h post-photoconversion, and (g) Time-course and quantification. h Confocal microscopy of MC38 tumours 72 h post-photoconversion. Arrows point to tumour-residing CCR7+ DCs (Kaede-red+CCR7+MHC-II+, dendritic morphology). Scale bar, 40μm. i Tumour DCs ordered by pseudotime, rooted in the cluster with highest proportion of Kaede-green DCs, and proposed maturation trajectory. j RNA velocity trajectory in tumour DCs. k Expression of Axl. Paired two-sided student’s t-test was used, data are shown as means ± s.d., and points represent independent mice (g). The results shown in (g) are from one experiment (24 h/72 h n = 5; 48 h n = 4 animals), representative of three independent experiments; and (h) are representative of three independent experiments (n = 7 animals).
Fig. 2
Fig. 2. CCR7+ DCs that migrate to the dLN become phenotypically and transcriptionally distinct from tumour-residing populations.
ac Flow cytometry of draining lymph nodes (dLN) and contralateral non-draining lymph nodes (ndLN) of MC38-Ova tumours; DCs that originate from photo-flashed tumours (tumour emigrants) carry the Kaede-red fluorescent profile, which enables tracking to LNs. a Representative plots 48 h after tumour photoconversion, b Time-course and quantification of Kaede-red cells among CCR7+ DCs, and c composition of Kaede-red DCs in dLNs. d, e UMAP of myeloid cells from scRNA-seq of FACS-sorted CD45+ Kaede-red cells from tumour-dLNs (MC38-Ova) and CD45+ cells from control LNs. f Gene set enrichment analysis (GSEA) of Kaede-red CCR7+ DCs in dLNs versus tumours (isotype control-treated). Signed P-adj indicate log10(Benjamini-Hochberg adjusted P value) with the direction of enrichment. g Selected gene expression in scRNA-seq of tumour CCR7+ DCs (isotype control-treated) and Kaede-red CCR7+ DC tumour emigrants in the dLN. h Flow cytometry of CD80 and CD86 on CCR7+ DCs from MC38-Ova tumours or dLNs 48 h after photoconversion, with representative histograms. i Integration of scRNA-seq of Kaede-red DCs from tumour-dLNs with tumour DCs and label transfer. j Proportion of DC clusters by tissue and Kaede fluorescence from i. Paired two-sided student’s t-test (b, c), two-sided Wilcoxon rank-sum test with Benjamini-Hochberg multiple-testing correction (g), or one-way analysis of variance (ANOVA) and Šidák’s multiple comparisons test (h) were used. Points represent independent mice (b, c, h). Data are shown as means ± s.d. (b, c, h), or box (median; box, 25th and 75th percentile; whiskers, 1.5*inter-quartile range) and violin plots (g). The results shown in a–c are from one experiment (24 h/72 h n = 5; 48 h n = 4 animals), representative of three independent experiments; and h from two independent experiments (n = 8 animals).
Fig. 3
Fig. 3. Tumour-residing CCR7+ DCs undertake an “exhausted” state with duration in the tumour, attenuated by anti-PD-L1 treatment.
a Expression of MHC-II transcripts and Cd74 over pseudotime in tumour DCs. Local regression (loess) was fit to scaled expression values. b Gene signature scores for “GO:BP dendritic cell antigen processing and presentation (GO:0002468)” by CCR7+ DC cluster. c Scaled enrichment scores of scRNA-seq of all DCs for “GO:BP DC antigen processing and presentation” and CCR7+ DC signature genes, coloured by pseudotime. d GSEA of Ccr7_DC.3 versus Ccr7_DC.1. Only significant pathways (Benjamini-Hochberg-adjusted p values (P-adj) < 0.05) are shown. e Differential gene expression between Ccr7_DC.1 and Ccr7_DC.3. Significant differentially expressed genes (DEG, P-adj < 0.01) from “Hallmark inflammatory response” are highlighted in red. f Milo differential abundance analysis. Bee-swarm plot shows treatment-associated differences in overlapping cellular neighbourhoods (individual points). Differentially abundant neighbourhoods at FDR < 0.05 are coloured. ‘Mixed’ refers to neighbourhoods where cells do not predominantly (>70%) belong to a single cluster. g Differential gene expression between Ccr7_DC.2 and Ccr7_DC.3. DEGs (P-adj < 0.01, log2Fold-change > 0.5) are coloured. h Flow cytometry of OX40L expression (OX40L+/Hu-CD4 reporter mice) on CCR7+ DC from MC38-Ova tumours and their dLN. Two-sided Wilcoxon rank-sum test (b), two-sided Wald test with Benjamini-Hochberg multiple testing correction (e, g), or paired two-sided student’s t-test (h) were used. Data are shown as box (median; box, 25th and 75th percentile; whiskers, 1.5*inter-quartile range) and violin plots (b), or means ± s.d. (h). Points represent independent mice (h). The results shown in h are from one experiment (n = 5 animals), representative of two independent experiments.
Fig. 4
Fig. 4. CCR7+ DCs interact with CD8+ T cells in human tumours.
a Pearson correlation between cell proportions, from deconvolution of 521 bulk transcriptomes of colorectal adenocarcinoma biopsies (TCGA), CD274, and a CCR7+ DC gene signature. Box highlights the high correlation between CCR7+ DC and CD8+ T cell transcripts. Only significant correlations (p < 0.05) shown. b Pearson correlation between CCR7+ DC signature genes and effector CD8+ T cell signature genes in colorectal adenocarcinoma (COAD), breast cancer (BRCA), skin cutaneous melanoma (SKCM) and lung adenocarcinoma (LUAD) from TCGA. Points represent individual patient samples. c CellPhoneDB cell-cell communication analysis between myeloid cells and effector CD8+ T cells in scRNA-seq of human CRC (n = 62 patients). Edge width scaled to standardised interaction scores. Only significant interactions (p < 0.05) shown. Mono, monocyte; Mac, macrophage; Mast, mast cell; pDC, plasmacytoid DC; CD8_Teff, effector CD8+ T cell. Spatial correlation (Pearson R-value) of CCR7+ DC and effector CD8+ T cell signature scores in spatial transcriptomics (10X Genomics Visium) of independent human CRC tumour sections (d, n = 2), a human melanoma section (e, n = 1), and independent human breast tumour sections (f, n = 2). g Expression of selected genes in myeloid-T cell doublets from sequencing of physically interacting cells (PICseq) of non-small cell lung cancer (NSCLC) tumours (n = 10 patients), grouped by the myeloid cell identity in each myeloid-T cell doublet. Two-sided Pearson correlation was used (a-b). Data are shown as linear regression with 95% confidence interval (b).
Fig. 5
Fig. 5. anti-PD-L1 promotes activating CCR7+ DC-cytotoxic CD8+ T cell interactions in the TME.
a UMAP of scRNA-seq of CD8+ T cells, from FACS-sorted CD45+ tumour-infiltrating lymphocytes 48 h after photoconversion of subcutaneous MC38-Ova tumours. b Proportion of TEX_CD8T cells by Kaede fluorescence and treatment group. c Number of significant DEGs in anti-PD-L1 versus isotype control-treated tumours by cluster. DEGs calculated using Wilcoxon rank-sum test with Benjamini-Hochberg multiple-testing correction. d CellPhoneDB ligand-receptor analysis between tumour DCs and CD8+ TEX cells. Only significant interactions (p < 0.05) shown. e Representative confocal microscopy images of MC38 tumours, showing co-localisation of CCR7+MHC-II+ DCs and CD3+CD8+ 4-1BB+/Ki-67+ T cells (arrows). Scale bar, 20 μm. (fk) In vitro and ex vivo cultures. f Phenotype of bone marrow-derived DCs (BMDC) following culture with UV-irradiated MC38-Ova tumour cells. g Experiment set-up of DC:OT-I co-culture. h Representative flow cytometry of CD44+ OT-I cell activation and proliferation following co-culture with MC38-Ova-experienced CCR7+ BMDC; +/– anti-PD-L1, OX40L-expressing (+, OX40Lfl/fl) or OX40L-deficient (–, CD11ccreOX40Lfl/fl) BMDCs. i Quantification of h and granzyme B (GzmB) expression. Values shown are relative to cultures with isotype control antibodies and OX40L-expressing DCs (baseline), to facilitate comparisons across experiments. j Flow cytometry of CCR7+ DCs from subcutaneous MC38-Ova tumours following 8 h culture ex vivo with antibodies or recombinant IFNγ (rIFNg). k Flow cytometry of OT-I cells following co-culture with MC38-Ova-experienced CCR7+ BMDC; CCR7+ BMDCs pre-treated with rIFNg (+) or PBS (–), OX40L-expressing (+) or OX40L-deficient (–) BMDCs. One-way analysis of variance (ANOVA) and Šidák’s multiple comparisons test was used, data are shown as means ± s.d. (f, ik). The results shown in (e) are representative of two independent experiments (n = 5 animals); f from one experiment (n = 4 biological samples) representative of three independent experiments; h, i from two independent experiments (n = 7 biological samples); j from one experiment (n = 5 animals), representative of two independent experiments; and (k) from one experiment (n = 4 biological samples), representative of two independent experiments.
Fig. 6
Fig. 6. Conserved CCR7+ DC heterogeneity and CD8+ T cell crosstalk in human cancer.
a UMAP of scRNA-seq of CCR7+ DCs from human CRC (GSE178341, n = 62 patients). b Selected gene expression in CCR7+ DCs from tumour (T), normal adjacent tissue (N) and tumour-dLN from independent scRNA-seq data of human CRC (syn26844071, n = 63 patients). c UMAP of myeloid cells from scRNA-seq of human metastatic urothelial carcinoma (mUC, n = 11 patients). d single-cell gene expression (scGEX) profiles associated with responders (complete responder, CR; partial responder, PR) or non-responders (stable disease, SD; progressive disease, PD) in the IMvigor 210 trial (atezolizumab) for treatment of mUC; Scissor integration of bulk RNA-seq of tumour samples from IMvigor210 (n = 208 patients) and scRNA-seq from c. e Differential gene expression in myeloid cells associated with responders (CR/PR) versus non-responders (SD/PD) from d. f Gene signature scores for Ccr7_DC.2 transcripts in scRNA-seq of CCR7+ DCs from human breast cancer following treatment with anti-PD-1 antibodies; T cell clonotype expanders (E, i.e. responders, n = 9 patients) versus non-expanders (NE, i.e. non-responders, n = 20 patients). g PICseq (myeloid-T cell doublets) of human NSCLC; expression of CCR7+ DC-T cell ligand-receptor pairs, grouped by myeloid cell identity in each myeloid-T cell doublet (n = 10 patients). h Gene signature scores for “GO:BP T cell mediated cytotoxicity (GO:0001913)” (HLA genes removed), in myeloid-CD8+ T cell PICs grouped by the myeloid cell identity. i, j Spatial correlation (Pearson R-value) of CCR7+ DC signature scores and selected CCR7+ DC-ligand receptors expressed by CD8+ T cells, in spatial transcriptomics (10X Genomics Visium) of independent human CRC tumour sections (n = 2). Two-sided Wilcoxon rank-sum test with Benjamini-Hochberg multiple-testing correction (b, e), or two-sided Wilcoxon rank-sum test (f, h) were used. Data are shown as box (median; box, 25th and 75th percentile; whiskers, 1.5*inter-quartile range) and violin plots (b, f, h).

References

    1. Wculek SK, et al. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2019;20:7–24. doi: 10.1038/s41577-019-0210-z. - DOI - PubMed
    1. Roberts EW, et al. Critical role for CD103(+)/CD141(+) Dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer Cell. 2016;30:324–336. doi: 10.1016/j.ccell.2016.06.003. - DOI - PMC - PubMed
    1. Sallusto F, et al. Rapid and coordinated switch in chemokine receptor expression during dendritic cell maturation. Eur. J. Immunol. 1998;28:2760–2769. doi: 10.1002/(SICI)1521-4141(199809)28:09<2760::AID-IMMU2760>3.0.CO;2-N. - DOI - PubMed
    1. Böttcher JP, Reis e Sousa C. The role of Type 1 conventional dendritic cells in cancer immunity. Trends Cancer. 2018;4:784–792. doi: 10.1016/j.trecan.2018.09.001. - DOI - PMC - PubMed
    1. Maier B, et al. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature. 2020;580:257–262. doi: 10.1038/s41586-020-2134-y. - DOI - PMC - PubMed