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 Jun;25(6):1033-1045.
doi: 10.1038/s41590-024-01828-7. Epub 2024 May 14.

The aged tumor microenvironment limits T cell control of cancer

Affiliations

The aged tumor microenvironment limits T cell control of cancer

Alex C Y Chen et al. Nat Immunol. 2024 Jun.

Abstract

The etiology and effect of age-related immune dysfunction in cancer is not completely understood. Here we show that limited priming of CD8+ T cells in the aged tumor microenvironment (TME) outweighs cell-intrinsic defects in limiting tumor control. Increased tumor growth in aging is associated with reduced CD8+ T cell infiltration and function. Transfer of T cells from young mice does not restore tumor control in aged mice owing to rapid induction of T cell dysfunction. Cell-extrinsic signals in the aged TME drive a tumor-infiltrating age-associated dysfunctional (TTAD) cell state that is functionally, transcriptionally and epigenetically distinct from canonical T cell exhaustion. Altered natural killer cell-dendritic cell-CD8+ T cell cross-talk in aged tumors impairs T cell priming by conventional type 1 dendritic cells and promotes TTAD cell formation. Aged mice are thereby unable to benefit from therapeutic tumor vaccination. Critically, myeloid-targeted therapy to reinvigorate conventional type 1 dendritic cells can improve tumor control and restore CD8+ T cell immunity in aging.

PubMed Disclaimer

Figures

Extended Data Fig. 1 ∣
Extended Data Fig. 1 ∣. Phenotyping of CD8+ T cells in young vs. aged tumors.
Related to Fig. 1. (a) Representative flow cytometry plots (left) and frequencies of splenic CD8+ T cells that were either naive (PD-1CD44; right-top), activated (PD-1+CD44+; right-center), or antigen experienced (PD-1+Tim-3+; right-bottom) cells in mice that ranged in 5-85 weeks of age. (b) Representative flow cytometry gating strategy utilized to quantify tumor-infiltrating CD8+ T cells as well as tetramer (Tet+) specific, and BrdU+ CD8+ T cells in young and aged tumor-bearing mice. (c) Representative flow cytometry gating strategy used to measure lymphatic CD8+ T cells as well as tetramer (Tet+) specific CD8+ T cells in young and aged tumor-bearing mice. (d) ScRNA-seq UMAP projection of 5862 aged tumor-infiltrating CD8+ T cells defined in Fig. 1d. (e) Corresponding violin plots of scRNA-seq that denote RNA read counts (nFeature_RNA; left), and the percentage of mitochondrial genes per cell (% mitochondrial reads; right) for each cluster. (f) Hashtag of scRNA-seq profiles reveals distinct biological replicates, denoted by color, in young (left) and aged (right) tumor-bearing mice. (g) Expression of indicated genes (grey: low, purple: high). (h) Heatmap shows top cluster-defining genes in each CD8+ T cell cluster/subset identified in Fig. 1d. (i) Quantification of subsets within live tetramer (Tet+) antigen-experienced (CD44+ PD-1+) CD8+ T cells, classified based on the Tim-3 and Slamf6 expression in young (blue, n = 8) and aged (red, n = 4) tumor-bearing mice. (j) Geometric mean fluorescence intensity (MFI) of IL-2, IFN-γ, TNF, Gzmb, and BrdU in live antigen-experienced (CD44+ PD-1+) tumor-infiltrating CD8+ T cells from young (blue; n = 7) and aged (red; n = 4) tumor-bearing mice, separated by subsets. For panels a, i and j: Summary of at least two independent experiments. Mean ± s.d. For i and j, significance was calculated using a two-sided Student’s t-test. Asterisks used to indicate significance corresponds to the following: N.S. (not significant, P > 0.05), *P ≤ 0.05, **P ≤ 0.01, and ***P ≤ 0.001).
Extended Data Fig. 2 ∣
Extended Data Fig. 2 ∣. Transcriptional profiling of T cell-intrinsic defects in aging.
Related to Fig. 2. (a) Schematic of single and co-transfers OT-1 CD8+ T cells into young tumor bearing recipient mice and the corresponding representative flow cytometry gating strategy utilized to quantify endogenous and transferred OT-1 CD8+ T cells in young tumor bearing recipient mice. (b) Schematic of young and aged splenic OT-1 CD8+ T cell co-transfer into young recipient mice experiments. scRNA-seq UMAP projection of 7731 tumor-infiltrating OT-1 CD8+ T cells from co-transfer of young and aged splenic OT-1 CD8+ T cell into young recipient mice, colored by cluster. Expression of indicated genes (grey: low expression, purple: high expression). (c) Violin plots that denote RNA read counts (nFeature_RNA; top), total number of genes in each cell (nCount_RNA; top second), and the percentage of mitochondrial genes (% mitochondrial reads; top third) and ribosomal genes (% ribosomal reads; bottom) per cell. (d) Heatmap showing top cluster-defining genes in each CD8+ T cell cluster/subset identified in Fig. 2b. (e) Relative proportion of young (OT-1Y>Y, n = 5) and aged (OT-1A>Y, n = 5) CD8+ T cells within progenitor-like exhausted (TProg), terminally exhausted (TTerm), and dividing (TDivi) clusters. (f) Experimental design for in vivo transfer assay to study TProg formation and maintenance at early time points, such as days 7 and 9, between OT-1Y>Y (blue) and OT-1A>Y (red) in young B16-OVA tumor-bearing mice on days 7, 9, and 15. Proportion of TProg, TTran, and TTerm between OT-1Y>Y and OT-1A>Y on days 7, 9, and 15 (n = 9-10). (g) UMAP projection of scRNA-seq profiles from 6231 live splenic OT-1 CD8+ T cells from young and aged mice, colored by cluster (C). C1, young naive OT-1 CD8+ T cells (Young Spl TNaiv); C2, aged naive OT-1 CD8+ T cells (Aged Spl TNaiv); C3, aged memory-like OT-1 CD8+ T cells (Aged Spl TMem-like); C4, aged effector-like OT-1 CD8+ T cells (Aged Spl TEff-like); C5, young effector-like OT-1 CD8+ T cells (young Spl TEff-like); C6, aged exhausted-like OT-1 CD8+ T cells (Aged Spl TExh). Expression of indicated genes (grey: low expression, purple: high expression). i: Tcf7, ii: Pdcd1, iii: Ccr7, iv: Sell, v: Tox. (h) Heatmap showing top cluster-defining genes in each of splenic OT-1 CD8+ T cell cluster/subset identified in Extended Data Fig. 2e, with a specific focus of the top 7 genes in cluster 2 (C2). (i) UMAP projection of scRNA-seq profiles of young splenic OT-1 (Spl OT-1Y), aged splenic OT-1 PD-1 (Spl OT-1A), aged splenic OT-1 PD-1+ (Spl OT-1A PD-1+), and wild-type young splenic (WTY) CD8+ T cells. Frequencies of each sample within different cell clusters. Mean ± s.d. For e and f, significance was calculated using a two-sided Student’s t-test. Asterisks used to indicate significance corresponds to the following: N.S. (not significant, P > 0.05), *P ≤ 0.05, **P ≤ 0.01, and ***P ≤ 0.001).
Extended Data Fig. 3 ∣
Extended Data Fig. 3 ∣. Epigenetic profiling of aged T cell-intrinsic defects at baseline.
Related to Fig. 3. (a) Transcriptional start site (TSS) enrichment of chromatin accessibility profiles from aged and young pre-transfer splenic as well as tumor-infiltrating OT-1 CD8+ T cell subsets. (b) Representative ATAC-seq tracks of aged (yellow) and young (green) splenic naive OT-1 CD8+ T cells at the indicated gene loci. (c) Pathway enrichment in splenic young OT-1 CD8+ T cells (Spl OT-1Y, green) and splenic aged OT-1 CD8+ T cells (Spl OT-1A, yellow) chromatin accessible regions (ChARs). The FDR values (hypergeometric test) are presented as −log10(q-value).
Extended Data Fig. 4 ∣
Extended Data Fig. 4 ∣. Cell-extrinsic effects of aging on differentiation and function of CD8+ T cells.
Related to Fig. 4, part 1. (a) Experimental design and tumor growth curves for B16-OVA tumors in young (10w; blue; n = 17) and aged (68w; red; n = 16) recipient mice treated with young spleen OT-1 CD8+ T cells (Spl OT-1Y). (b) Frequency of transferred OT-1 CD8+ T cells isolated from young (10w, blue; n = 17) and aged (68w; red; n = 16) recipient B16-OVA tumor-bearing mice. (c) B16-OVA tumor volumes at day 15 from the cell-intrinsic model (left, young mice receiving either young or aged splenic OT-1), and cell-extrinsic model (right, young or aged mice received young spleen OT-1). (d) Experimental design for in vitro killing assay and percentage of cytotoxicity of sorted young (blue) and aged (red) OVA tetramer+ (Tet+) intratumoral CD8+ T cell subsets at 1:1 effector to target (E:T; OT-1: B16-OVA) ratio (n = 4-6). (e) Experimental design for in vivo transfer and T cell differentiation. (f) Schematic of young and aged intratumoral Tet+ CD8+ T cell subsets transfer into young recipient mice experiments. The corresponding representative flow cytometry gating strategy is utilized to quantify transferred Tet+ CD8+ T cell subsets in young tumor-bearing recipient mice. (g) Frequency of transferred OT-1 CD8+ T cells differentiated into different CD8+ T cell subsets (n = 4-5). Mean ± s.d. For b, d, and g, significance was calculated using a two-sided Student’s t-test. Asterisks used to indicate significance corresponds to the following: N.S. (not significant, P > 0.05), *P ≤ 0.05, **P ≤ 0.01, and ***P ≤ 0.001).
Extended Data Fig. 5 ∣
Extended Data Fig. 5 ∣. Epigenetic and transcriptional profiling of T cell-extrinsic defects in aged tumors.
Related to Fig. 4, part 2. (a) ScRNA-seq UMAP projection of 4263 tumor-infiltrating OT-1 CD8+ T cells from transfer of young splenic OT-1 CD8+ T cell into young or aged recipient tumor-bearing mice, colored by cluster. Expression of indicated genes (grey: low, purple: high). (b) Heatmap showing top cluster-defining genes in each CD8+ T cell cluster/subset identified in Fig. 4e. (c) Violin plots that denote RNA read counts (nFeature_RNA; top), total number of genes in each cell (nCount_RNA; top second), and the percentage of mitochondrial genes (% mitochondrial reads; bottom) per cell, related to Fig. 4e, f. (d) Volcano plot of differentially expressed genes between transferred young OT-1 (OT-1Y) subsets: progenitor-like exhausted (TProg; brown) and terminally exhausted (TTerm; blue) as well as the differentially expressed genes between TTerm (blue) and tumor-infiltrating age-associated dysfunctional (TTAD; red) OT-1 CD8+ T cells. (e) Heatmap showing Z-scored TF regulon activity within cell-type clusters identified by Seurat. (f) Principal component analysis (PCA) projection and transcriptional start site (TSS) enrichment of chromatin accessibility profiles from tumor-infiltrating CD8+ T cell subsets in young and aged tumor-bearing mice. (g) Violin plots that denote RNA read counts (nFeature_RNA; top), total number of genes in each cell (nCount_RNA; top second), related to Fig. 4j-l. (h) Heatmap showing top cluster-defining genes in each CD8+ T cell cluster/subset identified in Fig. 4j. (i) Volcano plot of differentially expressed genes between human tumor-infiltrating T cell subsets: TTerm vs. TTAD CD8+ T cells.
Extended Data Fig. 6 ∣
Extended Data Fig. 6 ∣. Phenotyping of immune cells in young vs. aged tumors.
Related to Fig. 5. (a) Representative flow cytometry gating strategy (top) utilized to quantify intratumoral CD45+ cells in young and aged tumor-bearing mice, including conventional CD4+ T cells, CD8+ T cells, natural killer (NK) cells, and subsets of dendritic cells (DCs), macrophages (Mac), Ly6C+ monocytes (Mono), and conventional dendritic cells type1 (cDC1s) and 2 (cDC2s) within DCs. Representative flow cytometry gating strategy (bottom) utilized to lymphatic CD8+ T cells, tetramer (Tet+) specific CD8+ T cells, subsets of dendritic cells (DCs), and cDC1/cDC2 within DCs in young and aged tumor-bearing mice. (b) Heatmap shows top cluster-defining genes in each cell cluster/subset identified in Fig. 5c. (c) Volcano plot of differentially expressed genes between intratumoral T, NK, and myeloid cells from young (blue) and aged (red) tumor-bearing mice. (d) All the significant ligand-receptor interactions between NK, cDC1, and CD8+ T cells in young (blue) and aged (red) tumor-bearing mice. The dot color and size represent the calculated communication probability and p-values. (e) Percentage of intratumoral CD8+ T cells and OVA tetramer+ (Tet+) intratumoral CD8+ T cells from B16-OVA tumor-bearing mice with different ages, from 10w, 24w, 44w, up to 68w old (n = 6-8). (f) Percentage of intratumoral CD8+ T cell subsets (such as TTerm and TTAD) from B16-OVA tumor-bearing mice with different ages, from 10w, 24w, 44w, up to 68w old (n = 6-8). Mean ± s.d. For b, d, e, f, and g, significance was calculated using a two-sided Student’s t-test. Asterisks used to indicate significance corresponds to the following: N.S. (not significant, P > 0.05), *P ≤ 0.05, **P ≤ 0.01, and ***P ≤ 0.001).
Extended Data Fig. 7 ∣
Extended Data Fig. 7 ∣. Transcriptional profiling of CD8+ T cells after myeloid-targeted immunotherapy.
Related to Fig. 6. (a) ScRNA-seq UMAP projection of 3454 tumor-infiltrating CD8+ T cells defined in Fig. 6h. (b) Expression of indicated genes (grey: low, purple: high). (c) Corresponding violin plots of scRNA-seq that denote RNA read counts (nFeature_RNA; up), and the percentage of mitochondrial genes per cell (% mitochondrial reads; down) for each cluster. (d) Heatmap shows top cluster-defining genes in each CD8+ T cell cluster/subset identified in Fig. 6h. (e) ScRNA-seq UMAP projection of different tumor-infiltrating CD8+ T cell cluster proportion from untreated young/aged, and young/aged treated with CD40 agonist antibody mice. (f) Quantification of naive (TNaiv), transitory exhausted (TTran), IFN-reponse (TIFN), dividing (TDivi), and Lars2+ (TLars2+) subsets based on scRNA with individual mouse hashtags (n = 5). (g) Volcano plot of differentially expressed genes between transferred terminally exhausted (TTerm; brown) and different subsets, including tumor-infiltrating age-associated dysfunctional (TTAD; red), progenitor-like exhausted (TProg; golden yellow), memory-like (TMem-like), and effector-like (TEff-like) subsets. (h) Dot plot of different expression levels of critical surface markers and transcriptional regulators among each CD8+ T cell cluster. Mean ± s.d. For f, significance was calculated using a two-sided Student’s t-test. Asterisks used to indicate significance corresponds to the following: N.S. (not significant, P > 0.05), *P ≤ 0.05, **P ≤ 0.01, and ***P ≤ 0.001).
Fig. 1 ∣
Fig. 1 ∣. Aging promotes tumor growth and alters CD8+ T cell fate and effector function.
a, Schema of the experimental design for the B16-OVA melanoma model used to assess tumor growth in mice of the indicated ages; D0, day 0; D15, day 15. b, Tumor growth curves for B16-OVA and LLC-OVA tumors in young (10 weeks (w); blue; n = 17 and n = 8) and aged (68 weeks; red; n = 12 and n = 4) mice. c, Percentage of day 15 total tumor-infiltrating CD8+ T cells as well as tetramer-specific (Tet+) CD8+ T cells isolated from B16-OVA and LLC-OVA tumors, tdLNs and non-draining LNs (ndLN) from young (10 weeks; blue; n = 20) and aged (68 weeks; red; n = 12) mice. d, Uniform manifold approximation and projection (UMAP) of scRNA-seq profiles from 5,862 live tumor-infiltrating CD8+ T cells from day 15 B16-OVA tumor-bearing young and aged mice colored by cluster. Expression profiles of the indicated genes are shown on the right (gray, low expression; purple, high expression). e, Galaxy plots depicting the cell density in UMAP space for tumor-infiltrating CD8+ T cells from young (left) and aged (right) tumor-bearing mice. Cooler colors indicate low density, and warmer colors indicate high density. The relative proportion of CD8+ T cells from young (blue, n = 5) and aged (red, n = 5) tumors within TProg, TTerm, dividing T (TDivi) and TTAD cell clusters is shown. f, Representative flow plots of exhausted subsets in mice that were 10 weeks (young; blue; n = 19), 68 weeks (aged; red; n = 8) and over 90 weeks old (aged; purple; n = 2 groups; each biological replicate group contains three individual mice) bearing either B16-OVA or LLC-OVA tumors for 15 days. g, Quantification of TProg, TTerm, TDivi and TTAD cell subsets within live antigen-experienced (CD44+PD-1+) CD8+ T cells based on TIM-3 and SLAMF6 expression. h, Geometric mean fluorescence intensity (MFI) of IL-2, IFNγ, TNF, GZMB and BrdU in live antigen-experienced (CD44+PD-1+) tumor-infiltrating CD8+ T cells from young (blue; n = 16) and aged (red; n = 10) tumor-bearing mice. For b, c and fh, the data represent a summary of at least two independent experiments and are shown as mean ± s.d. For b, c and eh, significance was calculated using a two-sided Student’s t-test; NS, not significant (P > 0.05); *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001.
Fig. 2 ∣
Fig. 2 ∣. CD8+ T cell-intrinsic defects in aging lead to premature terminal differentiation and impaired proliferation.
a,b, Schema for in vivo co-transfer experiment (a) and scRNA-seq UMAP (b, left) of 7,731 live tumor-infiltrating OT-1 CD8+ T cells from day 15 B16-OVA tumor-bearing young mice colored by cluster. Expression of the indicated genes is shown (gray, low; purple, high). Density plots (b, right) in UMAP space for young T cells transferred into young mice (OT-1Y>Y, top) and aged T cells transferred into young (OT-1A>Y, bottom) mice are shown. Cooler colors indicate low density, and warmer colors indicate high density. c, Ratio of TTerm cells to TProg cells between OT-1Y>Y and OT-1A>Y T cells in scRNA-seq data. d, Volcano plot of differentially expressed genes between TTerm OT-1Y>Y and OT-1A>Y cells. e, Schema for the in vivo transfer assay to study TProg formation and maintenance at the indicated time points; i.v., intravenous. f, Representative flow plots of subset proportions within OT-1Y>Y (blue) and OT-1A>Y (red) T cells on the indicated days. g, Ratio of TTerm cells to TProg cells between OT-1Y>Y and OT-1A>Y on the indicated days (n = 9). h, scRNA-seq UMAP projection of 6,231 live splenic (Spl) OT-1 CD8+ T cells from young (10 weeks; blue; n = 5) and aged (68 weeks; red; n = 5) TNaiv cells colored by cluster. i, Volcano plot of differentially expressed genes between young and aged splenic OT-1 CD8+ TNaiv cells. j, Correlation of genes between aged and young splenic OT-1 CD8+ TNaiv cells (before transfer to young tumor-bearing mice) and between aged and young tumor-infiltrating TTerm OT-1 cells (after transfer). k, Preranked GSEA of the indicated gene signatures between aged and young splenic OT-1 TNaiv cells. l, Schema for in vitro killing assay and percent cytotoxicity of splenic aged (n = 4) and young (n = 4) OT-1 CD8+ T cells at different effector-to-target (E:T) ratios. m, Schema for the in vivo transfer assay to study CD8+ T cell-intrinsic responses. Young mice (n = 32) received either young (n = 63; blue) or aged (n = 43; red) OT-1 CD8+ T cells before B16-OVA tumor challenge. The data in l and m are derived from at least two independent experiments and are shown as mean ± s.d. For c, g, l and m, the significance was calculated using a two-sided Student’s t-test; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001.
Fig. 3 ∣
Fig. 3 ∣. Cell-intrinsic alternations in aged CD8+ T cells underlies low proliferation but not the formation of the TTAD subset.
a, Principal component analysis (PCA) projection and transcription start site enrichment of chromatin accessibility profiles from aged and young pretransfer splenic and tumor-infiltrating OT-1 CD8+ T cell subsets. b, Representative assay for transposase-accessible chromatin with sequencing (ATAC-seq) tracks of aged (yellow) and young (green) splenic OT-1 CD8+ TNaiv cells (Spl TNaiv) at the indicated gene loci. c, Pathway enrichment in TTerm OT-1Y>Y (purple) and TTerm OT-1A>Y (pink) chromatin-accessible regions (ChARs). The false discovery rate (FDR) values (hypergeometric test) are presented in −log10 (q value); eNOS, endothelial nitric oxide synthase. d, Representative flow cytometry plots and geometric MFI of γH2AX (left) and p21 (center) after 1–3 h of UV exposure in young and aged splenic OT-1 CD8+ T cells; FMO, fluorescence minus one. e, Frequency of BrdU+CD8+ T cells derived from the endogenous model (Endo.; top; young (n = 8) and aged (n = 4) mice without OT-1 T cell transfer), cell-intrinsic model (center; young mice that received either young (n = 5) or aged splenic OT-1 T cells (n = 4)) and cell-extrinsic model (bottom; young (n = 8) or aged mice (n = 7) that received young splenic OT-1 T cells). f, Representative flow cytometry plots of SLAMF6/TIM-3 gating and frequency of endogenous or transferred TTerm CD8+ T cells in young (10 weeks; blue; n = 14) or aged (68 weeks; red; n = 14) recipient mice 15 days after tumor inoculation. g, Representative flow cytometry plots of SLAMF6/TIM-3 gating and frequency of endogenous or transferred age-associated TTAD cells in young (10 weeks; blue; n = 14) or aged (68 weeks; red; n = 14) recipient mice 15 days after tumor inoculation. For dg, data are representative of at least two independent experiments and are shown as mean ± s.d. For eg, significance was calculated using a two-sided Student’s t-test. For c, enriched ChARs between each group were identified using GREAT analysis; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001.
Fig. 4 ∣
Fig. 4 ∣. Cell-extrinsic signals from the aged TME drive the TTAD subset state, which is distinct from CD8+ T cell exhaustion.
a, Schema and growth curves for B16-OVA tumors in young (10 weeks; blue; n = 17) and aged (68 weeks; red; n = 16) mice that received young splenic OT-1 CD8+ T cells (Spl OT-1Y). b, Geometric MFI of IL-2, IFNγ, TNF, GZMB and γH2AX within transferred young OT-1 TTerm (red) and TTAD (red dot) cell subsets in aged recipient mice (n = 7). c, In vitro killing assay and percent cytotoxicity of sorted young (blue) and aged (red) OT-1 intratumoral CD8+ T cell subsets at an effector:target ratio of 0.5:1 (n = 5). d, Schema for in vivo tumor control by transferring different OVA Tet+CD8+ T cell subsets from young (blue) and aged tumors (red) to young B16-OVA tumor-bearing mice (n = 4–6); w/o, without. e, UMAP projection of scRNA-seq profiles from 4,263 young OT-1 CD8+ T cells transferred into young and aged mice colored by cluster. Expression of the indicated genes is shown on the right (gray, low; purple, high). f, Density plots in UMAP space of young (left, OT-1Y>Y) and aged (right, OT-1Y>A) groups. Cooler colors represent low density, and warmer colors represent high density. Relative proportions of young (OT-1Y>Y, n = 5) and aged (OT-1Y>A, n = 5) CD8+ T cells within TProg, TTerm, TDivi and TTAD cell clusters are shown. g, Preranked GSEA enrichment of the indicated gene signatures within transferred OT-1 CD8+ TTerm and TTAD cells. h, Hypergeometric enrichment of effector, exhausted and memory core programs within ChARs differentially open in CD8+ TTerm and TTAD cells. i, Superenhancer plots based on ChARs differentially open in CD8+ TTerm and TTAD cells. The red dot indicates the superenhancer cutoff. j, scRNA-seq UMAP projection of 7,242 tumor-infiltrating CD8+ T cells from human pan-cancer aggregated tumor biopsies (n = 57) colored by cluster. Expression of the indicated genes (gray, low; red, high) is shown on the right; Hu., human; Mel, melanoma; LC, lung cancer; BC, breast cancer; CRC, colorectal cancer; OC, ovarian cancer. k, Density plots in UMAP space of CD8+ T cells from individuals with cancer at various age ranges. Cooler colors indicate low density, and warmer colors indicate high density. l, GSEA enrichment of mouse TTAD and TTerm cell signatures within a human intratumoral CD8+ T cell UMAP projection. The dark blue color indicates low enrichment, and the yellow-green color indicates high enrichment. The data in ad are representative of at least two independent experiments and are shown as mean ± s.d. For ad and f, the significance was calculated using a two-sided Student’s t-test; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001.
Fig. 5 ∣
Fig. 5 ∣. The aged TME erodes NK cell–cDC1–CD8+ T cell cross-talk and impairs CD8+ T cell priming and expansion.
a, Schema to assess intratumoral CD45+ cells from young (blue) and aged (red) tumor-bearing mice. b, Frequency of live intratumoral CD45+ cells in young and aged mice challenged with B16-OVA (young (blue), n = 15; aged (red), n = 8) and LLC-OVA (young (blue), n = 8; aged (red), n = 4) tumors. c, scRNA-seq UMAP projection of 2,660 intratumoral T cells, NK cells and myeloid cells subclustered from CD45+ cells in young and aged tumor-bearing mice colored by cluster. Expression of the indicated genes is shown on the bottom (gray, low; purple, high). Broad cell types were designated as follows: macrophages (Mac; clusters (C) 2, 3 and 5), NK cells (cluster 4), CD8+ T cells (clusters 1, 7 and 9), DCs (clusters 6, 8 and 10), CD4+ T cells (cluster 11) and monocytes (Mono; cluster 12). d, Volcano plot of differentially expressed genes between young (blue) and aged (red) intratumoral DCs (clusters 6, 8 and 10). e, scRNA-seq CellChat differential number of interactions between macrophages, CD4+ T cells (CD4), monocytes, type 2 cDCs (cDC2), migratory DCs (migDC), NK cells, cDC1s and CD8+ T cells (CD8) and relative value within each cell–cell interaction between young (blue) and aged (red) cells. f, Percentage of cDC subsets from B16-OVA tumor-bearing mice of the indicated ages (n = 6–10). g, Geometric MFI of CD40, CD86, FLT3, MHC class I and MHC class II in intratumoral cDC1s from day 15 tumors of tumor-bearing mice of the indicated ages (n = 6–10). h, Representative flow cytometry plots of cDC subsets from tumors (left), tdLNs (center) and non-draining LNs (right) in young (blue; top) and aged (red; bottom) mice. i, Percentage of cDC subsets from B16-OVA (top; n = 14) and LLC-OVA (bottom; n = 14) tumor-bearing young (blue; n = 10) and aged (red; n = 8) mice. j, Schema of potential CD8+ T cell-intrinsic and CD8+ T cell-extrinsic mechanisms of age-related immune dysfunction with progressive aging or in the aged TME, respectively. The data in b and fi are representative of at least two independent experiments and are shown as mean ± s.d. For b, f, g and i, the significance was calculated using a two-sided Student’s t-test; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001.
Fig. 6 ∣
Fig. 6 ∣. Limited CD8+ T cell priming and expansion by cDC1s in the aged TME can be rescued by myeloid-targeted immunotherapy.
a, Schema and growth curves after OVA-mRNA LNP vaccination in young (10 weeks; n = 10) and aged (68 weeks; n = 8) B16-OVA tumor-bearing mice; tx, treatment. b, Day 15 tumor volumes from a. c, Frequencies of cDC1s (first and second columns) and CD8+ T cells (third and fourth columns) within live CD45+ cells in tumor and tdLNs. The frequencies of indicated cells from OVA-mRNA LNP-treated (young tumor (top), n = 16; young tdLN (top), n = 8; aged tumor (bottom), n = 12; aged tdLN (bottom), n = 7) or nontreated (young tumor (top), n = 16; young tdLN (top), n = 8; aged tumor (bottom), n = 8; aged tdLN (bottom), n = 4) tumor-bearing mice are shown. d, Schema and growth curves after treatment with CD40 agonist antibody in young (10 weeks; blue; n = 16) and aged (68 weeks; red; n = 7) B16-OVA tumor-bearing mice. e, Day 15 tumor volumes from d. f, Frequencies of cDC1s (first and second columns) and CD8+ T cells (third and fourth columns) within live CD45+ cells in tumor and tdLNs. The frequencies of indicated cells from CD40 agonist antibody-treated (young tumor (top), n = 8; young tdLN (top), n = 8; aged tumor (bottom), n = 7; aged tdLN (bottom), n = 7) or nontreated (young tumor (top), n = 8; young tdLN (top), n = 8; aged tumor (bottom), n = 7; aged tdLN (bottom), n = 4) tumor-bearing mice are shown. g, Geometric MFI of CD40, CD86, FLT3, MHC class I and MHC class II in intratumoral cDC1 cells from day 15 tumors of young (10 weeks (Y)) and aged (68 weeks (A)) tumor-bearing mice with or without CD40 agonist antibody treatment. h, scRNA-seq UMAP projection of 3,454 live tumor-infiltrating CD8+ T cells from day 15 B16-OVA tumor-bearing young and aged mice with or without CD40 agonist antibody treatment colored by cluster. Expression of the indicated genes is shown along the bottom (gray, low; purple, high). i, Density plots in UMAP space for tumor-infiltrating CD8+ T cells from young (left top), young treated with CD40 agonist antibody (left bottom), aged (right top) and aged treated with CD40 agonist antibody (right bottom) tumor-bearing mice. Cooler colors indicate low density, and warmer colors indicate high density. j, Quantification of TProg, TTerm, TTAD, TMem-like and TEff-like cell subsets. Data in ag are representative of at least two independent experiments and are shown as mean ± s.d. For the data in ag and j, the significance was calculated using a two-sided Student’s t-test; *P ≤ 0.05; **P ≤ 0.01; ***P ≤ 0.001.

References

    1. Berben L, Floris G, Wildiers H & Hatse S Cancer and aging: two tightly interconnected biological processes. Cancers 13, 1400 (2021). - PMC - PubMed
    1. Tsukamoto H, Senju S, Matsumura K, Swain SL & Nishimura Y IL-6-mediated environmental conditioning of defective TH1 differentiation dampens antitumour immune responses in old age. Nat. Commun 6, 6702 (2015). - PMC - PubMed
    1. Sade-Feldman M et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013 (2018). - PMC - PubMed
    1. Schreiber RD, Old LJ & Smyth MJ Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science 331, 1565–1570 (2011). - PubMed
    1. Goronzy JJ, Fang F, Cavanagh MM, Qi Q & Weyand CM Naive T cell maintenance and function in human aging. J. Immunol 194, 4073–4080 (2015). - PMC - PubMed