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 Feb 20;5(2):101420.
doi: 10.1016/j.xcrm.2024.101420.

PD-L1-expressing tumor-associated macrophages are immunostimulatory and associate with good clinical outcome in human breast cancer

Affiliations

PD-L1-expressing tumor-associated macrophages are immunostimulatory and associate with good clinical outcome in human breast cancer

Lei Wang et al. Cell Rep Med. .

Abstract

Tumor-associated macrophages (TAMs) are the predominant cells that express programmed cell death ligand 1 (PD-L1) within human tumors in addition to cancer cells, and PD-L1+ TAMs are generally thought to be immunosuppressive within the tumor immune microenvironment (TIME). Using single-cell transcriptomic and spatial multiplex immunofluorescence analyses, we show that PD-L1+ TAMs are mature and immunostimulatory with spatial preference to T cells. In contrast, PD-L1- TAMs are immunosuppressive and spatially co-localize with cancer cells. Either higher density of PD-L1+ TAMs alone or ratio of PD-L1+/PD-L1- TAMs correlate with favorable clinical outcome in two independent cohorts of patients with breast cancer. Mechanistically, we show that PD-L1 is upregulated during the monocyte-to-macrophage maturation and differentiation process and does not require external IFN-γ stimulus. Functionally, PD-L1+ TAMs are more mature/activated and promote CD8+ T cells proliferation and cytotoxic capacity. Together, our findings reveal insights into the immunological significance of PD-L1 within the TIME.

PubMed Disclaimer

Conflict of interest statement

Declaration of interests The authors declare no competing interests.

Figures

None
Graphical abstract
Figure 1
Figure 1
Expression profile differences between PD-L1+/hi vs. PD-L1–/lo TAMs from human breast tumors revealed by scRNA-seq (A) scRNA-seq analysis of TAMs (n = 2,220 cells) from untreated primary breast tumor (n = 5 patients, ER+) shown as a UMAP, highlighting identified clusters. (B) UMAP showing mutually exclusive expression of PD-L1 and SIGLEC15 in TAMs. (C) Dichotomization of TAM clusters into PD-L1+/hi and PD-L1–/lo subpopulations. (D) PD-L1 expression of PD-L1+/hi and PD-L1–/lo subpopulations. (E) Representative flow cytometry plot showing PD-L1+% TAMs, peripheral monocytes, and intratumoral T cells from the same tumor tissue used for scRNA-seq (left panel), and PD-L1+% quantification was compared between analysis via scRNA-seq and flow cytometry (right panel). (F) Overlay of the expression of common M1 and M2 signature genes in the PD-L1+/hi vs. PD-L1–/lo TAM dichotomization. (G) Volcano plot showing differentially expressed genes (DEGs) between PD-L1+/hi vs. PD-L1–/lo TAMs. (H and I) Expression distribution of selected genes involved in maturation, pro-inflammatory or transcriptional activator (H), and anti-inflammatory, pro-tumor, fatty acid metabolic, or extracellular matrix (I) between PD-L1+/hi and PD-L1–/lo TAMs. ∗∗∗∗p < 0.0001. Wilcoxon rank-sum test. (J) PD-L1+/hi and PD-L1–/lo TAMs were flow sorted from freshly prepared single-cell suspension of digested breast tumor (n = 4) and supernatants were collected for ELISA after 16 h. Paired t test. ∗p < 0.05, ∗∗p < 0.01.
Figure 2
Figure 2
Expression profile of PD-L1+/− TAMs from patients with TNBC revealed by published scRNA-seq data (A) Published (Pal et al.27) scRNA-seq transcriptomic clustering of TAMs (n = 4,484 cells) from untreated primary TNBC breast tumors shown as a UMAP. (B) Mutually exclusive expression of PD-L1 and SIGLEC15 in TAMs. (C) Dichotomization of TAM clusters into PD-L1+/hi and PD-L1–/lo subpopulations. (D) PD-L1 expression of PD-L1+/hi and PD-L1–/lo subpopulations. (E) Volcano plot showing differentially expressed genes (DEGs) between PD-L1+/hi vs. PD-L1−/lo TAMs. (F) Expression distribution of selected genes involved in maturation, pro-inflammatory, anti-inflammatory, and pro-tumor between PD-L1+/hi and PD-L1–/lo TAMs. (G) Published (Bassez et al.28) scRNA-seq transcriptomic clustering and dichotomization of TAMs (n = 12,952 cells) from patients with TNBC treated with neoadjuvant anti-PD1 immunotherapy into PD-L1+/hi and PD-L1–/lo subpopulations. (H) PD-L1 expression of PD-L1+/hi and PD-L1–/lo subpopulations. ∗∗∗∗p < 0.0001. Wilcoxon rank-sum test. (I) TAMs were annotated based on whether patients exhibited clonal expansion of intratumoral PD1+ T cells after the anti-PD1 treatment as expanded or non-expanded. (J) The percentages of PD-L1+/hi TAMs were compared between tumors with anti-PD1-induced expanded vs. non-expanded clonal PD1+ T cells. ∗∗p < 0.01. Mann-Whitney test.
Figure 3
Figure 3
PD-L1+ TAMs are associated with favorable clinical outcome in two independent cohorts of patients with BC (A and B) Kaplan-Meier relapse-free survival (RFS) curves and log rank test generated for the gene signature of PD-L1+ vs. PD-L1 TAMs or the gene signature ratio of PD-L1+/PD-L1 TAMs in the luminal BC cohorts of METABRIC (n = 1098) (A) and TCGA (n = 789) (B) datasets. (C) Kaplan-Meier analyses for the M1, M2, or the ratio of M1/M2 gene signature in the luminal BC cohorts of METABRIC (n = 1098). Patients were divided into high- and low-expressing groups based on a 25% cutoff of the gene signature. (D) Schematic summarizing the histological quantification method in cohorts 1 and 2 of patients with luminal BC. (E) Representative immunofluorescence staining of PD-L1, CD68, and DAPI to identify PD-L1+ and PD-L1 TAMs. (F–H) Using Kaplan-Meier estimate and log-rank test, relapse-free survival (RFS) was compared between patients with low and high density of PD-L1+ TAMs in cohort 1 (n = 49) (F) and in cohort 2 (n = 93) (G) or low and high density ratio of PD-L1+/PD-L1 TAMs in combined cohorts 1 and 2 (n = 142) (H). Median density was used as the cutoff to divide patients into low vs. high groups. (I) Univariate and multivariate analysis for the prognostic significance of the density ratio of PD-L1+/PD-L1 TAMs. Hazard ratio calculated with below medium vs. above medium (n = 142).
Figure 4
Figure 4
PD-L1+ and PD-L1 TAMs have different cell-to-cell interaction preferences (A) Multiple immunofluorescence staining and corresponding phenotype map of representative breast tumor tissue section for PD-L1+ TAMs (CD68+PD-L1+), PD-L1 TAMs (CD68+PD-L1), CD8+ T cells (CD8+), CD4+ T cells (CD3+CD8), and cancer cells (CK+). (B) Whole-slide quantification of the ratio of PD-L1+ TAMs/cancer cells in total area. (C) Schematic representing the calculation of cell-cell interaction based on CD8+ T cells, CD4+ T cells, or cancer cells within a radius of 20 μm from the nuclei of PD-L1+ or PD-L1 TAMs. (D) CD8+ T cells, CD4+ T cells, or cancer cells within a radius of 20 μm from the nuclei of PD-L1+ or PD-L1 TAMs in untreated primary luminal breast tumors (n = 36). ∗∗∗∗p < 0.0001, ∗∗p < 0.01. Wilcoxon paired test. (E) The number of TAMs within 20 μm to PD-L1+ TAMs (left) or PD-L1 TAMs (right) (n = 36). Paired t test. ∗∗∗∗p < 0.0001. (F) Dot plots of ligand-receptor interactions between PD-L1+ or PD-L1 TAMs and CD4/8+ T cells (left panel), and cancer cells (right panel) based on our scRNA-seq transcriptomic analysis. Gray dot represents no significant interaction was found.
Figure 5
Figure 5
PD-L1 is upregulated during the monocyte-macrophage maturation/differentiation process (A and B) Freshly isolated PBMCs from newly diagnosed patients with BC (n = 12) were ex vivo rested in RPMI 1640 with 10% FBS for 8 h. PD-L1+% peripheral blood monocytes are shown in representative flow plots (A) and compared between fresh vs. rested monocytes (B). (C and D) PD-L1+% between in suspension vs. adherent monocytes after 8 h resting (n = 8) are shown in representative flow plots (C) and compared (D). (E and F) PD-L1+% monocytes between flow sorted fresh PBMCs vs. 8 h rested PBMCs (n = 5) are shown in representative flow plots (E) and compared (F). Paired t test. (G and H) MFI ratio of surface protein levels (G) and phosphorylated signal transduction protein levels (H) on PD-L1+ vs. PD-L1 monocytes in 8 h rested PBMCs from patients with BC. Wilcoxon paired test. (I) Peripheral blood monocytes from patients with BC (n = 6) were treated with small-molecule inhibitors (Selleck) against ERK1/2 (SCH772984 at 0. 5 μM), STAT1 (fludarabine at 50 μM), Akt1/2/3 (MK-2206 2HCl at 0.5 μM), PI3Kα/δ/β (LY294002 at 5 μM), NF-κB (QNZ at 5 μM), and mTOR (rapamycin at 0.1 μM) during the 8 h resting. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗p < 0.0001. Shown are mean ± SEM.
Figure 6
Figure 6
PD-L1 upregulation in TAMs could be IFN-γ independent (A–C) PBMCs from patients with BC were rested and stimulated with IFN-γ. Representative flow plots showing IFN-γ (50 ng/mL for 15 min) induced phosphorylation of STAT1 (pY701) in peripheral monocytes (A) from patients with BC. IFN-γ signaling response (n = 28) (B) and levels of IFN-γR1 (n = 20) (C) were compared between PD-L1–/lo vs. PD-L1+/hi monocytes. (D and E) Rested PBMCs from patients with BC were stimulated with IFN-γ at 0.2, 1, 5, or 25 ng/mL for 16 h. Levels of PD-L1 and IFN-γR1 on monocytes or T cells are shown in the representative flow plots (D) and compared (n = 6) (E). One-way ANOVA. (F–H) Single-cell suspensions from freshly prepared primary breast tumors were stimulated with IFN-γ. Representative flow plots (F) showing IFN-γ-induced pSTAT1 (G) and levels of IFN-γR1 (H) between PD-L1–/lo vs. PD-L1+/hi TAMs from untreated primary breast tumors (n = 8). (I and J) Multiplex immunofluorescence staining (I) and quantification of IFN-γR1+ PD-L1+/− cells (J) from untreated primary breast tumor tissues (n = 8). Scale bars, 100 μm. (K–M) Representative flow plots showing PD-L1 expression (K) and IFN-γ-induced pSTAT1 (L) and levels of IFN-γR1 (M) between PD-L1–/lo vs. PD-L1+/hi breast cancer cells (n = 8). Paired t test. ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001. Shown are mean ± SEM.
Figure 7
Figure 7
PD-L1+ TAMs are more activated and pro-inflammatory than PD-L1 TAMs (A and B) PBMCs from patients with BC (n = 16) were rested and the phagocytosis capacity of monocytes/macrophages were determined by using pHrodo Green E. Coli Bioparticles conjugate as shown in the representative flow plots (A) and compared between PD-L1–/lo vs. PD-L1+/hi monocytes/macrophages (B). Paired t test. (C and D) Freshly isolated PBMCs from patients with BC (n = 6) were rested and the PD-L1+/− monocytes were flow sorted. CellTrace Violet dilution by CD8+ T cells determined after 4 days of TCR-stimulated coculture with autologous PD-L1+ vs. PD-L1 monocytes/macrophages. (C) Representative flow plots showing percentage of proliferated CD8+ T cells. (D) Proliferation stimulation activity measured by cell number ratio of (CD8/CD4+CD14)/(CD8/CD4) as the stimulatory index. (E–H) Freshly isolated PBMCs from patients with BC (n = 6) were rested and the PD-L1+/− monocytes were flow sorted. Cytotoxic activity of CD8+ T cells determined using CD19/CD3 bispecific antibody (BiTE) after 2 days of coculture with CD19+ K562 cancer cells in the presence of autologous PD-L1+ or PD-L1 monocytes/macrophages. (E) Schematic representing the experiment setup. (F) Representative flow plots showing PD1 and CD137 expression on CD8+ T cells. (H) Cytotoxic activity calculated by the percentage of K562 cells killed by CD8+ T cells as shown in representative flow plots (G). One-way ANOVA. ∗p < 0.05, ∗∗∗∗p < 0.0001. Shown are mean ± SEM.

References

    1. Ma R.Y., Black A., Qian B.Z. Macrophage diversity in cancer revisited in the era of single-cell omics. Trends Immunol. 2022;43:546–563. doi: 10.1016/j.it.2022.04.008. - DOI - PubMed
    1. Mantovani A., Allavena P., Marchesi F., Garlanda C. Macrophages as tools and targets in cancer therapy. Nat. Rev. Drug Discov. 2022;21:799–820. doi: 10.1038/s41573-022-00520-5. - DOI - PMC - PubMed
    1. Vesely M.D., Zhang T., Chen L. Resistance Mechanisms to Anti-PD Cancer Immunotherapy. Annu. Rev. Immunol. 2022;40:45–74. doi: 10.1146/annurev-immunol-070621-030155. - DOI - PubMed
    1. Liu Y., Zugazagoitia J., Ahmed F.S., Henick B.S., Gettinger S.N., Herbst R.S., Schalper K.A., Rimm D.L. Immune Cell PD-L1 Colocalizes with Macrophages and Is Associated with Outcome in PD-1 Pathway Blockade Therapy. Clin. Cancer Res. 2020;26:970–977. doi: 10.1158/1078-0432.Ccr-19-1040. - DOI - PMC - PubMed
    1. Lau J., Cheung J., Navarro A., Lianoglou S., Haley B., Totpal K., Sanders L., Koeppen H., Caplazi P., McBride J., et al. Tumour and host cell PD-L1 is required to mediate suppression of anti-tumour immunity in mice. Nat. Commun. 2017;8 doi: 10.1038/ncomms14572. - DOI - PMC - PubMed

Publication types

Associated data