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 Sep 3;12(9):e009236.
doi: 10.1136/jitc-2024-009236.

Targeting IL-33 reprograms the tumor microenvironment and potentiates antitumor response to anti-PD-L1 immunotherapy

Affiliations

Targeting IL-33 reprograms the tumor microenvironment and potentiates antitumor response to anti-PD-L1 immunotherapy

Yanyang Nan et al. J Immunother Cancer. .

Erratum in

Abstract

Background: The main challenge against patients with cancer to derive benefits from immune checkpoint inhibitors targeting PD-1/PD-L1 appears to be the immunosuppressive tumor microenvironment (TME), in which IL-33/ST2 signal fulfills critical functions. However, whether IL-33 limits the therapeutic efficacy of anti-PD-L1 remains uncertain.

Methods: Molecular mechanisms of IL-33/ST2 signal on anti-PD-L1 treatment lewis lung carcinoma tumor model were assessed by RNA-seq, ELISA, WB and immunofluorescence (IF). A sST2-Fc fusion protein was constructed for targeting IL-33 and combined with anti-PD-L1 antibody for immunotherapy in colon and lung tumor models. On this basis, bifunctional fusion proteins were generated for PD-L1-targeted blocking of IL-33 in tumors. The underlying mechanisms of dual targeting of IL-33 and PD-L1 revealed by RNA-seq, scRNA-seq, FACS, IF and WB.

Results: After anti-PD-L1 administration, tumor-infiltrating ST2+ regulatory T cells (Tregs) were elevated. Blocking IL-33/ST2 signal with sST2-Fc fusion protein potentiated antitumor efficacy of PD-L1 antibody by enhancing T cell responses in tumor models. Bifunctional fusion protein anti-PD-L1-sST2 exhibited enhanced antitumor efficacy compared with combination therapy, not only inhibited tumor progression and extended the survival, but also provided long-term protective antitumor immunity. Mechanistically, the superior antitumor activity of targeting IL-33 and PD-L1 originated from reducing immunosuppressive factors, such as Tregs and exhausted CD8+ T cells while increasing tumor-infiltrating cytotoxic T lymphocyte cells.

Conclusions: In this study, we demonstrated that IL-33/ST2 was involved in the immunosuppression mechanism of PD-L1 antibody therapy, and blockade by sST2-Fc or anti-PD-L1-sST2 could remodel the inflammatory TME and induce potent antitumor effect, highlighting the potential therapeutic strategies for the tumor treatment by simultaneously targeting IL-33 and PD-L1.

Keywords: Immune Checkpoint Inhibitor; Immune modulatory; Immunotherapy; T regulatory cell - Treg; Tumor microenvironment - TME.

PubMed Disclaimer

Conflict of interest statement

Competing interests: None declared.

Figures

Figure 1
Figure 1. IL-33/ST2 signal upregulated after anti-PD-L1 treatment. Volcano plots generated from analyzing differential gene expression between control and anti-PD-L1 treatment group (A). IL1RL1 gene level in tumor tissues of LLC tumor-bearing mouse injected with anti-PD-L1 (n=3) (B). ST2 protein level in tumor tissues was analyzed by WB (C). The quantification of ST2 expression in tumor tissues during anti-PD-L1 using ImageJ software (n=4) (D). IL-33 tumor tissue expression was analyzed by ELISA (n=4) (E). The expression of Myd88 and activation of JAK2/STAT3 pathway in tumor tissues treated with anti-PD-L1 (F). The gene correlation among CD274 and IL1RL1 in LUAD was analyzed using GEPIA (G). Representative images of ST2 (green) and FOXP3 (red) labeled in the LLC tumor tissues after anti-PD-L1 treatment (H). Scale bar=100 µm. White arrows point to ST2+FOXP3+ cells. *P < 0.05, **P < 0.01. GEPIA, Gene Expression Profiling Interactive Analysis; LLC, lewis lung carcinoma; LUAD, lung adenocarcinoma.
Figure 2
Figure 2. Dual targeting of PD-L1 and IL-33 enhanced antitumor effect of anti-PD-L1 therapy in mouse models. Treatment scheme (top), tumor growth curves (bottom left), and tumor weight (bottom right) of CT26 (A) or LLC (B) tumor-bearing mice in four groups (n=5). Schematic diagram of treatment protocol (left) and survival curves (right) of metastasis CT26 tumor model (n=5) (C). Scheme (left), H&E-stained lung tissues together with their representative photos (middle), and the quantity of tumor burden (right) from each group with metastasis LLC tumor (n=5) (D). *P < 0.05, **P < 0.01. LLC, lewis lung carcinoma.
Figure 3
Figure 3. Combined blockade of IL-33 and PD-L1 enhanced T cells response. Bulk RNA-seq to evaluate immune landscape in LLC tumors (A–E). Gene expression levels of Prf1, Ifng, and Gzmb in tumor tissue of LLC tumor-bearing mice (n=3) (A). The data were presented in Log2TPM and every group was compared with the combination therapy group. Heat map of genes that under the category of immune signatures (n=3) (B–D). Gene set enrichment analysis enrichment plot of KEGG pathway for genes in Combo-treated tumors, relative to anti-PD-L1-treated tumors (E). Flow cytometry was used to analyze the absolute number of CD4+ (F), CD8+ (G), CD8+GranB+ (H) and CD8+IFN-γ+ (I) cells and CD4+FOXP3+ cells (J) in CT26 tumor tissues (n=3). *P < 0.05, **P < 0.01. LLC, lewis lung carcinoma.
Figure 4
Figure 4. Construction and characterization of bifunctional fusion proteins targeting both PD-L1 and IL-33. Structure of bifunctional fusion proteins (A). Atezolizumab and sST2 were used to targeting PD-L1 and IL-33, respectively, and were fused by a flexible (Gly4Ser)3Gly linker. The purity determination of anti-PD-L1-sST2 (B) and sST2-anti-PD-L1 (C). Affinity of bifunctional fusion proteins for hPD-L1 (D) or mIL-33 (E) proteins measured by SPR. PD-1/PD-L1 blockade bioassay of bifunctional fusion proteins was confirmed (n=2) (F). Naïve CD4+ T cells were incubated with anti-CD3/CD28 and TGF-β1, subjected to the specified treatments for 3 days to determine the frequencies of FOXP3+ T cells (n=3) (G). Effects of bifunctional fusion proteins on LPS-induced production of IL-6 (H) and TNF-α (I) from Raw264.7 in vitro (n=3). * and $ represent the Student’s t-test of each groups compared with control group and IL-33 group, respectively. *P < 0.05, **P < 0.01, $P < 0.05.
Figure 5
Figure 5. Antitumor activity of bifunctional fusion proteins in murine LLC models. Tumor growth (A) and tumor weight (B) of LLC tumor-bearing mice therapied with bifunctional fusion proteins (11.5 mg/kg) or anti-PD-L1 (7.5 mg/kg) and sST2-Fc (6.3 mg/kg) combined therapy (n=5). Schematic overview of the experimental study (C). Mice were intravenously injected with LLC cells (2*105), treated at day 0, 4, 7, and 11 after injection (red arrows) and observed until day 100 (n=8). Then, the survived mice were rechallenged with injection of 5*105 LLC cells and observed for three additional weeks. Survival curves of tumor-bearing mice in five groups (D). H&E-stained lung tissues of mice in rechallenge assay (E). *P < 0.05, **P < 0.01. LLC, lewis lung carcinoma.
Figure 6
Figure 6. Dual targeting of PD-L1 and IL-33 changed T-cell cellular landscapes. Design of scRNA-seq experiment (A). UMAP of all cell types from four groups (B). UMAP plots showing all CD4+ (C) and CD8+ (E) T cells from tumor tissues in four groups. Bar plots exhibiting the proportion of CD4+ (D) and CD8+ (F) T cells within each cluster. Selected significantly enriched pathways in KEGG analyses based on the differentially expressed genes in T cell clusters (G). Gene Ontology in biological function enrichment analysis of the upregulated (H, J) or downregulated (I, K) expressed genes in T cells of anti-PD-L1-sST2 versus atezolizumab group, and anti-PD-L1-sST2 versus combination group, respectively. LLC, lewis lung carcinoma.
Figure 7
Figure 7. Dual targeting of PD-L1 and IL-33 reprograms immuno-inflammatory phenotype TME. Absolute number of CD8+ cells (A), CD3+CD8+IFN-γ+ T cells (B), CD8+CD44+ cells (C), CD8+TCF-1+ cells (D), CD8+TIM-3+ cells (E) within the CD45+ cells from tumor tissues (n=3). The quantification of collagen (n=3) (F). The quantification of CD4+FOXP3+ cells in LLC tumor-bearing mice (n=5) (G). White arrows point to CD4+FOXP3+ cells. The quantification of α-SMA (H), Vimentin (I), TGF-β1 (J), and E-cadherin (K) (n=4). In figure 7G–K, data in groups of control, sST2-Fc, anti-PD-L1, anti-PD-L1 + sST2 Fc, and anti-PD-L1-sST2 are shown in color of blank, blue, green, red, and violet, respectively. Schematic diagram illustrating the potent antitumor impact of anti-PD-L1-sST2 bifunctional fusion protein (l). *P < 0.05, **P < 0.01. LLC, lewis lung carcinoma; TME, tumor microenvironment.

References

    1. van Gulijk M, van Krimpen A, Schetters S, et al. PD-L1 checkpoint blockade promotes regulatory T cell activity that underlies therapy resistance. Sci Immunol. 2023;8:eabn6173. doi: 10.1126/sciimmunol.abn6173. - DOI - PubMed
    1. Wang R, Liu H, He P, et al. Inhibition of PCSK9 enhances the antitumor effect of PD-1 inhibitor in colorectal cancer by promoting the infiltration of CD8+ T cells and the exclusion of Treg cells. Front Immunol. 2022;13:947756. doi: 10.3389/fimmu.2022.947756. - DOI - PMC - PubMed
    1. Wang Y, Zhang X, Xu C, et al. Targeting 4-1BB and PD-L1 induces potent and durable antitumor immunity in B-cell lymphoma. Front Immunol. 2022;13:1004475. doi: 10.3389/fimmu.2022.1004475. - DOI - PMC - PubMed
    1. Abril-Rodriguez G, Torrejon DY, Liu W, et al. PAK4 inhibition improves PD-1 blockade immunotherapy. Nat Cancer. 2020;1:46–58. doi: 10.1038/s43018-019-0003-0. - DOI - PMC - PubMed
    1. Vesely MD, 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

LinkOut - more resources