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
. 2021 Feb;160(3):781-796.
doi: 10.1053/j.gastro.2020.10.036. Epub 2020 Oct 29.

PD-1 Signaling Promotes Tumor-Infiltrating Myeloid-Derived Suppressor Cells and Gastric Tumorigenesis in Mice

Affiliations

PD-1 Signaling Promotes Tumor-Infiltrating Myeloid-Derived Suppressor Cells and Gastric Tumorigenesis in Mice

Woosook Kim et al. Gastroenterology. 2021 Feb.

Abstract

Background & aims: Immune checkpoint inhibitors have limited efficacy in many tumors. We investigated mechanisms of tumor resistance to inhibitors of programmed cell death-1 (PDCD1, also called PD-1) in mice with gastric cancer, and the role of its ligand, PD-L1.

Methods: Gastrin-deficient mice were given N-methyl-N-nitrosourea (MNU) in drinking water along with Helicobacter felis to induce gastric tumor formation; we also performed studies with H/K-ATPase-hIL1B mice, which develop spontaneous gastric tumors at the antral-corpus junction and have parietal cells that constitutively secrete interleukin 1B. Mice were given injections of an antibody against PD-1 or an isotype control before tumors developed, or anti-PD-1 and 5-fluorouracil and oxaliplatin, or an antibody against lymphocyte antigen 6 complex locus G (also called Gr-1), which depletes myeloid-derived suppressor cells [MDSCs]), after tumors developed. We generated knock-in mice that express PD-L1 specifically in the gastric epithelium or myeloid lineage.

Results: When given to gastrin-deficient mice before tumors grew, anti-PD-1 significantly reduced tumor size and increased tumor infiltration by T cells. However, anti-PD-1 alone did not have significant effects on established tumors in these mice. Neither early nor late anti-PD-1 administration reduced tumor growth in the presence of MDSCs in H/K-ATPase-hIL-1β mice. The combination of 5-fluorouracil and oxaliplatin reduced MDSCs, increased numbers of intra-tumor CD8+ T cells, and increased the response of tumors to anti-PD-1; however, this resulted in increased tumor expression of PD-L1. Expression of PD-L1 by tumor or immune cells increased gastric tumorigenesis in mice given MNU. Mice with gastric epithelial cells that expressed PD-L1 did not develop spontaneous tumors, but they developed more and larger tumors after administration of MNU and H felis, with accumulation of MDSCs.

Conclusions: In mouse models of gastric cancer, 5-fluorouracil and oxaliplatin reduce numbers of MDSCs to increase the effects of anti-PD-1, which promotes tumor infiltration by CD8+ T cells. However, these chemotherapeutic agents also induce expression of PD-L1 by tumor cells. Expression of PD-L1 by gastric epithelial cells increases tumorigenesis in response to MNU and H felis, and accumulation of MDSCs, which promote tumor progression. The timing and site of PD-L1 expression is therefore important in gastric tumorigenesis and should be considered in design of therapeutic regimens.

Keywords: Immunosuppression; Mouse Model; Resistance; Stomach Cancer.

PubMed Disclaimer

Figures

Figure 1.
Figure 1.. Early anti-PD-1 treatment inhibits tumor growth in GAS-KO mice
(A) Expression of Pdl1 and Pdcd1 in the antrum following Hf/MNU by qPCR (n = 4/group). (B) E-cadherin and PD-L1 immunostaining on Hf/MNU/GAS-KO tumors at 30 weeks post-MNU. (C) Experimental design for early treatment. (D-E) Gross images (D) and tumor area measured (E) from GAS-KO mice treated with isotype control (n = 8) or anti-PD-1 (n = 6). Dotted lines indicate tumor area. (F) CD3 immunostaining on treated tumors. (G-H) The proportion of tumor-infiltrating CD3+ T cells (G) and the subsets (H) among CD45+ cells in treated mice (n = 3-4/group) by flow cytometry. (I) CD11b immunostaining on treated tumors. (J-K) The percentage of intratumoral MDSCs (CD11b+Gr-1+) (J), M-MDSCs (CD11b+Ly6ChiLy6G) and PMN-MDSCs (CD11b+Ly6CloLy6G+) (K) among CD45+ cells from treated mice (n = 3-4/group). (L) Immunosuppressive activity of PMN-MDSCs isolated from tumors (n = 3/group). Statistically significant differences from No MDSC group. (M-N) The proportion of macrophages (CD11b+Gr-1F4/80+) among CD45+ cells (M) and Tregs (CD25+Foxp3+) in CD4+ T cells (N) in treated tumors (n = 3-4/group). Scale bars, 100 μm (B); 5 mm (D); 50 μm (F and I). Mean ± SEM. one-way ANOVA (A); Student’s t-test (E, G-H and J-N). *P < .05; **P < .01; ***P < .001.
Figure 2.
Figure 2.. Early anti-PD-1 treatment in H/K-ATPase-IL-1β mice fails to delay tumor growth
(A) E-cadherin and PD-L1 immunostaining on Hf/MNU/IL-1β tumors at 30 weeks post-MNU. (B) PD-L1+ cells in EpCAM+ and CD45+ cells isolated from 30-week IL-1β tumors by flow cytometry (n = 3). (C-D) Gross images (C) and tumor area measured (D) from control (n = 9) or anti-PD-1-treated (n = 14) IL-1β mice. Dotted lines indicate tumor area. (E) Gr-1 immunostaining on tumors from WT and treated IL-1β mice. (F-G) Contour plots showing tumor MDSCs (F) and quantification (G) from treated groups (n = 6-8/group). (H-J) The proportion of MDSC subsets (H), T cells (I-J) in treated tumors (n = 6-8/group). (K) CD8 immunostaining on treated tumors. Scale bars, 100 μm (A); 5 mm (C); 50 μm (E and K). Mean ± SEM. Student’s t-test. *P < .05; **P < .01; ****P < .0001.
Figure 3.
Figure 3.. Late treatment with anti-PD-1 is effective when administered in combination with chemotherapy
(A) Experimental scheme for late treatment. (B-C) Gross images (B) and tumor area measured (C) from GAS-KO mice in indicated treatment groups (n = 12-17/group). Dotted lines indicate tumor area. One-way ANOVA (C). (D) H&E stains of GAS-KO tumors in different treatment groups. Dashed boxes indicate magnified areas shown at the bottom. (E-G) Immunostaining for cleaved caspase-3 (E), Ki-67 (F) and β-catenin (G) on treated GAS-KO tumors and quantification (n = 3/group). Arrowheads indicate nuclear β-catenin (G). Student’s t-test. Scale bars, 5 mm (B); 100 μm (D-G). Mean ± SEM. *P < .05; **P < .01; ***P < .001; ****P < .0001.
Figure 4.
Figure 4.. Immune effects of combination chemo-immunotherapy
(A) The proportion of intratumoral T cells in treated GAS-KO tumors (n = 8-13/group). (B) CD8 immunostaining on treated tumors. (C) Frequency of effector cytokine-secreting CD8+ T cells (n = 3/group). (D) CD11b immunostaining on treated tumors. (E) The proportion of MDSCs in treated GAS-KO tumors (n = 8-13/group). (F) Linear regression analyses between tumor area and the percentage of CD8+ T cells and PMN-MDSCs in GAS-KO tumors (n = 44). r2 and P-values are shown. (G) MFI (mean fluorescence intensity) of PD-L1 expressed on EpCAM+ cells isolated from GAS-KO tumors treated as indicated (n = 3-5/group). Scale bars, 50 μm. Mean ± SEM. Student’s t-test. *P < .05; **P < .01; ***P < .001.
Figure 5.
Figure 5.. PD-L1 overexpression in gastric epithelial cells promotes gastric tumorigenesis
(A) Gene construct of R26-LSL-Pdl1-IRES-EGFP mice. (B) Endogenous GFP expression in the stomach. The targeted tissues by Cre-expressing mice are gastric epithelial cells (Tff2-Cre) and immune/myeloid cells (LysM-Cre). (C-D) Gross images (C) and tumor area measured (D) from R26-PD-L1 and Tff2-Cre; R26-PD-L1 mice at 36 weeks post-MNU +/− Hf. Dotted lines indicate tumor area (n = 6-11/group). (E) H&E stains of gastric tumors. Dashed lines indicate intramucosal well-differentiated adenocarcinoma. An arrowhead indicates submucosal immune cell infiltrates. (F) Ki-67 and β-catenin staining on gastric tissues and quantification (n = 3/group). (G-H) The proportion of MDSCs by flow cytometry (G) and Gr-1 immunostaining (H) on tumors (n = 5-6/group). (I) The percentage of intratumoral MDSC subsets (n = 5-6/group). (J-K) mRNA expression of chemokines in tumors (J, n = 4/group) and isolated EpCAM+ cells from tumors (K, n = 3/group) by qPCR. (L) The percentage of tumor-infiltrating T cells (n = 5-6/group). (M) The percentage of cytokine secreting-intratumoral CD8+ T cells (n = 3/group). Scale bars, 100 μm (B and E-F); 5 mm (C); 50 μm (H). Mean ± SEM. One-way ANOVA (D); Student’s t-test (F, G and I-M). *P < .05; **P < .01; ***P < .001; n.s., not significant.
Figure 6.
Figure 6.. PD-L1-overexpressing myeloid cells facilitate gastric tumorigenesis
(A-B) Gross images (A) and tumor area measured (B) from R26-PD-L1 and LysM-Cre; R26-PD-L1 mice at 36 weeks post-MNU +/− Hf (n = 5-12/group). Dotted lines indicate tumor area. Scale bars, 5 mm (A). (C-D) The percentage of tumor-infiltrating MDSCs and T cells (n = 4-9/group). (E-F) The percentage of Ki-67+ (E), effector cytokine-positive cells (F) in CD8+ T cells isolated from MNU-induced R26-PD-L1 and LysM-Cre; R26-PD-L1 tumors (n = 3/group). Mean ± SEM. one-way ANOVA (B); Student’s t-test (C-F). *P < .05; **P < .01; ***P < .001; n.s., not significant.
Figure 7.
Figure 7.. Epithelial-derived PD-L1 increases susceptibility to gastric carcinogenesis
(A) Experimental design to assess susceptibility to MNU-induced gastric tumorigenesis. (B-D) Gross images (B), tumor incidence rates (C), and tumor area measured (D) in R26-PD-L1 and Tff2-Cre; R26-PD-L1 mice (n = 10-11/group) at 24 weeks post-MNU. (E-F) Gross images (E) and tumor area measured (F) from R26-PD-L1 and LysM-Cre; R26-PD-L1 mice at 24 weeks (n = 5/group). (G) Pdl1 expression in antral tissues at 10 weeks by qPCR (n = 3/group). (H-I) The proportion of T cells (H) and MDSCs (I) in antral tissues from R26-PD-L1 and Tff2-Cre; R26-PD-L1 mice at various time points following MNU induction (n = 3-5/group). Scale bars, 5 mm (B and E). Mean ± SEM. Student’s t-test. *P < .05; **P < .01; ****p < .0001.

Comment in

References

    1. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394–424. - PubMed
    1. Howlader N, Noone AM, Krapcho M, et al. SEER Cancer Statistics Review, 1975-2016, National Cancer Institute; Bethesda, MD, https://seer.cancer.gov/csr/1975_2016/, based on November 2018 SEER data submission, posted to the SEER web site, April 2019. 2019.
    1. Smyth EC, Verheij M, Allum W, et al. Gastric cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2016;27:v38–v49. - PubMed
    1. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443–54. - PMC - PubMed
    1. Fashoyin-Aje L, Donoghue M, Chen H, et al. FDA Approval Summary: Pembrolizumab for Recurrent Locally Advanced or Metastatic Gastric or Gastroesophageal Junction Adenocarcinoma Expressing PD-L1. Oncologist 2019;24:103–109. - PMC - PubMed

Publication types

MeSH terms