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
. 2018 Feb 1;128(2):580-588.
doi: 10.1172/JCI96061. Epub 2018 Jan 16.

PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression

Affiliations

PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression

Haidong Tang et al. J Clin Invest. .

Abstract

Programmed death-ligand 1 (PD-L1) expression on tumor cells is essential for T cell impairment, and PD-L1 blockade therapy has shown unprecedented durable responses in several clinical studies. Although higher expression of PD-L1 on tumor cells is associated with a better immune response after Ab blockade, some PD-L1-negative patients also respond to this therapy. In the current study, we explored whether PD-L1 on tumor or host cells was essential for anti-PD-L1-mediated therapy in 2 different murine tumor models. Using real-time imaging in whole tumor tissues, we found that anti-PD-L1 Ab accumulates in tumor tissues, regardless of the status of PD-L1 expression on tumor cells. We further observed that, while PD-L1 on tumor cells was largely dispensable for the response to checkpoint blockade, PD-L1 in host myeloid cells was essential for this response. Additionally, PD-L1 signaling in defined antigen-presenting cells (APCs) negatively regulated and inhibited T cell activation. PD-L1 blockade inside tumors was not sufficient to mediate regression, as limiting T cell trafficking reduced the efficacy of the blockade. Together, these findings demonstrate that PD-L1 expressed in APCs, rather than on tumor cells, plays an essential role in checkpoint blockade therapy, providing an insight into the mechanisms of this therapy.

Keywords: Cancer immunotherapy; Cellular immune response; Immunology; Oncology.

PubMed Disclaimer

Conflict of interest statement

Conflict of interest: The authors have declared that no conflict of interest exists.

Figures

Figure 1
Figure 1. Tumor-expressed PD-L1 is dispensable for responses to checkpoint blockade therapy.
(A) C57BL/6 mice were inoculated with 1 × 106 MC38 cells. Spleen, dLN, and tumor tissues were collected on day 22. PD-L1 expression was measured by flow cytometry. FMO, fluorescence minus one. (BD) Mean fluorescent intensities (MFIs) of PD-L1 staining in spleen (B), dLN (C), and tumor (D) are shown (n = 3 per group). (E) PD-L1 expression in MC38.WT, MC38.PD-L1–/–, A20.WT, and A20.PD-L1–/– cells was measured by flow cytometry. To induce PD-L1 expression, cells were treated with 500 U/ml IFN-γ for 24 hours. (F and G) C57BL/6 mice (n = 5 or 6) were inoculated with 1 × 106 MC38.WT or MC38.PD-L1–/– cells. After tumors were established, mice were treated with 200 μg anti–PD-L1 on days 7, 10, and 13. Tumor growth (F) and survival curve (G) are shown. (H and I) BALB/c mice (n = 5) were inoculated with 3 × 106 A20.WT or A20.PD-L1–/– cells. Mice were treated with 200 μg anti–PD-L1 on days 10 and 13. Tumor growth (H) and survival curve (I) are shown. (JL) Tissues were collected from MC38.PD-L1–/– tumor-bearing mice. Mean fluorescent intensities of PD-L1 staining in spleen (J), dLN (K), and tumor (L) are shown (n = 3). Data indicate mean ± SEM and are representative of at least 2 independent experiments. Statistical analysis was performed using an unpaired Student’s 2-tailed t test.
Figure 2
Figure 2. Anti–PD-L1 Ab targeting to tumor tissue is independent of tumor PD-L1.
(A) MC38.WT or MC38.PD-L1–/– tumor-bearing mice were injected with 50 μCi of 89Zr-radiolabeled deferoxamine-conjugated anti–PD-L1 (89Zr-anti–PD-L1) Abs (n = 3 per group). Ab distribution was imaged by PET/CT on 1, 2, 3, and 6 d.p.i. One representative mouse from each group is shown. Yellow arrows indicate tumors. (B) The uptake of 89Zr–anti–PD-L1 Abs in MC38 tumors was measured by PET/CT and quantitated in various organs on 1, 2, 3, and 6 d.p.i. (C) Ex vivo biodistribution of 89Zr–anti–PD-L1 Ab uptake on 6 d.p.i. is shown. (D) The uptake of 89Zr–anti–PD-L1 Abs was measured and quantitated by PET/CT in mice bearing A20.WT or A20.PD-L1–/– tumors (n = 3 per group). Statistical analysis was performed using an unpaired Student’s 2-tailed t test.
Figure 3
Figure 3. Preexisting TIL is insufficient for the effects of PD-L1 blockade.
(A) B6.Rag–/– mice (n = 5) were inoculated with 1 × 106 MC38 cells. After tumors were established, mice were treated with 200 μg anti–PD-L1 on days 8 and 11. Tumor growth was measured twice a week. (B) C57BL/6 mice (n = 5) were inoculated with 1 × 106 MC38 cells. Mice were treated with 200 μg anti–PD-L1 on days 8 and 11. For CD8+ T cell depletion, mice were treated with 200 μg anti-CD8 on days 8, 11, and 14. (CE) MC38 tumor–bearing mice (n = 5 per group) were treated with 200 μg IgG or anti–PD-L1 on days 8 and 11. Mice were also treated with control (C) or FTY720 from day 0 (D) or day 8 (E). (F) MC38 tumor-bearing mice were treated with IgG or anti–PD-L1 (n = 3 per group). Two days later, dLN were isolated and single-cell suspensions were prepared. Cells were cocultured with or without MC38 cells for 2 days. IFN-γ+ cells were measured by ELISPOT. (G) MC38 tumor–bearing mice were treated with anti–PD-L1 and FTY720 as in CE. Three days after anti–PD-L1 treatment, dLNs were isolated. ELISPOT assay was performed. Data indicate mean ± SEM and are representative of 2 independent experiments. Statistical analysis was performed using an unpaired Student’s 2-tailed t test. *P < 0.05; **P < 0.01; ***P < 0.001. ND, not detectable.
Figure 4
Figure 4. Host PD-L1 is essential for the responses to PD-L1 blockade.
(A) PD-L1–/– mice were inoculated with 1 × 106 WT MC38 cells. Mice were treated with 200 μg IgG or anti–PD-L1 on days 9 and 12 (n = 4). Tumor growth was measured twice weekly. (B and C) C57BL/6 mice were reconstituted with bone marrow cells from WT (B) or PD-L1–/– (C) mice. Eight weeks after reconstitution, mice were inoculated with 1 × 106 MC38 cells and treated with 200 μg anti–PD-L1 on days 7, 10, and 13 (n = 7 for WT, n = 5 for PD-L1–/–). Data indicate mean ± SEM and are from a pool of 2 independent experiments. Statistical analysis was performed using an unpaired Student’s 2-tailed t test. **P < 0.01.
Figure 5
Figure 5. PD-L1 signaling in myeloid cells harnesses antitumor immunity.
(A) C57BL/6 mice (n = 5) were inoculated with 1 × 106 WT MC38 cells and treated with 200 μg IgG or anti–PD-L1 on days 9 and 12. For cell depletion, 300 μg IgG, anti-CSF1R, or anti–Gr-1 Abs were injected from day 8. (BD) C57BL/6 mice were reconstituted with mixed bone marrow cells from CD11b-DTR and PD-L1–/– mice. Eight weeks after reconstitution, mice were inoculated with 1 × 106 MC38 cells and treated with 200 μg anti–PD-L1 treatment on days 11 and 14 (n = 5). DT was injected intraperitoneally every other day from day 11. Twenty-four hours after the second DT injection, PD-L1 levels in tumor-infiltrating-CD11b+ cells were measured by flow cytometry (B). Tumor growth without (C) or with (D) DT was measured twice a week. (E and F) BMDMs from WT or PD-L1–/– mice were loaded with SIY peptide, then cocultured with 2C T cells for 3 days. (E) IFN-γ levels in culture supernatant were measured by CBA. (F) T cell activation was evaluated by flow cytometry. (G) DCs and CD8+ T cells were isolated from dLNs of MC38 tumor–bearing mice and cocultured for 4 days. Anti–PD-L1 or control IgG was added into medium at a concentration of 10 μg/ml. Cell culture supernatants were harvested, and IFN-γ levels were measured by CBA. Data indicate mean ± SEM and are representative of 2 (A, B, G) or 3 (E, F), or a pool of 2 (C, D) independent experiments. Statistical analysis was performed using an unpaired Student’s 2-tailed t test. *P < 0.05; **P < 0.01; ***P < 0.001.

Comment in

References

    1. Zou W, Wolchok JD, Chen L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med. 2016;8(328):328rv4. doi: 10.1126/scitranslmed.aad7118. - DOI - PMC - PubMed
    1. Topalian SL, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443–2454. doi: 10.1056/NEJMoa1200690. - DOI - PMC - PubMed
    1. Brahmer JR, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455–2465. doi: 10.1056/NEJMoa1200694. - DOI - PMC - PubMed
    1. Sznol M, Chen L. Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced human cancer. Clin Cancer Res. 2013;19(5):1021–1034. doi: 10.1158/1078-0432.CCR-12-2063. - DOI - PMC - PubMed
    1. Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A. 2002;99(19):12293–12297. doi: 10.1073/pnas.192461099. - DOI - PMC - PubMed

Publication types