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
. 2019 Jul 22;20(1):164.
doi: 10.1186/s12931-019-1137-4.

The canonical TGF-β/Smad signalling pathway is involved in PD-L1-induced primary resistance to EGFR-TKIs in EGFR-mutant non-small-cell lung cancer

Affiliations

The canonical TGF-β/Smad signalling pathway is involved in PD-L1-induced primary resistance to EGFR-TKIs in EGFR-mutant non-small-cell lung cancer

Yang Zhang et al. Respir Res. .

Abstract

Background: Approximately 30% of patients with epidermal growth factor receptor (EGFR)-activating mutations have no response to EGFR-tyrosine kinase inhibitors (TKIs) (primary resistance). However, little is known about the molecular mechanism involved in primary resistance to EGFR-TKIs in EGFR-mutant non-small cell lung cancer (NSCLC). Programmed death ligand-1 (PD-L1) plays important regulatory roles in intracellular functions and leads to acquired resistance to EGFR-TKIs in NSCLC. Here, we investigated the mechanistic role of PD-L1 in primary resistance to EGFR-TKIs in EGFR-mutant NSCLC cells.

Methods: The expression levels of PD-L1 and the sensitivity to gefitinib in H1975, HCC827 and PC-9 cells were determined by quantitative real-time PCR analysis (qRT-PCR) and Cell Counting Kit-8 (CCK-8) assays, respectively. Molecular manipulations (silencing or overexpression) were performed to assess the effect of PD-L1 on sensitivity to gefitinib, and a mouse xenograft model was used for in vivo confirmation. Western blotting and qRT-PCR were used to analyse the expression of epithelial-mesenchymal transition (EMT) markers. The effect of PD-L1 on migratory and invasive abilities was evaluated using the Transwell assay and mice tail intravenous injection.

Results: Higher expression of PD-L1 was related to less sensitivity to gefitinib in EGFR-mutant NSCLC cell lines. The overexpression or knockdown of PD-L1 presented diametrical sensitivity to gefitinib in vitro and in vivo. Furthermore, the overexpression of PD-L1 led to primary resistance to gefitinib through the induction of EMT, which was dependent on the upregulation of Smad3 phosphorylation. Moreover, in the mouse model, the knockdown of PD-L1 inhibited transforming growth factor (TGF)-β1-induced cell metastasis in vivo.

Conclusion: PD-L1 contributes to primary resistance to EGFR-TKI in EGFR-mutant NSCLC cells, which may be mediated through the induction of EMT via the activation of the TGF-β/Smad canonical signalling pathway.

Keywords: Drug resistance; EGFR-TKI; NSCLC; PD-L1; TGF-β/Smad signalling.

PubMed Disclaimer

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Correlation between PD-L1 expression and sensitivity to EGFR-TKIs in EGFR-mutant NSCLC cells (a) Three types of EGFR-mutant NSCLC cells were treated with different concentrations of gefitinib, and the IC50 value was calculated from the dose-response survival curve determined by the CCK-8 assay. b The mRNA expression level of PD-L1 in three types of EGFR-mutant NSCLC cells was detected by qRT-PCR. ***P < 0.001
Fig. 2
Fig. 2
PD-L1 is responsible for generating primary resistance to gefitinib. a PD-L1 mRNA and protein expression levels in stable HCC827 cell lines transfected with a PD-L1-expressing plasmid (PLVX-PD-L1) or a negative control (PLVX-NC). b PD-L1 mRNA and protein expression levels in stable HCC827 cell lines transfected with two PD-L1 ShRNAs (Sh-PD-L1–1 and Sh-PD-L1–2) or a negative control (Sh-PD-L1-NC). c and (d) Stable PD-L1-overexpressing and PD-L1-silenced HCC827 cells were treated with different concentrations of gefitinib, and the IC50 value was calculated from the dose-response survival curve determined by the CCK-8 assay. ***P < 0.001
Fig. 3
Fig. 3
Overexpression of PD-L1 attenuated sensitivity to gefitinib in vivo. a The tumour volume was measured at the indicated time intervals and calculated. See the Methods for details. b and (c) At the end of treatment, the tumours were excised, photographed as indicated and weighed. d Immunohistochemical staining of PD-L1, E-cadherin and vimentin was quantified based on staining intensity. *P < 0.05; **P < 0.01; ***P < 0.001
Fig. 4
Fig. 4
PD-L1 induced EMT and promoted migration and invasion. a and (b) E-cadherin, N- cadherin, vimentin and Snail mRNA and protein expression levels in stable PD-L1-overexpressing or PD-L1-silenced HCC827 cells were detected by qRT-PCR and western blotting, respectively. c and (d) Stable PD-L1-overexpressing or PD-L1-silenced HCC827 cells were allowed to migrate through an 8 μM pore Transwell insert, and the migrated cells were stained and counted in at least three microscopic fields (magnification, 100×). The cells were allowed to invade through Matrigel-coated Transwell membranes, and invasive cells were stained and counted under a light microscope. *P < 0.05; **P < 0.01; ***P < 0.001
Fig. 5
Fig. 5
PD-L1 induced EMT by upregulating Smad3 phosphorylation. a Western blot analysis of p-Smad3 and Smad3 in stable PD-L1-overexpressing or PD-L1-silenced HCC827 cells. The PD-L1 blot is the same blot shown in Fig. 2a and b. b After serum starvation for 24 h, PD-L1-overexpressing and PD-L1-silenced HCC827 cells were treated with SIS3(3 μM) or TGF-β1 (10 ng/ml) for 24 h. The expression of p-Smad3, Smad3, E-cadherin and vimentin was analysed by western blotting. c Knockdown of PD-L1 moderated TGF-β1-induced PAI-1 promoter activation. The relative PAI-1 promoter activities of PD-L1-silenced HCC827 cell lines were measured by a Dual-Luciferase Reporter Assay kit. Relative luciferase activity was expressed as the mean fold change from the basal level ± SD of three independent experiments. d and (e) Stable PD-L1-overexpressing and PD-L1-silenced HCC827 cells were treated with or without SIS3(3 μM) and TGF-β (10 ng/ml) for 24 h, respectively, and allowed to migrate through 8-μM pore Transwell inserts. The migrated cells were stained and counted in at least three microscopic fields (magnification, 100×). The cells were allowed to invade through Matrigel-coated Transwell membranes, and invasive cells were stained and counted under a light microscope
Fig. 6
Fig. 6
Knockdown of PD-L1 inhibits cell migration and invasion in vivo. Surgically resected lung tissues were fixed in Bouin’s fluid, and representative images of micrometastases detected by H&E staining are presented; red arrowheads indicate micrometastases

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30. doi: 10.3322/caac.21442. - DOI - PubMed
    1. Rothschild SI. Targeted therapies in non-small cell lung cancer-beyond EGFR and ALK. Cancers (Basel) 2015;7(2):930–949. doi: 10.3390/cancers7020816. - DOI - PMC - PubMed
    1. Wu YL, Cheng Y, Zhou X, et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): a randomised, open-label, phase 3 trial. Lancet Oncol. 2017;18(11):1454–1466. doi: 10.1016/S1470-2045(17)30608-3. - DOI - PubMed
    1. Yang JJ, Zhou C, Huang Y, et al. Icotinib versus whole-BRAIN irradiation in patients with EGFR-mutant non-small-cell lung cancer and multiple BRAIN metastases (BRAIN): a multicentre, phase 3, open-label, parallel, randomised controlled trial. Lancet Respir Med. 2017;5(9):707–716. doi: 10.1016/S2213-2600(17)30262-X. - DOI - PubMed
    1. Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010;362(25):2380–2388. doi: 10.1056/NEJMoa0909530. - DOI - PubMed

MeSH terms