Elsevier

Toxicology Letters

Volume 338, 1 March 2021, Pages 1-9
Toxicology Letters

Crosstalk between alveolar macrophages and alveolar epithelial cells/fibroblasts contributes to the pulmonary toxicity of gefitinib

https://doi.org/10.1016/j.toxlet.2020.11.011Get rights and content

Highlights

  • Macrophages contribute to gefitinib-induced pulmonary toxicity.

  • Macrophage-secreted MCP-1 is a key mediator of gefitinib-induced pulmonary toxicity.

  • MCP-1 promotes alveolar epithelial cells to undergo EMT.

  • MCP-1 participates in gefinitib-induced activation and antiapoptosis in fibroblasts.

  • Gefitinib increases Mcp-1 transcription level via the nuclear import of STAT3.

Abstract

Gefitinib is an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor indicated for the first-line treatment of patients with metastatic or advanced non-small cell lung cancer (NSCLC) whose tumors have specific EGFR mutations. Pulmonary toxicity is one of the fatal adverse effects of gefitinib and the underlying mechanism remains unclear. Here we demonstrated that alveolar macrophages contributed to gefitinib-induced pulmonary toxicity through promoting alveolar epithelial cells to undergo epithelial to mesenchymal transition (EMT) and inducing activation and antiapoptotic effect in fibroblasts. Further, we found that alveolar macrophage-secreted MCP-1 worked as a key factor in the pathologic changes of these two cell types. Gefitinib increased Mcp-1 transcription level via the nuclear import of the transcription factor STAT3. In conclusion, our data uncovered the underlying mechanisms of macrophage-promoted pulmonary toxicity in the presence of gefitinib. MCP-1 antibody or inhibition of STAT3 activation may represent novel therapeutic strategies for preventing gefitinib-induced pulmonary toxicity.

Introduction

Gefitinib, the first inhibitor of epidermal growth factor receptor (EGFR), has been approved for patients with specific EGFR mutation-positive non-small cell lung cancer (NSCLC) since 2003 (Cohen et al., 2003). Considering the role of EGFR in tumor progression, gefitinib has also been used to treat other malignant solid cancers in clinical trials and in vitro studies (Peng et al., 2016; Shao et al., 2016). However, increases in pulmonary toxicity are reported in patients receiving gefitinib, especially interstitial lung disease (ILD) (Takeda et al., 2015), which occurs with a low incidence but can be critical and fatal (Abdel-Rahman and Elhalawani, 2015). According to the U.S. Food and Drug Administration (FDA) database, cases of ILD have been observed in patients receiving gefitinib at an overall incidence of about 1%. Approximately 1/3 of the cases have been fatal. Patients with ILD are usually associated with a poor prognosis due to respiratory failure and severe comorbidities (Adegunsoye and Strek, 2016), leading to a 5-year survival rate of approximately 15–30 % (Walsh and Hansell, 2010). It has been reported that macrophages accumulated in the alveolar space from ILD patients after gefitinib treatment (Aoe et al., 2005). However, the functions of macrophages in gefitinib-induced pulmonary toxicity are unclear and need to be explored.

Fibrotic responses include the stimulation of epithelial-mesenchymal transition (EMT) in alveolar epithelial cells (Kasper and Haroske, 1996) and the formation of fibroblast and myofibroblast foci. EMT in alveolar epithelial cells have been widely observed in patients suffering from ILD, which may give rise to the fibroblasts (Cho et al., 2011). The fibroblast and myofibroblast foci secrete excessive amounts of extracellular matrix, mainly collagens, resulting in scarring, tissue destruction and the loss of lung function (Inoue et al., 2003). These reports suggest that alveolar epithelial cell EMT and fibroblast activation might participate in pulmonary toxicity of gefitinib.

Here, we found that alveolar macrophages contributed to gefitinib-induced pulmonary toxicity by promoting alveolar epithelial cells to undergo EMT and inducing activation and antiapoptotic effect in fibroblasts. Further, we uncovered that alveolar macrophages secreted-MCP-1 played a vital role in the pathologic changes of these two cell types. Gefitinib could increase Mcp-1 transcription level via the nuclear import of the transcription factor STAT3. Collectively, MCP-1, as a mediator, linked alveolar macrophages and the phenotypes associated with pulmonary toxicity. Our work illustrated the mechanism of gefitinib-induced pulmonary toxicity and suggested strategies for protecting against pulmonary toxicity by targeting MCP-1 or STAT3.

Section snippets

Cell lines and cell culture

The 3T6 mouse embryonic fibroblast cell line (Bates and Levene, 1971), TC-1 mouse alveolar epithelial cells (Lin et al., 1996), and MH-S mouse alveolar macrophages (Mbawuike and Herscowitz, 1989) were purchased from Anhui Qingzhi Biological Technology Company. Cells were maintained in high-glucose DMEM (Gibco, 10569010) supplemented with 10 % fetal bovine serum (FBS, HyClone, SV30160.03), 100 U/mL penicillin and 100 μg/mL streptomycin (Gibco, 10378016) in a humid atmosphere with 5% CO2 and 95 %

Alveolar macrophages triggered gefitinib-induced pulmonary toxicity via inducing alveolar epithelial cell EMT and fibroblast activation

Distinct cell populations appear to contribute to the complex and diverse pathogenesis of pulmonary toxicity. The stimulation of alveolar epithelial cell EMT and the formation of fibroblast and myofibroblast foci occur with the development of fibrosis (Kasper and Haroske, 1996). First, we detected EMT with the epithelial marker E-cadherin (E-CAD) and the mesenchymal markers Vimentin (VIM) and Fibronectin (FN). Dose selection of gefitinib was based on the range of 2−10-fold max serum

Discussion

As we reported before, pulmonary toxicity is a complex adverse drug reaction requiring the synergistic action of multiple cells (Li et al., 2019). It has been confirmed that parenchymal injury is the initial event, followed by alveolar epithelial cytokine signaling and cell recruitment. Our findings suggested that gefitinib-induced pulmonary toxicity were independent on the direct effect on alveolar epithelial cells and fibroblast activation. In our experiments, we found that alveolar

Conclusion

In conclusion, alveolar macrophages are crucial for gefitinib-induced pulmonary toxicity, which leads to EMT in alveolar epithelial cells and induces activation and antiapoptotic effect in fibroblasts. These effects might be achieved by secreted MCP-1, which is transcriptionally activated by STAT3 in alveolar macrophages. MCP-1 acts as a key mediator in the progression of gefitinib-induced pulmonary toxicity and may serve as an early detection marker. Furthermore, MCP-1 neutralizing antibody or

Funding

This work was supported by the National Natural Science Foundation of China [No. 81673457]; Medical Health Science and Technology Project of Zhejiang Provincial Health Commission [No. 2018KY519]; and Natural Science Foundation of Zhejiang Province [No. LSY19H310001].

Declaration of Competing Interest

The authors report no declarations of interest.

References (43)

  • M. Takeda et al.

    Pooled safety analysis of EGFR-TKI treatment for EGFR mutation-positive non-small cell lung cancer

    Lung Cancer

    (2015)
  • Z. Xu et al.

    High-mobility group box 1 protein-mediated necroptosis contributes to dasatinib-induced cardiotoxicity

    Toxicol. Lett.

    (2018)
  • H. Yan et al.

    ROS-dependent DNA damage contributes to crizotinib-induced hepatotoxicity via the apoptotic pathway

    Toxicol. Appl. Pharmacol.

    (2019)
  • J. Yang et al.

    Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis

    Cell

    (2004)
  • J. Zhao et al.

    Cerebrospinal fluid concentrations of gefitinib in patients with lung adenocarcinoma

    Clin. Lung Cancer

    (2013)
  • O. Abdel-Rahman et al.

    Risk of fatal pulmonary events in patients with advanced non-small-cell lung cancer treated with EGF receptor tyrosine kinase inhibitors: a comparative meta-analysis

    Future Oncol.

    (2015)
  • K. Aoe et al.

    Sudden onset of interstitial lung disease induced by gefitinib in a lung cancer patient with multiple drug allergy

    Anticancer Res.

    (2005)
  • J.H. Cho et al.

    Systems biology of interstitial lung diseases: integration of mRNA and microRNA expression changes

    BMC Med. Genomics

    (2011)
  • M.H. Cohen et al.

    FDA drug approval summary: gefitinib (ZD1839) (Iressa) tablets

    Oncologist

    (2003)
  • S.L. Deshmane et al.

    Monocyte chemoattractant protein-1 (MCP-1): an overview

    J. Interferon Cytokine Res.

    (2009)
  • K. Hacke et al.

    Regulation of MCP-1 chemokine transcription by p53

    Mol. Cancer

    (2010)
  • Cited by (6)

    • Resection of Scar Tissue in Rats With Spinal Cord Injury Can Promote the Expression of βⅢ-tubulin in the Injured Area

      2023, World Neurosurgery
      Citation Excerpt :

      The frozen tissue sections were cut at 20 μm (Leica CM1950, Wetzlar, Germany). Hematoxylin and eosin (HE) (Beyotime C0105M, Shanghai, China), Nissl (Beyotime C0117, Shanghai, China), and Tunel (Beyotime C1088, Shanghai, China) staining tissue sections were stained according to the kit.21-26 The images were captured with a digital slide scanner (3DHISTECH Pannoramic, Budapest, Hungary).

    1

    Equal contribution to this work.

    View full text