Abstract

BackgroundImmune related interferon regulatory factor 4 (IRF4) is a member of the IRF family, whereas the clinical significance and possible role of IRF4 in lung adenocarcinoma (LUAD) remains unclear. We aimed to investigate the role of IRF4 in predicting the prognosis of LUAD patients.MethodsUsing The Cancer Genome Atlas (TCGA) database and our immunohistochemical (IHC) cohort, we analyzed the correlation between IRF4 expression and clinical characteristics, and the prognostic value of IRF4 was also evaluated in LUAD. The potential biological functions of IRF4 in LUAD were analyzed by Gene Set Enrichment Analysis (GSEA). The relationship between IRF4 and immune cell infiltration were evaluated by TISIDB database and our own IHC cohort. In addition, an immune checkpoint inhibitor (ICI) treated cohort from Gene Expression Omnibus database was used to determine the role of IRF4 in LUAD patients with immunotherapy.ResultsWe found that either mRNA or protein expression level of IRF4 was significantly higher in LUAD than in normal tissues (P < 0.001). The elevate in IRF4 expression in LUAD was significantly associated with the earlier clinical stage (P = 0.002). Patients with LUAD and IRF4 high expression correlated with significant longer overall survival in both TCGA database (P < 0.05) and our IHC-cohort (P = 0.001). Our results also demonstrated that IRF4 could serve as an independent favorable prognostic factor in patients with LUAD. GSEA analysis indicated that high IRF4 expression group enriched with several immune-related pathways, such as B cell receptor signaling pathway, T cell receptor signaling pathway and cytokine-cytokine receptor interaction signaling pathway. In LUAD, IRF4 positively correlated with several different immune infiltrations including various B cells, CD8+ T cells and CD4+ T cells both in mRNA and protein levels. Additionally, we found that the expression of IRF4 was positively associated with PD-1 and PD-L1 mRNA expression levels, and IRF4 high expression predicted moderate better survival in LUAD with immunotherapy (P = 0.071).ConclusionsOur results suggested that IRF4 was associated with higher B cells and T cells infiltration levels and might be a favorable prognostic biomarker in LUAD patients, whereas the potential prognostic role of IRF4 in ICI-treated patients needed further exploration.

Highlights

  • Lung cancer is among the leading causes of cancer-related deaths, and it is the second frequent common cancer in the world [1, 2]

  • The interferon regulatory factor 4 (IRF4) expression levels were lower in Bladder urothelial carcinoma (BLCA), Breast invasive carcinoma (BRCA), Colon adenocarcinoma (COAD), Kidney chromophobe (KICH), Liver hepatocellular carcinoma (LIHC) and Rectum adenocarcinoma (READ)

  • Using UALCAN database, we identified that IRF4 expression was significantly higher in Lung adenocarcinoma (LUAD) tumor compared to normal samples both at the transcriptional and protein levels (P < 0.001 and P < 0.001) (Figures 1B, F)

Read more

Summary

Introduction

Lung cancer is among the leading causes of cancer-related deaths, and it is the second frequent common cancer in the world [1, 2]. Non-small-cell lung carcinoma (NSCLC) is the major type of lung cancer and has two main subtypes: Lung adenocarcinoma (LUAD) and Lung squamous cell carcinoma (LUSC) [3]. The efficacy of ICI was shown to be varied and only a limited percentage of NSCLC patients got promising prognoses [5]. The expression of PD-L1 has been served as an imperfect biomarker in NSCLC with ICI in our clinical practice [6]. Considering the importance of tumor immune-evasion in cancer treatment, identifying novel prognostic and immune-related biomarkers is necessary to guide clinical treatment in NSCLC. Immune related interferon regulatory factor 4 (IRF4) is a member of the IRF family, whereas the clinical significance and possible role of IRF4 in lung adenocarcinoma (LUAD) remains unclear. We aimed to investigate the role of IRF4 in predicting the prognosis of LUAD patients

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call