Abstract
Epidermal growth factor receptor wild type lung adenocarcinoma (EGFRWT LUAD) still has limited treatment options and unsatisfactory clinical outcomes. Ferroptosis, as a form of cell death, has been reported to play a dual role in regulating tumor cell survival. In this study, we constructed a 3-ferroptosis-gene signature, FeSig, and verified its accuracy and efficacy in predicting EGFRWT LUAD prognosis at both the RNA and protein levels. Patients with higher FeSig scores were found to have worse clinical outcomes. Additionally, we explored the relationship between FeSig and tumor microenvironment, revealing that enhanced interactions between fibroblasts and tumor cells in FeSighigh patients causing tumor resistance to ferroptosis. To address this challenge, we screened potential drugs from NCI-60 (The US National Cancer Institute 60 human tumour cell line anticancer drug screen) and Connectivity map database, ultimately identifying 6-mercatopurine (6-MP) as a promising candidate. Both in vitro and in vivo experiments demonstrated its efficacy in treating FeSighigh EGFRWT LUAD tumor models. In summary, we develop a novel FeSig for predicting prognosis and guiding drug application.
Similar content being viewed by others
Introduction
With an estimated 235,760 new cases and 131,880 deaths in the United States in 2021, lung cancer has become the most common cancer and the leading cause of cancer-related deaths worldwide1. Based on histopathological morphology, lung cancer can be classified into small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC), with NSCLC accounting for more than 80% of cases. According to the 2015 World Health Organization Lung Tumor Classification Guidelines, NSCLC comprises lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), and large cell carcinoma (LCC), with LUAD being the most common subtype, accounting for approximately 40–45% of cases2,3,4. Of note, the morbidity and mortality of LUAD are among the highest, making it a serious public health problem.
Epidermal growth factor receptor (EGFR) is a tyrosine kinase receptor that is associated with cell proliferation, migration, and apoptosis, and EGFR mutations are major drivers of NSCLC5. Approximately 15-30% of LUAD cases possess EGFR mutations, in which L858R missense mutations and exon 19 deletions accounting for 85% of EGFR-activating mutations. EGFR-targeted tyrosine kinase inhibitors (TKIs) have proven to be highly effective treatments for LUAD harboring the oncogenic EGFR mutation6. The successful use of EGFR-TKIs has shifted cancer treatment from empirical cytotoxic chemotherapy to molecularly targeted therapies. Nevertheless, TKIs treatments have limited efficacy in patients with EGFRWT LUAD. Therefore, there is an urgent need to investigate new strategies to combat EGFRWT LUAD.
Ferroptosis is an iron-dependent programmed cell death caused by massive iron accumulation and lipid peroxidation7. In recent years, the induction of ferroptosis has become a promising cancer treatment approach, which triggers the death of cancer cells, particularly in malignancies resistant to conventional therapies8,9. Accumulating evidence suggests that activating ferroptosis can facilitate anti-tumor treatments for various tumors10,11. For instance, erastin enhances the therapeutic effect of cisplatin in NSCLC by inducing ferroptosis, offering a new strategy for treating drug-resistant tumors12. Combined treatment with erastin and acetaminophen offers a new strategy for ferroptosis induction in lung cancer13. Consequently, studying the expression of ferroptosis-related genes in NSCLC and their prognostic value could lead to new strategies for the treatment of NSCLC. However, a comprehensive understanding of the role of ferroptosis-related genes in EGFRWT LUAD prognosis is still deficient, and the role of ferroptosis in EGFRWT LUAD remains largely unexplored.
In this study, we developed a 3-gene ferroptosis-related prognostic signature by analyzing RNA-seq data and corresponding clinical information of EGFRWT LUAD patients from TCGA (The Cancer Genome Atlas) database. This 3-gene model was further validated in 3 independent EGFRWT LUAD cohorts. Patients were classified into high-risk and low-risk groups based on this model. We also validated the protein expression profiles of the core genes using clinical tissue microarrays. By integrating the FeSig model and single-cell sequencing data, we obtained a deeper understanding of tumor microenvironment (TME) of high-risk patients, which may assist future TME-targeted drug investigations and developments. Notably, to enhance the clinical applicability of the FeSig model, we screened 6-Mercaptopurine (6-MP) as a promising drug for treating high-risk patients with EGFRWT LUAD.
Results
Identification of ferroptosis-associated genes as a prognostic feature of EGFRWT lung adenocarcinoma
The workflow for identifying ferroptosis-associated genes and constructing signature is delineated in Fig. 1a. We collected ferroptosis-related genes from ferroptosis database (FerroDb)14, while gene expression matrix and clinical traits of EGFRWT LUAD patients were extracted from The Cancer Genome Atlas (TCGA) lung adenocarcinoma database. Firstly, univariate cox analysis was performed, identifying 32 prognostic genes (Supplementary Fig. 1a). Subsequently, 11 prognostic genes were further refined by multivariate cox analysis, including 6 risk genes (PARK7, PRKCA, DUOX1, NOX4, DDIT4, GCLC) and 5 protective genes (SNCA, GDF15, PEBP1, HERPUD1, MFN2) (Fig. 1b). DDIT4, NOX4 and GCLC were significantly upregulated in tumor tissue (LogFC > 1 and p.adj < 0.05) and correlated with poor clinical outcomes (Supplementary Fig. 1b–d). After excluding protective genes, we established a 3-gene-based ferroptosis-related risk signature (FeSig) (Fig. 1c). The single-sample Gene Set Enrichment Analysis (ssGSEA)15 algorithm was applied to EGFRWT LUAD patients to obtain FeSig scores, categorizing patients into two groups according to the median score of FeSig (FeSighigh: n = 143, FeSiglow: n = 143). To evaluate FeSig’s predictive capabilities of survival among TCGA EGFRWT LUAD patients, Kaplan-Meier survival analysis, time-dependent receiver operating characteristic curve (ROC) and nomogram analysis were conducted. Results showed that FeSighigh patients had significantly shorter overall survival (OS) time than FeSiglow patients (Fig. 1d, p = 0.0048). In addition, the FeSig model can also predict survival across different tumor stages, with death events increasing as FeSig scores increased (Supplementary Fig. 1e–h). The area under curve (AUC) values of time-dependent ROC for OS were 0.71(1-year survival), 0.63(3-year survival) and 0.62(5-year survival) (Fig. 1e). Nomograms of 1 year, 3 years, and 5 years survival and corresponding calibration curves were constructed (Fig. 1f). Calibration curve indicated the favorable predictive accuracy of the nomogram model (Fig. 1g). To further validate predictive performance of FeSig, the FeSig was applied to 3 additional EGFRWT LUAD validation cohorts: GSE3121016 (n = 68), oncoSG17 (n = 73) and GSE1196918 (n = 58, Fig. 1h–j). FeSig performed robustly in predicting survival across these 3 datasets (Supplementary Fig. 2a–f). Collectively, these results support the FeSig model’s ability to predict the prognosis of EGFRWT LUAD patients.
a Overall flowchart of gene screening, analysis strategy and experiments designing. b expression differences, log2 fold change and hazard ratio of potential hazard genes. c Venn diagram of tumor upregulated ferroptosis genes and prognostic ferroptosis genes. Boxplot of DDIT4, NOX4, and GCLC expression in paired tumor and normal sample from TCGA-EGFRWT-LUAD (N = 35, T = 35, paired t test). d Kaplan–Meier survival plot (KM plot) of FeSighigh and FeSiglow patients. e AUC analysis of FeSig model. f Nomogram of 1-year,3-years and 5-years survival. g Calibration curve of nomogram. h KM plot of FeSighigh and FeSiglow patients in validation dataset GSE31210. i KM plot of FeSighigh and FeSiglow patients in validation dataset oncoSG. j KM plot of FeSighigh and FeSiglow patients in validation dataset GSE11969. Error bars depict as mean ± SD.
Functional investigations of FeSig genes
The impacts of FeSig genes on lung adenocarcinoma cell lines were evaluated by knocking down DDIT4, NOX4 and GCLC in A549, H441 and H1299, respectively (Fig. 2a, Supplementary Fig. 3a). Cell viabilities of these 3 cell lines were significantly impeded after the knockdown of these FeSig genes (Fig. 2b, Supplementary Fig. 3d). Clone formation assays also confirmed that the FeSig genes are closely associated with cell proliferation, as the number of colonies in the knockdown group was reduced compared to the control group (Fig. 2c, d, Supplementary Fig. 3b, f). Cell scratch assay was performed to assess the cell mobilities and scratch width was measured at 0 h and 36 h after treatment. We observed that after knocking down FeSig genes in A549, H441 and H1299, all 3 cell lines exhibited slower healing capabilities compared to the control group (Fig. 2e, f, Supplementary Fig. 3c, e).
a Western blot analysis of knocking down (KD) of DDIT4, NOX4 and GCLC in A549 and H441, respectively. b Cell viability analysis (Student’s t test, two-tailed). c Clone formation assay of control A549 and A549 with DDIT4, NOX4 and GCLC knocking down, respectively. d Quantitative analysis of clone formation assay of A549 (Student’s t test, two tailed). e Scratch assay of control A549 and A549 with DDIT4, NOX4 and GCLC knocking down, respectively. f Quantitative analysis of migration rate of A549 and corresponding gene silenced group (Student’s t test, two-tailed). g Flowcytometry assay of ferroptosis detection after RSL3 treatment (Student’s t test, two-tailed). Error bars depict as mean ± SD.
Since FeSig genes were initially screened based on their ferroptosis-related functions, we subsequently evaluated the alteration in ferroptosis sensitivity of A549 and H1299 after silencing FeSig genes. Results suggested that in the FeSig-gene silenced group, the percentage of ferroptotic cells were significantly increased after treatment with ferroptosis inducer RSL3 (Fig. 2g, Supplementary Fig. 4a, b). All these findings confirmed that DDIT4, NOX4 and GCLC act as ferroptosis inhibitors, suggesting higher expression of these genes inhibits ferroptosis in cancer cell, leading to unsatisfactory clinical outcomes.
Protein level validation of FeSig on EGFRWT lung adenocarcinoma microarray
Given the imperfect alignment between mRNA expression and protein levels19, we utilized a lung adenocarcinoma tissue microarray to validate survival states with FeSig at protein level (Fig. 3a). The mean fluorescence intensity of DDIT4 and GCLC was calculated by randomly selecting 5 views per sample. Samples were evaluated and categorized into FeSighigh and FeSiglow groups based on the median FeSig score. After excluding EGFRMUT patients, we obtained a cohort containing 61 EGFRWT LUAD patients (Supplementary Data). As expected, combining DDIT4 and GCLC fluorescence and clinical information, patients who fell into the FeSighigh group have significantly upregulated DDIT4 and GCLC protein expression (Fig. 3b–e). Meanwhile, a higher FeSig score predicted worse clinical outcomes (Fig. 3f), which is consistent with previous results derived from mRNA levels. We next combined the clinical traits of these 61 EGFRWT LUAD patients with FeSig score. Notably, the FeSig score increased with the progression of the tumor stage (Fig. 3g). Consequently, the FeSig score demonstrates efficacy at both mRNA and protein levels, emphasizing the promising potential for clinical applications.
a Tissue array information of EGFRWT LUAD sample. b Immunoflorescence of DDIT4 (red) and GCLC (white) in FeSighigh and FeSiglow sample, respectively. Scalebar: 20 μm. c Quantitative analysis of DDIT4 expression difference between FeSighigh (n = 31) and FeSiglow (n = 30) (Wilcoxon test). d Quantitative analysis of GCLC expression difference between FeSighigh (n = 31) and FeSiglow (n = 30) (Wilcoxon test). e Quantitative analysis of FeSig difference between FeSighigh (n = 31) and FeSiglow (n = 30) (Wilcoxon test). f KM plot of FeSighigh and FeSiglow patients in tissue array. g The relationship between FeSig and tumor stages (Fisher exact test). Error bars depict as mean ± SD.
FeSig closely related with fibroblasts infiltration in tumor microenvironment
To profile the relationship between TME and FeSig score, we leveraged immune and stromal cell feature genes derived from single-cell RNA sequencing data20. Each cell type was scored using ssGSEA algorithm, and cell type scores were then correlated with FeSig score to decode TME composition differences between FeSighigh and FeSiglow patients (Fig. 4a). Notably, across all 4 independent datasets, fibroblasts exhibited significant positive correlation with FeSig score, and the expression of fibroblast feature genes such as COL1A1, COL1A2, LUM were upregulated with the increasing of FeSig score, underscoring fibroblasts may be critical in modeling FeSighigh TME or vice versa (Fig. 4a, b). To validate this phenomenon, we checked hematoxylin-eosin (HE) slides from the TCGA cohort (Fig. 4c), including 4 patients in the FeSighigh group and another 4 patients in the FeSiglow group. Interestingly, FeSighigh patients displayed abundant fibroblasts infiltration in their TME (Fig. 4c), while the TME in FeSiglow patients primarily consisted of tumor cells and microvessels (Fig. 4c). We also observed similar TME composition differences in our LUAD microarray samples: FeSighigh samples showing more α-SMA area compared to FeSiglow samples (Fig. 4d, e). Altogether, these data highlight that fibroblasts may contribute to ferroptosis regulation in EGFRWT LUAD TME, potentially impacting the clinical outcomes of LUAD patients.
a Correlation analysis of FeSig and 20 TME cell signatures score. b Heatmap of fibroblast-related genes in TCGA-EGFRWT LUAD patients. c HE staining of FeSighigh and FeSiglow samples in TCGA-EGFRWT LUAD, scale bar: 500 μm. d Immunofluorescence staining of α-SMA (green) in FeSighigh and FeSiglow patients. Scale bar: 20 μm. e Quantitative analysis of mean percentage difference of α-SMA between FeSighigh (n = 31) and FeSiglow (n = 30) patients (Wilcoxon test). Error bars depict as mean ± SD.
Upregulation of fibroblast-cancer cell interaction in FeSighigh TME
To further elucidate relationships between ferroptosis state and fibroblasts, we queried a lung adenocarcinoma single-cell sequencing data from Gene Expression Omnibus (GEO) database, GSE13190720, to explore the interaction between fibroblasts and tumor cells under different FeSig conditions at single-cell resolution. After dimensionality reduction using Uniform Manifold Approximation and Projection (UMAP) and combining with marker-based cell type identification and annotation, we grouped 33632 cells into 9 clusters, including endothelial (n = 257), fibroblast (n = 663), mast cells (n = 680), plasma cells (n = 1705), NK cells (n = 1786), T cells (n = 11,380), epithelial (n = 9824), B cells (n = 2040) and myeloid cells (n = 5297) (Fig. 5a, b, Supplementary Fig. 5a, b). FeSig scores were applied to each patient using pseudobulk algorithm from muscat package. As expected, an enrichment of fibroblasts in FeSighigh patient was observed (2.85% in FeSighigh group, 0.53% in FeSiglow group), consistent with our results mentioned above in bulk RNA-seq data (Fig. 5c). We then performed CellChat analysis on this single-cell data and verified enhanced fibroblast-cancer cell interaction than other cell types, and this interaction in FeSighigh group was greater than that in FeSiglow group (Fig. 5d). To find out potential mechanisms by which fibroblasts regulate ferroptosis status of EGFRWT LUAD cells, we performed ligand-target analysis with Nichenetr package (Fig. 5e, top 80 targets were shown). Gene Ontology (GO) analysis of target genes enriched in insulin-like growth factor (IGF) binding-related pathways (Supplementary Fig. 5c). Interestingly, when examining specific Cellchat pathways in FeSighigh and FeSiglow groups, the IGF pathway was also identified in the FeSighigh cohort (Supplementary Fig. 5d). 4 independent datasets consistently demonstrated that patients with higher FeSig score exhibited higher IGF binding score (Supplementary Fig. 5e), indicating insulin-like growth factor binding-related pathways may contribute to tumor cell resistance to ferroptosis. To validate the correlation of these predicted ligand-receptor interactions with ferroptosis, we constructed a ligand-receptor-target (L-R-T) signature based on Nichenetr outputs. Results showed that this L-R-T signature is negatively correlated with ferroptosis state (TCGA-EGFRWT-LUAD: Rs = −0.293, p < 0.0001; GSE31210: Rs = −0.405, p = 0.0006; oncoSG: Rs = −0.296, p = 0.0109. Figure 5f–i), except dataset GSE11969 (Rs = −0.033, p = 0.803. Figure 5i). In conclusion, the interaction between fibroblasts and tumor epithelial cells in EGFRWT LUAD TME appears to regulate the ferroptosis state, stronger interaction and communication between fibroblasts and tumor epithelial cells leading to resistance to ferroptosis in tumor cells.
a UMAP (Uniform Manifold Approximation and Projection) plot depicts 9 cell types in EGFRWT LUAD tissue. b Dotplot showing cell type definition and marker gene average expression. c Percentage differences of 9 cell types between FeSighigh and FeSiglow cohorts. d Circos plot showing predicted ligand–receptor interactions between cell types. The width of lines indicates communication count number. Dotplot below shows the interaction differences between fibroblast and other cell types in FeSighigh and FeSiglow cohorts. e Heatmap showing top-ranked ligands’ activities which may regulate malignant epithelial cells according to NicheNet (Left). Heatmap of the downstream target genes in malignant cells which regulated by top-ranked ligands (Right). Correlation analysis of nichenet L-R-T communication score and ferroptosis score of (f) TCGA-LUAD, g GSE31210, h oncoSG and i GSE11969 cohort, respectively.
Drug screening and testing based on FeSig
Given the significant difference in prognosis between FeSighigh and FeSiglow patients, we combined both the NCI-60 database (NCI-60 Human Tumor Cell Lines Screen) and the Connectivity Map database to identify compounds that could potentially improve the prognosis of FeSighigh patients. The NCI-60 cell line expression profile was acquired, and the FeSig score for each cell line was calculated using the ssGSEA algorithm. The FeSig scores were then correlated with half-maximal inhibitory concentration (IC50) of compounds, which finally resulted 17 compounds, including 4 sensitive drugs and 13 resistant drugs (Fig. 6a). To further refine the list of potential compounds for FeSighigh patients, top 150 overexpressed genes in FeSighigh patients of TCGA-EGFRWT-LUAD were inputted to Connectivity map database to screen a secondary drug set. By intersecting the NCI-60 sensitive drugs with the Connectivity map drug sets, we identified 6-Mercaptopurine (6-MP), a purine antagonist, as a promising candidate (Fig. 6b). The effects of 6-MP on lung adenocarcinoma cell lines were subsequently tested both in vitro and in vivo.
a Correlation analysis of FeSig score and drug IC50 concentration from NCI-60 dataset. b Drug screening combining connectivity map dataset and sensitive drug screened from 6a. c Clone formation assay of H3122 and A549 under 0 mM, 0.5 mM and 1 mM 6-MP. d Quantitative analysis of clone formation assay in 6c (Student’s t test, two tailed). e Representative pictures of H3122 and A549 subcutaneous tumor with PBS control or 6-MP treatment. f Quantitative analysis of control tumor wight and 6-MP treated tumor wight of H3122 and A549, respectively. g IHC of α-SMA in control and 6-MP treated H3122 and A549 subcutaneous tumors. Scale bar: 100μm. h Relative fold change of decreased α-SMA area in 6-MP treated H3122 and A549 subcutaneous tumors. Error bars depict as mean ± SD.
We first pulled lung adenocarcinoma cell line RNA-sequencing data from Genomics of Drug sensitivity in Cancer (GDSC) database and profiled FeSig score for each lung adenocarcinoma cell line. We then selected 2 cell lines, H3122 (FeSig = 0.993, FeSiglow), A549 (FeSig = 1.407, FeSighigh), to assess the impact of 6-MP on cancer cell elimination. In vitro, clone formation assay demonstrated the efficacy of 6-MP in killing cancer cells increased with higher compound concentration (Fig. 6c, d). Notably, the cell line with a higher FeSig score (A549) displayed greater sensitivity to 6-MP compared to those with a lower FeSig score cell line (H3122) (Fig. 6c, d). To assess the in vivo tumor elimination capacities, we inoculated A549 and H3122 cancer cell lines subcutaneously into 6-week-old Balb/c nude mice and intraperitoneally administrated 6-MP or control PBS (Phosphate Buffered Saline), respectively. Results showed that 6-MP had limited effects on H3122 tumors (FeSiglow), while A549 tumors (FeSighigh) were more sensitive to 6-MP, consistent with in vitro colony formation assay (Fig. 6e, f).
Given the critical role of fibroblasts in TME modeling, we quantified fibroblasts infiltration alteration with or without 6-MP treatment using immunohistochemistry (IHC) (Fig. 6g). Surprisingly, 6-MP treatment significantly reduced fibroblast infiltration in A549 tumor models (sensitive to 6-MP), whereas H3122 tumor models (resistant to 6-MP) did not show any changes (Fig. 6g, Supplementary Fig. 6a). These findings suggested that 6-MP not only directly targets tumor cells, but also remodels the TME to improve chemotherapy outcomes in FeSighigh patients, indicating combining drug administration with 6-MP may offer significant benefits for FeSighigh EGFRWT LUAD patients.
Discussion
Ferroptosis, as a non-apoptotic and iron-dependent form of cell death, has been proven to play a regulatory role in carcinogenesis and cancer progression across various cancer types21,22,23,24,25. Meanwhile, ferroptosis is closely related to outcomes of clinical treatment of cancer, such as radiotherapy26,27, chemotherapy28,29, and immunotherapy30,31,32. Unlike EGFRMUT LUAD, the prognosis of EGFRWT LUAD patients is unsatisfactory due to the absent of targetable genes. To address this issue, many efforts have been paid to characterize specific subtypes of EGFRWT LUAD33,34,35. In this study, we integrated transcriptomic and ferroptotic features to identify potential ferroptosis-related subtypes of EGFRWT LUAD patients. Through univariate and multivariate Cox regression analysis, we got 11 risk-associated and 5 protective ferroptosis genes. To further refine molecule signature, we focused on overexpressed ferroptosis genes in EGFRWT LUAD and intersected them with the 11 risk genes, resulting in a three-gene signature: DDIT4, NOX4 and GCLC. This molecular signature, termed FeSig, was constructed based on these three genes. Survival analysis, time-dependent ROC of TCGA data and another three validation cohorts suggested FeSig can precisely predict the prognosis of EGFRWT LUAD patients (Fig. 7).
DDIT4 (DNA Damage Inducible Transcript 4) is known to be overexpressed in various of solid tumors, including head and neck squamous carcinoma36, triple-negative breast cancer37, colon cancer38, and is related to unsatisfactory clinical prognosis. Overexpression of DDIT4 in lung adenocarcinoma has been reported39,40 and the silence of DDIT4 enhances apoptosis and sensitizes lung adenocarcinoma cell lines, H358 and H838, to chemotherapies like ispinesib and cabazitaxel39. In addition, DDIT4 was included in many prognostic gene signatures for LUAD41,42,43, further confirming its role in regulating tumor growth, proliferation and predicting the prognosis of lung cancer patients. However, the detailed mechanisms by which DDIT4 regulates ferroptosis of EGFRWT-LUAD cancer cells still stay in opaque.
NOX4 (NADPH Oxidase 4) belongs to NOX family which functions as superoxide or hydrogen peroxide producer. Research has confirmed that NOX family members are closely related to cancer progression44. NOX4 has been implicated in resistance to EGFR-TKI therapy in NSCLC via enhancing IL-8/PD-L1 pathways45. Fan et al.46 demonstrated that EFHD2 induces the expression of NOX4 and through the EFHD2-NOX4-ROS-ABCC1 axis, promotes drug efflux, leading to chemotherapy resistance in non-small cell lung cancer. Chen et al.47 identified Fangchinoline (Fan) as a compound that reduces NSCLC metastasis by directly targeting NOX4, lowering cytoplasm ROS levels, and inhibiting the Akt-mTOR pathway, highlighting NOX4 as a promising therapeutic target in NSCLC. Besides affecting cancer cells directly, NOX4 also influences the TME by altering macrophages and fibroblast phenotypes. NOX4-derived ROS in the TME induces M2 macrophage polarization through ROS/PI3K signaling to facilitate cancer cell progression and immune evasion48. Cancer-associated fibroblasts (CAFs), as a component of TME, maintain an immune desert microenvironment characterized by the exclusion of CD8+ T cells through the expression of NOX4. Silencing NOX4 in CAFs remodels CAF phenotype and TME, potentially enhancing the effectiveness of immunotherapy49.
GCLC (Glutamate-cysteine ligase catalytic subunit) has been demonstrated to have an anti-ferroptosis function, which promotes cancer cell survival and progression in gastric cancer50, non-small-cell lung cancer51, and hepatocellular carcinoma52. GCLC regulates the synthesis of glutathione (GSH), which protects cancer cells from oxidative stress-induced ferroptosis50. Apart from the classic way of ferroptosis inhibition, Kang et al. discovered GCLC synthesize γ-glutamyl-peptide in a GSH-independent manner, further preventing ferroptosis in NSCLC51. Epigenetically53, KrasLSL-G12D/+ mouse NSCLC models demonstrated that histone variant H1.2 stabilizes nuclear NRF2 levels and enhances NRF2 binding to GCLC promoter, leading to upregulated GCLC expression in tumor cells. Depletion of H1.2 significantly reduced GCLC expression and induced oxidative stress, resulting in NSCLC regression and increased sensitivity to cisplatin. In our gene functional assay, all three FeSig exhibited anti-ferroptosis properties, and knocking down these genes significantly increased the sensitivity to ferroptosis inducer. By staining FeSig genes using IHC or mIF (multiple color immunofluorescence) and quantifying mean fluorescence intensities, we could profile the ferroptosis state of each patient, allowing for tailored clinical treatment strategies that could lead to a better prognosis.
Resistance to immunotherapies poses challenges to current immunotherapeutic strategies. Tumor patients exhibit varying responses to immunotherapy. TME of non-responders often enriched in exhausted T cells (Tex)54,55,56 and regulatory T cells (Treg), which are commonly targeted by mainstream immunotherapies57,58. Additionally, some non-responders exhibit a lack of immune effector cells within TME, which is defined as an immune-desert TME59,60,61. In such cases, even if T cells are targeted, they remain ineffective due to their exclusion from the tumor site. This type of TME is often enriched with tumor-associated fibroblasts. Liu et al. reported such cancer-fibroblast niche in hepatocellular carcinoma, where CAFs and SPP1 macrophages together form a barrier-like structure surrounding the tumor border, preventing the infiltration of T cells and other immune cells, ultimately leading to the failure of immunotherapy62.
Here, we utilized α-SMA to label fibroblasts. α-SMA, encoded by ACTA2, was reported as a marker of cancer-associated fibroblasts (CAFs)63,64, and ACTA2+ fibroblasts have been reported closely related to worse prognosis of gastric cancer65 and colorectal cancer64. However, Hui Ding et al reported that alveolar epithelial cells could also express α-SMA under the induction of TGF-β66. In the tumor microenvironment, TGF-β is expressed and secreted by multiple cell types, such as fibroblast, macrophage and tumor cell itself, which may lead heterologous expression of α-SMA out of fibroblasts. Also, α-SMA can be expressed by endothelial cells which undergo endothelial-mesenchymal transition (EndoMT) process under systematic sclerosis67, but whether such transition occurs in tumor microenvironment still unknown which needs further investigation and validation. In our analysis, FeSighigh patients characterized a phenotype where a CAF-barrier surrounds tumor regions, dividing the tumor into nest-like structure, which may impede T cell infiltration. Therefore, patients with this phenotype might benefit from CAF-targeting therapies.
To apply our FeSig feature to clinical practice, we screened FDA (Food and Drug Administration)-approved compounds to identify those that could benefit FeSighigh patients, with the goal of improving their survival. By integrating NCI-60 database and Connectivity Map compound library, we identified the purine antagonist 6-MP as a potential candidate. Both in vitro and in vivo experiments verified its beneficial effects on EGFRWT LUAD. However, our findings have certain limitations. 6-MP showed partial tumor elimination capacities in in vitro experiments. Notably, under the same concentration of 6-MP, the OD595 values of A549 were significantly lower than those of H3122. This suggests that A549 (FeSighigh) was more sensitive to 6-MP compared to H3122 (FeSiglow). The limited effects on FeSiglow in in vivo tumor models are likely due to the complex tumor microenvironments in vivo. Unlike in vitro culture systems, where tumor cells are directly exposed to the drug, in vivo tumor cells interact with multiple cell types, such as fibroblasts, macrophages, and immune cells. Cytokines, secretory proteins, and ligand-receptor interactions all contribute to complicated intracellular signaling pathways in tumor cells, affecting their response to drug treatment. Thus, in vitro experiments sometimes cannot perfectly simulate in vivo results.
The role of 6-MP as a purine antagonist in regulating tumor ferroptosis and affecting fibroblasts infiltration in TME remains unclear. Further experimental evidence is needed to determine whether 6-MP could enhance immunotherapeutic effects. In addition, the mechanisms by which interactions between fibroblasts and EGFRWT LUAD cancer cells lead to resistance to ferroptosis require further investigation and explanation. Potential mechanisms involving IGF and associated pathways in regulating ferroptosis need to be explored, and whether IGF could serve as a promising target for the treatment of EGFRWT LUAD also warrants detailed studies.
In summary, our study identified a 3-ferroptosis-inhibitor-gene signature, FeSig, which can be used to predict clinical prognosis at both mRNA and protein levels. FeSig can be conveniently applied to clinical use through techniques such as immunohistochemistry (IHC) or fluorescence in situ hybridization (FISH). Besides, FeSig can also characterize a fibroblast-enriched TME, aiding in the evaluation of immunotherapeutic responses in specific patients. Of note, we screened the purine antagonist, 6-MP, which shows promising potential in enhancing existing chemotherapies or neoadjuvant chemotherapies for FeSighigh EGFRWT LUAD patients.
Methods
Data collection and processing
RNA expression data, mutation information, and clinical characteristics for EGFRWT LUAD were downloaded from the UCSC-XENA database (https://xenabrowser.net/datapages/). Validation datasets GSE3121016 and GSE1196918 were obtained from GEO database, and the oncoSG cohort was downloaded from the cbioportal database17,68. EGFRMUT patients were excluded from subsequent analysis. Single-cell RNA sequencing data, GSE13190720, was also downloaded from the GEO database. Ferroptosis-related genes were obtained from FerroDB14.
Risk gene screening
Univariate and multivariate Cox regression analysis were conducted on the TCGA-EGFRWT LUAD cohort. Genes with significant prognostic properties in the univariate Cox regression analysis were selected as input for the multivariate cox regression analysis to narrow down candidate genes. Both univariate and multivariate Cox regression analysis were performed using survival and survminer R package.
FeSig scoring and survival analysis
GSVA R package was used to calculate FeSig score by setting methods as “ssgsva” and algorithm as “Gaussian” for array data or FPKM normalized data, detailed GSVA function and parameter settings as follow: gsva(as.matrix(Normalized gene expression matrix), gene signature, method = “ssgsea”, verbose = FALSE, kcdf = “Gaussian”). Gene signatures used in this manuscript were listed in Supplementary Data. Survival analysis of all four RNA sequencing data and one tissue array data were conducted using survival R package. Patients of TCGA-EGFRWT LUAD cohort were split into FeSighigh or FeSiglow according to median FeSig score, and another 3 validation cohorts’ patients were divided by survival and survminer R package.
Time-dependent ROC and nomogram analysis
Time-dependent ROC analysis was applied to all four RNA sequencing datasets using survivalROC R package. For TCGA-EGFRWT-LUAD, GSE31210 and oncoSG cohorts, ROC of 1 year, 3 years, and 5 years were evaluated, and for GSE11969, ROC of 3 years, 5 years, and 7 years were evaluated. Nomogram was conducted based on TCGA-EGFRWT-LUAD cohort and calibration curve was also plotted by utilizing survival and rms R package.
Correlation analysis
To correlate the FeSig score and cell types of TME, TME cell signature genes were obtained from GSE131907. TME cell scores were calculated using GSVA R package. In addition, for testing the relationship between ferroptosis state and ligand-receptor (L-R-T) interaction of all four RNA sequencing cohorts, ferroptosis signature score and L-R-T score were calculated and scored using GSVA package and cor.test function, respectively. Spearman correlation analysis was performed on all four datasets and |Rs| > 0.3 and p value < 0.05 was set to filter cell types that significantly correlated with FeSig.
Single-cell RNA sequencing data processing
Single-cell sequencing data was obtained from GEO database, and data normalization and dimensional reduction were performed by utilizing Seurat V469. Patients who harbor mutated EGFR were screened out and EGFRWT patients were retained for the subsequent analysis. To evaluate patients’ FeSig score, we employed pseudobulk algorithm from muscat70 R package and the yielded pseudobulk data matrix was subjected to the GSVA algorithm to calculate corresponding FeSig score and patients were categorized into FeSighigh and FeSiglow two groups based on the mean FeSig scores. Cell type annotation was based on canonical marker genes: Endothelial cells (PECAM1, CLDN5, FLT1), fibroblasts (DCN, COL1A1, LUM), mast cells (KIT, MS4A2, GATA2), plasma cells (IGHM, IGHG3, IGHA2), NK cells (NKG7, GNLY, NCAM1), T cells (CD3D, CD3E, TRAC), epithelial cells (EPCAM, KRT19, KRT18), B cell (CD79A, MS4A1, CD19), myeloid cells (LYZ, MACRO, CD68). To elucidate the interaction alterations under single-cell resolution, cell-cell interactions were predicted and evaluated by R package Cellchat71 and Nichenetr72 with default settings.
Drug screening
To screen potential clinical applicable drugs based on FeSig, drug information of IC50 and corresponding cell line gene expression profiles were downloaded from NCI-60 database. Spearman correlation analysis was applied to all drug IC50 and cell line FeSig score which was calculated by GSVA R package. The drugs with Rs < −0.3 and p < 0.05 were seen as sensitive drugs, while the drugs with Rs > 0.3 and p < 0.05 were resistant drugs. Meanwhile, FeSighigh patient’s overexpression genes were inputted into the Connectivity Map database for screening another drug set. The intersection of the NCI-60 drug set and the Connectivity Map drug was employed to narrow down potential drugs.
Cell culture
Human Embryonic Kidney (HEK) 293T, A549, H1299, H441, H3122 (A549 and H3122 were kindly provided by Guangya Zhu Lab) and indicated knock-down (KD) cells were maintained in DMEM/RPMI-1640 (Servicebio, Wuhan, China) supplemented with 10% FBS (Lonsera, catalog S711-001) and 1% penicillin-streptomycin (Gibco, catalog 15140148) in a 5% CO2 incubator at 37°C. HEK293T, A549, H1299, H441, and H3122 were authenticated by short tandem repeat (STR) genotyping. All cells were tested routinely to avoid infection of Mycoplasma.
Construction of DDIT4/NOX4/GCLC KD cells
To make stable KD cells (sequences are listed in Supplementary Data), sgRNA were cloned into a lentiCRISPR vector. 6 μg lentiviral plasmid DNA was co-transfected with 3 μg pMD2.G, 6 μg psPAX2 to a 10 cm dish of HEK293T cells with around 80% confluence of cells. After 48 hours transfection, culture medium containing lentivirus was collected and centrifuged for 10 min at 3,000 g at room temperature, lentivirus supernatant was then filtered using 0.22 μm Millipore Express PES membrane filter unit. A549/H441/H1299 cells were cultured in infection medium (culture medium: lentivirus supernatant: polybrene = 1 ml : 1 ml : 2 μl), infection medium was then replaced with normal culture medium after 12 h-infection. Two days after incubation, infected cells were selected with puromycin (Merck, catalog 540411-25MGCN, working concentration, 1 μg/mL) for 3 generations. Knocking down the efficacy of corresponding target protein was tested by immunoblotting. For siRNA mediated knocking down of genes, cells were seeded and cultured in 6-well plates (10,000 cells per well) with RPMI-1640 medium with 10% FBS and cultured until around 60% of confluence. 9 μl RNAiMAX regent (Invitrogen, catalog 13778150) was diluted in 150 μl Opti-MEM (Gibico, catalog 31985070) medium (solution A), 30 pmol siRNA was diluted in 150 μl Opti-MEM (solution B). Mix solution B to solution A at a ratio of 1:1 gently and incubated under room temperature for 5 min, then add the mixture solution to the corresponding well and cultured for 48 h. After 48-h culturation, culture medium was replaced with a fresh RPMI-1640 medium.
Cell viability
Cell viability was measured using the Cell Counting Kit (CCK-8) (Yeasen Biotechnology, Shanghai, China, catalog 40203ES60). In brief, cells were seeded into 96-well plates at a density of 1000 cells/well. Cells were then cultivated at 37 °C for 72 h. Upon measurement, 10 μl of the CCK8 reagent was added to each well and incubated for 60 min. The supernatant’s optical density (OD) values were then measured at wavelengths of 450 nm and compared within different groups. Each experiment was independently repeated three times.
Cell colony formation assay
The colony formation assay was performed, and indicated cells were plated into 6-well plate at 1,500 cells per well and cultured for 10 days. Colonies were fixed with 4% polyoxymethylene (Servicebio, Wuhan, China, catalog G1101-500ML) for 30 min and stained with 2% crystal violet (Sangon, Shanghai, China, catalog E607309-0100) for 30 min for colony visualization. To quantify clone formation, crystal violet stained cell clones were eluted by 10% acetic acid solution (Sinopharm, Shanghai, China). The value was measured using a microplate reader at a wavelength of 595 nm. The relative colony number stands for fold change in relation to the cell number of control group.
Wound-healing assay
After cells were cultured to a confluence around 90%, a small area was disrupted by scratching the monolayer with a 200 μl plastic pipette tip. Afterward, cells were cultured for 36 h with low serum medium (1%) and then taken images by microscope. The migration rate was calculated and compared within different groups. Each experiment was independently repeated three times.
Flow cytometric analysis
A549 and H1299 cells were plated into 6-well plate and cultured to confluence around 80%, ferroptosis was induced by RSL3 (Selleck, Shanghai, China) for 24 hours. After treatment of RSL3, cells were washed with cold PBS and stained with Invitrogen™ BODIPY™ 581/591 C11 at room temperature for 30 minutes. When staining finished, cells were washed with cold PBS twice and resuspended in flowcytometry buffer (PBS containing 1% FBS). The fluorescence of stained cells was captured and analyzed at the FITC channel.
Multi-fluorescence staining
Formalin-fixed and paraffin-embedded tumor samples were deparaffined, rehydrated, and pre-treated with 3% hydrogen peroxidase (Sangon, Shanghai, China) for 10 min. Antigen retrieval with 1× Citrate Antigen Retrieval Solution buffer (Beyotime, Shanghai, China) was conducted in a steam cooker for 20 min at 100 °C, followed by slowly cooling down to room temperature. After blocking with 10% Bovine Serum Albumin (BSA, Beyotime, Shanghai, China), sections were incubated with anti-DDIT4 antibody (1:200, Proteintech, 10638-1-AP) for overnight at 4°C. After incubation with signalstain(R) Boost IHC Det. (HRP, Rab), positive signals were visualized by AF 555 TYRAMIDE (Invitrogen, Shanghai, China). Then, sections were incubated with anti-GCLC antibody (1:200, Proteintech, 12601-1-AP) overnight at 4 °C. After incubation with signalstain (R) Boost IHC Det. (HRP, Rab), positive signals were visualized by AF 647 TYRAMIDE (Invitrogen, Shanghai, China). Last, sections were incubated with α-SMA antibody (1:200, CST,19245S) overnight at 4 °C. After incubation with signalstain (R) Boost IHC Det. (HRP,Rab), positive signals were visualized by AF 488 TYRAMIDE (Invitrogen, Shanghai, China). After staining of DAPI (Sangon, Shanghai, China), slides were sealed and imaged with microscope (TG TissueFAXS PLUS).
Animals
We have complied with all relevant ethical regulations for animal use. Mouse studies were approved by the Institutional Animal Care and Use Committee of The First Affiliated Hospital of USTC (2023-N(A)-0167). Four-week-old BALB/c-nu/nu male mice were purchased from SLAC Laboratory Animal Co. Ltd. (Shanghai, China). Mice were bred in a pathogen-free animal facility in The First Affiliated Hospital of USTC for 1 week before experiments. Mice were randomly grouped and injected subcutaneously with 1 × 106 A549 cells and NCI-H3122 cells within Matrigel-Matrix (Corning, catalog 354234). Experiments were terminated before the tumor volumes reached 1000 mm3. The volume of tumors was measured with calipers and calculated by the formula: Volume = 1/2 × length × width2. 6-MP (MedChemExpress, Shanghai, China) was applied 20 mg/kg, i.p., every 2 days when tumor volume reached 100 mm3. The maximal tumor volume (1000 mm3) was permitted by Institutional Animal Care and Use Committee of The First Affiliated Hospital of USTC and none of experiments exceeded the maximal limit.
Statistics and reproducibility
Data are presented as mean ± standard deviation. Experiments were conducted and at least three biological replicates were set. Paired t test was performed on paired-sample data (Fig. 1c). For a two-group comparison, the Wilcoxon test or t test were used depending on the distribution of data (Figs. 2b, d, f, g, 3c–e, 4e, 6d, f, h, Supplementary Fig 3b–f, 4a, b, 5e). Spearman correlation analysis was performed to determine the correlations between two variants (Figs. 4a, 5f–i, 6a). Fisher exact test was performed to determine the relationship of variants (Fig. 3g). Sample sizes of each graph were described in figures and figure captions or presented as dots in figures. Data analyses and visualization were performed by R version 4.2.2. p < 0.05 was considered a statistically significant difference.
Reporting summary
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.
Data availability
Public datasets used in this study can be found and downloaded with accession code: UCSC-XENA-TCGA-LUAD, GSE31210, GSE11969, oncoSG (data downloaded from cBioPortal), GSE131907. Uncropped images including western blots, scratch assays, clone formation assays and flow cytometry gating strategy can be found in Supplementary Information (Supplementary Fig. 7–9). Original source data supporting the results of this study are listed in Supplementary Data, which contains numerical data for Figs. 2b, d, f, 3c–e, 4e, 6d, f, h and Supplementary Fig. 3b–f. Other original image data supporting this study also available upon reasonable request from the corresponding authors.
Code availability
Analysis codes are deposited in https://github.com/wuqi12138/FeSig .
References
Siegel, R. L., Miller, K. D., Fuchs, H. E. & Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 71, 7–33 (2021).
Marentakis, P. et al. Lung cancer histology classification from CT images based on radiomics and deep learning models. Med. Biol. Eng. Comput. 59, 215–226 (2021).
Su, L. et al. CircRNAs in lung adenocarcinoma: diagnosis and therapy. Curr. Gene Ther. 22, 15–22 (2022).
Bade, B. C. & Dela Cruz, C. S. Lung Cancer 2020: epidemiology, etiology, and prevention. Clin. Chest Med. 41, 1–24 (2020).
Tang, J. C. et al. Shikonin enhances sensitization of gefitinib against wild-type EGFR non-small cell lung cancer via inhibition PKM2/stat3/cyclinD1 signal pathway. Life Sci. 204, 71–77 (2018).
Kleczko, E. K. et al. Novel EGFR-mutant mouse models of lung adenocarcinoma reveal adaptive immunity requirement for durable osimertinib response. Cancer Lett. 556, 216062 (2023).
Li, J. et al. Ferroptosis: past, present and future. Cell Death Dis. 11, 88 (2020).
Hassannia, B., Vandenabeele, P. & Vanden Berghe, T. Targeting ferroptosis to iron out cancer. Cancer Cell 35, 830–849 (2019).
Liang, C., Zhang, X., Yang, M. & Dong, X. Recent progress in ferroptosis inducers for cancer therapy. Adv. Mater. 31, e1904197 (2019).
Ma, S., Henson, E. S., Chen, Y. & Gibson, S. B. Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 7, e2307 (2016).
Lou, J. S. et al. Ginkgetin derived from Ginkgo biloba leaves enhances the therapeutic effect of cisplatin via ferroptosis-mediated disruption of the Nrf2/HO-1 axis in EGFR wild-type non-small-cell lung cancer. Phytomedicine 80, 153370 (2021).
Yamaguchi, H. et al. Caspase-independent cell death is involved in the negative effect of EGF receptor inhibitors on cisplatin in non-small cell lung cancer cells. Clin. Cancer Res. 19, 845–854 (2013).
Gai, C. et al. Acetaminophen sensitizing erastin-induced ferroptosis via modulation of Nrf2/heme oxygenase-1 signaling pathway in non-small-cell lung cancer. J. Cell Physiol. 235, 3329–3339 (2020).
Zhou, N. et al. FerrDb V2: update of the manually curated database of ferroptosis regulators and ferroptosis-disease associations. Nucleic Acids Res. 51, D571–d582 (2023).
Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinform. 14, 7 (2013).
Okayama, H. et al. Identification of genes upregulated in ALK-positive and EGFR/KRAS/ALK-negative lung adenocarcinomas. Cancer Res. 72, 100–111 (2012).
Chen, J. et al. Genomic landscape of lung adenocarcinoma in East Asians. Nat. Genet. 52, 177–186 (2020).
Takeuchi, T. et al. Expression profile-defined classification of lung adenocarcinoma shows close relationship with underlying major genetic changes and clinicopathologic behaviors. J. Clin. Oncol. 24, 1679–1688 (2006).
Anurag, M. et al. Proteogenomic markers of chemotherapy resistance and response in triple-negative breast cancer. Cancer Discov. 12, 2586–2605 (2022).
Kim, N. et al. Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nat. Commun. 11, 2285 (2020).
Gao, R. et al. YAP/TAZ and ATF4 drive resistance to Sorafenib in hepatocellular carcinoma by preventing ferroptosis. EMBO Mol. Med. 13, e14351 (2021).
Chen, Q. et al. SOCS2-enhanced ubiquitination of SLC7A11 promotes ferroptosis and radiosensitization in hepatocellular carcinoma. Cell Death Differ. 30, 137–151 (2023).
Yang, W. H. et al. The hippo pathway effector TAZ regulates ferroptosis in renal cell carcinoma. Cell Rep. 28, 2501–2508.e2504 (2019).
Jiang, K. et al. STC2 activates PRMT5 to induce radioresistance through DNA damage repair and ferroptosis pathways in esophageal squamous cell carcinoma. Redox Biol. 60, 102626 (2023).
Qiu, S. et al. Mitochondria-localized cGAS suppresses ferroptosis to promote cancer progression. Cell Res. 33, 299–311 (2023).
Lang, X. et al. Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 9, 1673–1685 (2019).
Zhang, Z. et al. Holo-lactoferrin: the link between ferroptosis and radiotherapy in triple-negative breast cancer. Theranostics 11, 3167–3182 (2021).
Niu, B. et al. Application of glutathione depletion in cancer therapy: Enhanced ROS-based therapy, ferroptosis, and chemotherapy. Biomaterials 277, 121110 (2021).
Wang, Y. et al. Overcoming cancer chemotherapy resistance by the induction of ferroptosis. Drug Resist Updat. 66, 100916 (2023).
Yang, F. et al. Ferroptosis heterogeneity in triple-negative breast cancer reveals an innovative immunotherapy combination strategy. Cell Metab. 35, 84–100.e108 (2023).
Gao, W., Wang, X., Zhou, Y., Wang, X. & Yu, Y. Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal Transduct. Target Ther. 7, 196 (2022).
Wang, W. et al. CD8(+) T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569, 270–274 (2019).
Zhao, F., Wang, M. & Zhu, J. Hypoxia-related lncRNAs to build prognostic classifier and reveal the immune characteristics of EGFR wild type and low expression of PD-L1 squamous and adenocarcinoma NSCLC. Cancer Med. 10, 6099–6113 (2021).
Wang, M., Zhu, J., Zhao, F. & Xiao, J. Transcriptome analyses identify a metabolic gene signature indicative of antitumor immunosuppression of EGFR wild type lung cancers with low PD-L1 expression. Front. Oncol. 11, 643503 (2021).
Planck, M., Isaksson, S., Veerla, S. & Staaf, J. Identification of transcriptional subgroups in EGFR-mutated and EGFR/KRAS wild-type lung adenocarcinoma reveals gene signatures associated with patient outcome. Clin. Cancer Res. 19, 5116–5126 (2013).
Yu, F. et al. RBM33 is a unique m(6)A RNA-binding protein that regulates ALKBH5 demethylase activity and substrate selectivity. Mol. Cell 83, 2003–2019.e2006 (2023).
Chen, X. et al. Identification of DDIT4 as a potential prognostic marker associated with chemotherapeutic and immunotherapeutic response in triple-negative breast cancer. World J. Surg. Oncol. 21, 194 (2023).
Fattahi, F. et al. High expression of DNA damage-inducible transcript 4 (DDIT4) is associated with advanced pathological features in the patients with colorectal cancer. Sci. Rep. 11, 13626 (2021).
Zhang, Y., Wang, Y., Chen, J., Xia, Y. & Huang, Y. A programmed cell death-related model based on machine learning for predicting prognosis and immunotherapy responses in patients with lung adenocarcinoma. Front. Immunol. 14, 1183230 (2023).
Song, X. et al. Streptococcus pneumoniae promotes migration and invasion of A549 cells in vitro by activating mTORC2/AKT through up-regulation of DDIT4 expression. Front. Microbiol. 13, 1046226 (2022).
Guo, Q. et al. Clinical roles of risk model based on differentially expressed genes in mesenchymal stem cells in prognosis and immunity of non-small cell lung cancer. Front. Genet. 13, 823075 (2022).
Shi, Y. et al. TKI resistant-based prognostic immune related gene signature in LUAD, in which FSCN1 contributes to tumor progression. Cancer Lett. 532, 215583 (2022).
Tang, B. et al. Identification of critical ferroptosis regulators in lung adenocarcinoma that RRM2 facilitates tumor immune infiltration by inhibiting ferroptotic death. Clin. Immunol. 232, 108872 (2021).
Skonieczna, M., Hejmo, T., Poterala-Hejmo, A., Cieslar-Pobuda, A. & Buldak, R. J. NADPH oxidases: insights into selected functions and mechanisms of action in cancer and stem cells. Oxid. Med. Cell Longev. 2017, 9420539 (2017).
Liu, W. J. et al. Elevated NOX4 promotes tumorigenesis and acquired EGFR-TKIs resistance via enhancing IL-8/PD-L1 signaling in NSCLC. Drug Resist Updat. 70, 100987 (2023).
Fan, C. C. et al. EFHD2 contributes to non-small cell lung cancer cisplatin resistance by the activation of NOX4-ROS-ABCC1 axis. Redox Biol. 34, 101571, (2020).
Chen, B. et al. Fangchinoline inhibits non-small cell lung cancer metastasis by reversing epithelial-mesenchymal transition and suppressing the cytosolic ROS-related Akt-mTOR signaling pathway. Cancer Lett. 543, 215783 (2022).
Zhang, J. et al. Tumoral NOX4 recruits M2 tumor-associated macrophages via ROS/PI3K signaling-dependent various cytokine production to promote NSCLC growth. Redox Biol. 22, 101116 (2019).
Ford, K. et al. NOX4 inhibition potentiates immunotherapy by overcoming cancer-associated fibroblast-mediated CD8 T-cell exclusion from tumors. Cancer Res 80, 1846–1860 (2020).
Yang, Z. et al. ACTL6A protects gastric cancer cells against ferroptosis through induction of glutathione synthesis. Nat. Commun. 14, 4193 (2023).
Kang, Y. P. et al. Non-canonical glutamate-cysteine ligase activity protects against ferroptosis. Cell Metab. 33, 174–189.e177 (2021).
Cui, Z. et al. Dihydroartemisinin enhances the inhibitory effect of sorafenib on HepG2 cells by inducing ferroptosis and inhibiting energy metabolism. J. Pharm. Sci. 148, 73–85 (2022).
Chen, Y. et al. An antioxidant feedforward cycle coordinated by linker histone variant H1.2 and NRF2 that drives nonsmall cell lung cancer progression. Proc. Natl Acad. Sci. USA 120, e2306288120 (2023).
Chow, A., Perica, K., Klebanoff, C. A. & Wolchok, J. D. Clinical implications of T cell exhaustion for cancer immunotherapy. Nat. Rev. Clin. Oncol. 19, 775–790 (2022).
Wang, Q., Qin, Y. & Li, B. CD8(+) T cell exhaustion and cancer immunotherapy. Cancer Lett. 559, 216043 (2023).
Bell, H. N. et al. Microenvironmental ammonia enhances T cell exhaustion in colorectal cancer. Cell Metab. 35, 134–149.e136 (2023).
Kang, J. H. & Zappasodi, R. Modulating Treg stability to improve cancer immunotherapy. Trends Cancer 9, 911–927 (2023).
Moreno Ayala, M. A. et al. CXCR3 expression in regulatory T cells drives interactions with type I dendritic cells in tumors to restrict CD8(+) T cell antitumor immunity. Immunity 56, 1613–1630.e1615 (2023).
Liu, Y. T. & Sun, Z. J. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics 11, 5365–5386 (2021).
Hornburg, M. et al. Single-cell dissection of cellular components and interactions shaping the tumor immune phenotypes in ovarian cancer. Cancer Cell 39, 928–944.e926 (2021).
Bagaev, A. et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell 39, 845–865.e847 (2021).
Liu, Y. et al. Identification of a tumour immune barrier in the HCC microenvironment that determines the efficacy of immunotherapy. J. Hepatol. 78, 770–782 (2023).
Jain, S. et al. Single-cell RNA sequencing and spatial transcriptomics reveal cancer-associated fibroblasts in glioblastoma with protumoral effects. J. Clin. Invest. 133, e147087 (2023).
Kobayashi, H. et al. The Origin and Contribution of Cancer-Associated Fibroblasts in Colorectal Carcinogenesis. Gastroenterology 162, 890–906 (2022).
Park, S. et al. ACTA2 expression predicts survival and is associated with response to immune checkpoint inhibitors in gastric cancer. Clin. Cancer Res 29, 1077–1085 (2023).
Ding, H., Chen, J., Qin, J., Chen, R. & Yi, Z. TGF-β-induced α-SMA expression is mediated by C/EBPβ acetylation in human alveolar epithelial cells. Mol. Med. 27, 22 (2021).
Manetti, M. et al. Endothelial-to-mesenchymal transition contributes to endothelial dysfunction and dermal fibrosis in systemic sclerosis. Ann. Rheum. Dis. 76, 924–934 (2017).
Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e3529 (2021).
Crowell, H. L. et al. muscat detects subpopulation-specific state transitions from multi-sample multi-condition single-cell transcriptomics data. Nat. Commun. 11, 6077 (2020).
Jin, S. et al. Inference and analysis of cell-cell communication using CellChat. Nat. Commun. 12, 1088 (2021).
Browaeys, R., Saelens, W. & Saeys, Y. NicheNet: modeling intercellular communication by linking ligands to target genes. Nat. Methods 17, 159–162 (2020).
Acknowledgements
This work was supported by the Natural Science Foundation of Anhui Province (2208085MH241, 2208085MH237), research funds of the Center for Advanced Interdisciplinary Science and Biomedicine of IHM (QYPY20220005), and Anhui Provincial Health Research Project (AHWJ2023BAa20192).
Author information
Authors and Affiliations
Contributions
Q.H. C.Z., and B.Y. conceived and supervised the project. C.Z., H.Y., and H.Z. designed and performed the research. Q.W. performed data analysis. C.Z., C.L., and H.Y. developed mouse models and related experiments. C.Z., B.Y., and Q.H. interpreted the results. C.Z., Q.W., and Q.H. wrote the manuscript with input from all other authors.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Communications Biology thanks Bo Chu, and the other anonymous reviewer for their contribution to the peer review of this work. Primary Handling Editor: Johannes Stortz. [A peer review file is available.]
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Zhang, C., Wu, Q., Yang, H. et al. Ferroptosis-related gene signature for predicting prognosis and identifying potential therapeutic drug in EGFR wild-type lung adenocarcinoma. Commun Biol 7, 1416 (2024). https://doi.org/10.1038/s42003-024-07117-1
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s42003-024-07117-1