Abstract Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) are the most applied targeted therapy for EGFR-mutant lung adenocarcinoma (LUAD). The third-generation EGFR-TKI, osimertinib, is widely used throughout lung cancer treatment, with single or combination modes. One of the main barriers in osimertinib treatment is the acquired resistance and mechanisms are not fully understood. Gene expression other than genetic mutations might predict drug response and mediate resistance occurrence. We analyzed six datasets of osimertinib-resistant LUAD cells from the Gene Expression Omnibus (GEO) database and identified two hub genes, named MRAS and HEG1. We found that the expression mode of MRAS/HEG1 in LUAD was osimertinib-dependent and contributed to drug resistance. We also explored potential mechanisms of hub genes related osimertinib resistance and emphasized the M2 infiltration involved. Moreover, potential therapeutic agents conquering MRAS/HEG1-related resistance were also identified. In conclusion, MRAS and HEG1 might be responsible for osimertinib resistance and could be promising prognostic biomarkers for osimertinib response in LUAD, which might provide insights into therapeutic strategies. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-024-01552-6. Keywords: Lung adenocarcinoma (LUAD), Osimertinib resistance, Biomarker, Muscle RAS (MRAS), Heart of glass 1 or EF-like domains 1 (HEG1) Introduction Lung cancer is the cancer with the highest morbidity and mortality in China and LUAD is the most common type of non-small cell lung cancer (NSCLC) [[36]1]. The epidermal growth factor receptor (EGFR) is one of the common alterations in NSCLC patients and its frequency ranges from 11.9% (global) to 49.1% (Asia) [[37]2]. Studies from multiple centers in China show the frequency of EGFR mutation is 39.0–57.7% [[38]3]. The most frequent somatic mutations are in the kinase domain of EGFR and cause the constitutive activation independent of ligand, promoting the survival and proliferation of cancer cells [[39]4]. Most EGFR mutations comprise of exon 19 deletion and exon 21 L858R point mutation that is sensitive to first- (gefitinib and erlotinib) or second-generation (afatinib and dacomitinib) EGFR tyrosine kinase inhibitors (TKIs) [[40]5]. The third-generation EGFR TKI osimertinib showed superior efficacy in LUAD patients with EGFR mutation, especially acquired EGFR T790M-related resistance [[41]6]. However, the majority of LUAD patients who initially reported an osimertinib response will ultimately develop disease progression, which makes it crucial to identify biomarkers that predict resistance and guide treatment adjustments. The occurrence of acquired EGFR mutations (e.g., C797S) [[42]7] and loss or maintenance of the EGFR T790M mutation [[43]8] have been reported in tumors of patients with acquired osimertinib resistance. Additionally, off-target genetic alterations and amplification, as well as the activation of other signaling pathways have been discovered in a majority of LUAD patients with acquired osimertinib resistance [[44]2]. Increasing evidence has shown that the high proportion of osimertinib-resistant LUAD don’t harbor the new occurrence of genetic alterations [[45]9]. Recent studies demonstrated that EGFR-independent mechanisms without gene mutation include the activation of insulin-like growth factor (IGF)-1 receptor and Src/Akt [[46]10, [47]11]. However, the detailed resistant mechanisms remain unclear. A comprehensive understanding of acquired osimertinib-resistant mechanisms is essential to inform future therapeutic decisions for patients with EGFR mutation advanced LUAD. In the present study, we aimed to identify the osimertinib-resistant biomarkers and explored related mechanisms. We first analyzed six datasets of osimertinib-resistant LUAD cells and identified the hub genes associated with osimertinib resistance. The expression levels of muscle RAS (MRAS) and heart of glass 1 or EF-like domains 1 (HEG1) were verified in the published RNA-seq data online and tumor tissues collected from the osimertinib-sensitive/resistant patients in our hospital. We also assessed the prognostic value of MRAS and HEG1 in predicting the survival of LUAD. Additionally, we established osimertinib-resistant (OR) clones and conducted a series of experiments in vitro. We found that the expressions of MRAS and HEG1 in LUAD were in a dose/time-dependent manner until the osimertinib resistance developed. Molecular elucidations of MRAS and HEG1 based on the KEGG pathway and immune analysis were essential for understanding the osimertinib resistance occurrence. Finally, we discussed the prospect of clinical agents conquering the MRAS/HEG1-related osimertinib-resistant LUAD by performing oncoPredict analysis and in vitro crystal violet viability assay. Results Identification of the DEGs/hub genes by two bioinformatics in acquired osimertinib-resistant LUAD cells To explore the hub genes associated with acquired osimertinib resistance in LUAD, six datasets of LUAD cells sensitive or resistant to osimertinib were downloaded from the GEO database and subsequently processed for DEGs analysis. The results showed that there were 437 DEGs (GSE1033350), 2610 DEGs ([48]GSE153183), 125 DEGs ([49]GSE106765), 491 DEGs ([50]GSE163913), 2539 DEGs ([51]GSE201608), and 743 DEGs ([52]GSE193258) (Fig. [53]1A and Table S1). The DEGs in the six datasets were further analyzed for GO enrichment and pathway enrichment analysis (GSEA). The top 10 GO terms and the top 15 pathways are shown in Fig. [54]1B and C, respectively. The DEGs were enriched in the pathways related to drug metabolism and resistance, such as the metabolism of xenobiotics by cytochrome P450 (hsa00980), drug metabolism-cytochrome P450 (hsa00982), and DNA replication (hsa03030) (Fig. [55]1C, Tables S2 and S3). Additionally, two genes (MRAS and HEG1) were identified as increasing in osimertinib-resistant cells by intersecting the DEGs in the six datasets using the Venn plot (Fig. [56]1D). Fig. 1. [57]Fig. 1 [58]Open in a new tab Identification and analyses of DEGs in six datasets of osimertinib-resistant LUAD cells. A The volcano plots of the differential gene expressions in six datasets of osimertinib-resistant NSCLC cells. The -log10 (P-values) (Y-axis) is plotted against the average log2 (Fold Change) (X-axis) in gene expressions. The log2 (FC) and P-value are set at > 1 and < 0.05, respec-tively. B GO enrichment analysis based on the DEGs of six GEO datasets. The corresponding terms with GO number are in Table S2. C GSEA analysis based on the DEGs of GEO six datasets. The corresponding terms with KEGG number are in Table S3. D The Venn diagram shows the intersection of the DEGs in the six GEO datasets. Two genes are spotted Six datasets were merged to create a combined dataset which was used for DEGs analysis. First, the batch effects were eliminated from the combined matrix (Fig. [59]2A). The analysis revealed that there were 128 downregulated DEGs and 158 upregulated DEGs between osimertinib sensitive and resistant LUAD cells (Fig. [60]2B and C). The functional enrichment analysis showed that 158 DEGs related to osimertinib resistance were significantly associated with 195 GO terms and 66 KEGG pathways (Table S4 and S5). The top 10 GO terms and top 20 KEGG pathways are shown in Fig. [61]2D and E, respectively. The top five enriched pathways were cell cycle (hsa04110), spliceosome (hsa03040), ribosome biogenesis in eukaryotes (hsa03008), nucleocytoplasmic transport (hsa03013), and nucleotide metabolism (hsa03008). Genomic instability is known as one of the hallmarks of cancer. These findings may provide a new insight into osimertinib resistance in LUAD by examining DNA replication and repair. Fig. 2. [62]Fig. 2 [63]Open in a new tab Identification and analyses of DEGs from the combined datasets of osimertinib-resistant NSCLC cells. A Principal component analysis of six datasets before (upper panel) and after (low panel) elimination of batch effects. B The volcano plots of the differential gene expressions in the merged dataset of osimertinib-resistant NSCLC cells. C The -log10 (P-values) (Y-axis) is plotted against the average log2 (Fold Change) (X-axis) in gene expressions. The log2 (FC) and P-value are set at > 1 and < 0.05, respectively. D GO enrichment analysis based on 284 DEGs of the merged dataset. The top 10 GO terms are shown. The corresponding terms with GO number are in Table S4. E GESA analysis based the concordant differences genes (P < 0.05) of the merged dataset. The top 20 terms are shown. The corresponding terms with KEGG number are in Table S5 MRAS and HEG1 are the hub genes in LUAD with acquired osimertinib-resistance As shown in Fig. [64]3A, MRAS and HEG1 were also identified as the DEGs in the merged dataset. We examined the expression of MRAS and HEG1 in the six GEO datasets. The heatmap analysis revealed that the expression levels of both genes in osimertinib-resistant LUAD cells were higher than those in osimertinib-sensitive LUAD cells (Fig. [65]3B). We also observed higher expression of MRAS and HEG1 in acquired TKI-resistant LUAD tumors by analyzing the online transcriptome sequencing data [[66]12] (Fig. [67]3C). To further confirm the clinical significance of the MRAS and HEG1 expression in predicting response to osimertinib, we conducted immunohistochemical (IHC) analysis on tumor tissues collected from LUAD patients. The patients were assigned to the sensitive subgroup and the resistant subgroup. The tumor biopsies of the sensitive subgroup were surgically taken from LUAD patients who carried EGFR L858R before any treatment. The tumor biopsies of resistant subgroup were collected from LUAD patients who had disease progression after achieving osimertinib treatment for 12–23 months. The clinicopathological information of eight patients was shown in Table S6. We found that LUAD patients with acquired osimertinib resistance showed significantly higher expression of MRAS and HEG1, compared to LUAD patients prior to treatment (Fig. [68]3D and E). Moreover, patients with high levels of MRAS had poor clinical results in LUAD (P = 0.027) (Fig. [69]3F). As shown in Fig. [70]3G, higher expression of HEG1 is prone to shorter lifetimes but with no statistical differences (P = 0.15), which needs larger sample sizes to verify further (Fig. [71]3G). The above results indicate that MRAS has a good prediction performance in osimertinib resistance and prognosis, however, HEG1 seems only can be an osimertinib resistance predictor in LUAD. More data about the MRAS and HEG1 expressions after osimertinib treatment in LUAD from the real world are demanded to validate the predictive accuracy. Fig. 3. [72]Fig. 3 [73]Open in a new tab MRAS and HEG1 are highly expressed in osimertinib-resistant cells and tissues. A The Venn plot of DEGs from the merged dataset and six datasets. B The heatmap of MRAS and HEG1 in osimertinib-sensitive and resistant LUAD cells from six GEO datasets. C Bare mean of counts of MRAS and HEG1 from the online RNA-seq data [[74]9]. D IHC analysis of MRAS in tumor biopsies from LUAD patients (left panel). The DAB positive signals were analyzed by the Halo data analysis system (right panel). E IHC analysis of HEG1 in tumor biopsies from LUAD patients (left panel). The DAB positive signals were analyzed by the Halo data analysis system (right panel). F-G The Kaplan–Meier plots of the survival analysis of LUAD patient data from the [75]GSE14814 dataset (N = 265) MRAS and HEG1 expressions in LUAD are in osimertinib used-dependent manner until the resistance develops Next, we investigated the relationship between MRAS and HEG1 expressions and osimertinib sensitivity. We analyzed the expression in the tumor and adjacent normal tissues from the TCGA cohort. As shown in Fig. [76]4A and B, adjacent normal tissues showed higher expression levels of both MRAS and HEG1 compared to that in tumor tissues, which hinted the baseline levels of MRAS and HEG1 expressions in LUAD were low. We further examined the expression levels of MRAS and HEG1 in NCI-H1975 cells treated with osimertinib. We found that both mRNA and protein expression of MRAS and HEG1 were elevated in a dose-dependent manner (Fig. [77]4C–E). NCI-H1975 was treated with or without 10 nM of osimertinib at different time points. The qRT-PCR results showed that osimertinib induced MRAS and HEG1 expression in a time-dependent way, starting at two days after treatment (Fig. [78]4F and G). Immunoblotting data showed that MRAS and HEG1 proteins were significantly increased three days after treatment (Fig. [79]4H). Fig. 4. [80]Fig. 4 [81]Open in a new tab MRAS and HEG1 are in osimertinib use-dependent manner in LUAD. A The paired expression of MRAS in the LUAD cohort from the TCGA dataset. B The paired expression of HEG1 in the LUAD cohort from the TCGA dataset. C NCI-H1975 cells were treated with indicated concentations of osimertinib for 72 h. QRT-PCR was conducted to analyze MRAS mRNA. D NCI-H1975 cells were treated with indicated concentations of osimertinib for 72 h. QRT-PCR was conducted to analyze HEG1 mRNA. E NCI-H1975 cells were treated with indicated concentrations of osimertinib for 72 h. WB was conducted to analyze MRAS and HEG1 proteins. F NCI-H1975 cells were treated with 10 nM of osimertinib for the indicated time. QRT-PCR was conducted to analyze MRAS mRNA. G NCI-H1975 cells were treated with 10 nM of osimertinib for the indicated time. QRT-PCR was conducted to analyze HEG1 mRNA. H NCI-H1975 cells were treated with 10 nM of osimertinib for the indicated time. WB was conducted to analyze MRAS and HEG1 protein. I The schematic figure of establishing osimertinib-resistant (OR) clones. J Crystal violet cytotoxic assay was conducted to assess IC[50] of osimertinib in NCI-H1975 parental (Par) and OR clones. K QRT-PCR (top) and WB (bottom) analysis of MRAS in NCI-H1975 Par and OR cells. L. QRT-PCR (top) and WB (bottom) analysis of HEG1 in NCI-H1975 Par and OR cells. *, P < 0.05; ***, P < 0.001; ****, P < 0.0001 To further explore the roles of MRAS and HEG1 in acquired osimertinib resistance, we generated osimertinib-resistant NCI-H1975 clones (osimertinib-resistant-1/2, OR-1/2) as shown in the schematic figure (F[82]ig. [83]4I). The sensitivities of parental and resistant NCI-H1975 cells to osimertinib were evaluated using a crystal violet cytotoxic assay. As shown in Fig. [84]4J, OR-1/2 clones displayed remarkably enhanced resistance to osimertinib, with the IC[50] increasing by approximately two hundred thousand-fold. We found that mRNA and protein expressions of MRAS and HEG1 were remarkably upregulated in OR-1/2 clones compared to their parental counterparts (Fig. [85]4K and L). Combining all these data above, MRAS and HEG1 were proven to play important roles in the osimertinib-resistant LUAD but the mechanism remained unknown. Roles of MRAS and HEG1 in osimertinib resistant LUAD We further investigated the potential resistant mechanisms of MRAS and HEG1 by performing the KEGG pathway analysis in the TCGA cohort. The median MRAS and HEG1 expression were used as the cut-off value to assign samples with higher expression to the MRAS^high and HEG1^high subgroups (Fig. [86]5A and C), respectively. The EGFR mutation status were also analyzed in the TCGA cohort (Table S9). The MRAS^high subgroup showed highly enriched pathways in viral protein interaction with cytokine and receptor, cytokine-cytokine receptor interaction, cell adhesion molecules, malaria, and protein digestion and absorption (Fig. [87]5B). On the other hand, the MRAS^low subgroup had highly enriched pathways in maturity-onset diabetes of the young, arginine and proline metabolism, complement and coagulation cascades, gastric cancer, and metabolism of xenobiotics by cytochrome P450 (Fig. [88]5B). The most highly enriched pathways in the HEG1^high subgroup were ECM-receptor interaction, protein digestion and absorption, PI3K-Akt signaling pathway, focal adhesion, and neuroactive lig-and-receptor interaction; the most enriched pathways in the HEG1^low subgroup were ascorbate and aldarate metabolism, biosynthesis of nucleotide sugars, aldosterone-regulated sodium reabsorption, and inositol phosphate metabolism (Fig. [89]5D). Fig. 5. [90]Fig. 5 [91]Open in a new tab KEGG analysis based on the hub genes. A The volcano plot of DEGs in MRAS^high and MRAS^low subgroups from TCGA dataset. The -log10 (P-values) (Y-axis) is plotted against the average log2 (Fold Change) (X-axis) in gene expressions. The log2 (FC) and P-value are set at > 1 and < 0.05, respectively. B KEGG analysis based on the MRAS^high and MRAS^low subgroups. C The volcano plot of DEGs in HEG1^high and HEG1^low subgroups from TCGA dataset. The -log10 (P-values) (Y-axis) is plotted against the average log2 (Fold Change) (X-axis) in gene expressions. The log2 (FC) and P-value are set at > 1 and < 0.05, respectively. D KEGG analysis based on the HEG1^high and HEG1^low subgroups Evidence has shown that acquired resistance to osimertinib in NSCLC patients is associated with the mutual effects between the tumor and its surrounding microenvironment [[92]13]. We tried to clarify whether MRAS and HEG1 contributed to building the immunosuppressive environment after osimertinib treatment. The CIBERSOR method was used to calculate the degree of immune cell infiltration in each sample from the TCGA cohort and analyze the correlation between MRAS and HEG1 and immune infiltration (Fig. [93]6A and B). Of note, both MRAS and HEG1 had positive relations with M2 infiltration. We also explored the expression relation between two hub genes and 60 representative genes in the immune checkpoint pathway [[94]14], which showed closely positive correlations (Fig. [95]6C and D). Fig. 6. [96]Fig. 6 [97]Open in a new tab Relations between the hub genes and immune-related cells and genes. A-B The relationship between MRAS/HEG1 expressions and immune cell infiltration. C-D The relationship between MRAS/HEG1 expressions and immune markers We also analyzed the expression of MRAS and HEG1 in LUAD patients from the TCGA database who had the most common clinical mutations linked to EGFR TKI resistance [[98]15, [99]16] (Figure S1 and S2). These findings indicate that the bypassing pathway activating, and immune microenvironment remodeling contribute to MRAS and HEG1-related-acquired resistance to osimertinib in LUAD patients. Identification of potential therapeutic agents for the MRAS/HEG1-related osimertinib resistance Upregulation of MRAS and HEG1 in LUAD is closely related to osimertinib resistance but there is no existing strategy using MRAS or HEG1 inhibition to conquer the resistance. The signaling pathways accompanied by overexpressing MRAS and HEG1 might be potential targets in LUAD. We used the oncoPredict model to identify potential therapeutic agents for the MRAS/HEG1-related osimertinib resistance. Our findings predicted that six agents, including AZD8186, AZD1332, cerdiranib, dasatinib, doramapimod, and WIKI4 were effective against osimertinib-resistant NSCLC cells, MRAS^high LUAD patients, and HEG1^high LUAD patients (Fig. [100]7). The sensitivities of parental NCI-H1975 and resistant OR-1/2 cells to these agents were subsequently examined using a crystal violet cytotoxic assay (Fig. [101]8). OR-1/2 cells showed relatively higher sensitivity to doramapimod compared to parental NCI-H1975 cells (Fig. [102]8C). However, compared to parental NCI-H1975 cells, OR-1 and OR-2 displayed reduced and enhanced sensitivities to WIKI4, respectively (Fig. [103]8D). Fig. 7. [104]Fig. 7 [105]Open in a new tab OncoPredict analysis of osimertinib-resistant LUAD cells and patients with differential expression of MRAS and HEG1. A OncoPredict analysis of osimertinib-sensitive or resistant LUAD cells to six agents. B OncoPredict analysis of LUAD patients with low and high levels of MRAS to six agents. C OncoPredict analysis of LUAD patients with low and high levels of HEG1 to six agents Fig. 8. [106]Fig. 8 [107]Open in a new tab Crystal violet cytotoxic assay in parental NCI-H1975 and OR-1/2 clones. A Crystal violet cytotoxic assay was conducted to assess IC[50] of AZD1332 in NCI-H1975 parental (Par) and osimertinib-resistant (OR) clones. B Crystal violet cytotoxic assay was conducted to assess IC[50] of dasatinib in NCI-H1975 Par and OR clones. C Crystal violet cytotoxic assay was conducted to assess IC[50] of doramapimod in NCI-H1975 Par and OR clones. D Crystal violet cytotoxic assay was conducted to assess IC[50] of WIKI4 in NCI-H1975 Par and OR clones. E Crystal violet cytotoxic assay was conducted to assess IC[50] of AZD8186 in NCI-H1975 Par and OR clones. F Crystal violet cytotoxic assay was conducted to assess IC[50] of cediranib in NCI-H1975 Par and OR clones Methods Data download We downloaded six datasets related to the EGFR-TKI resistance from the Gene Expression Omnibus (GEO, [108]https://www.ncbi.nlm.nih.gov/gds), including [109]GSE103350, [110]GSE106765, [111]GSE153183, [112]GSE163913, [113]GSE201608 and [114]GSE193258. Only osimertinib sensitive and resistant samples could be included in the study. The specific dataset taken for LUAD resistant or sensitive cells for osimertinib are listed as follows: Osimertinib sensitive cells datasets: [115]GSE103350 dataset: [116]GSM2768998 [117]GSE193258 dataset: HCC2935_DMSO_1, HCC2935_DMSO_2, HCC2935_DMSO_3, HCC827_DMSO_1, HCC827_DMSO_2, HCC827_DMSO_3, H1975_DMSO_1, H1975_DMSO_2, H1975_DMSO_3, PC9_DMSO_1, PC9_DMSO_2 and PC9_DMSO_3. [118]GSE106765 dataset: [119]GSM2850068 and [120]GSM2850071. [121]GSE153183 dataset: [122]GSM4635290 and [123]GSM4635291. [124]GSE163913 dataset: [125]GSM4990622 and [126]GSM4990625. [127]GSE201608 dataset: [128]GSM6068510. Osimertinib resistant cells datasets: [129]GSE103350 dataset: [130]GSM2769000 and [131]GSM2769003. [132]GSE193258 dataset: HCC2935_osi_DTP_1, HCC2935_osi_DTP_2, HCC2935_osi_DTP_3, HCC827_osi_DTP_1, HCC827_osi_DTP_2, HCC827_osi_DTP_3, H1975_osi_DTP_1, H1975_osi_DTP_2, H1975_osi_DTP_3, PC9_osi_DTP_1, PC9_osi_DTP_2 and PC9_osi_DTP_3. [133]GSE106765 dataset: [134]GSM2850070 and [135]GSM2850073. [136]GSE153183 dataset: GSM4635292and [137]GSM4635293. [138]GSE163913 dataset: [139]GSM4990624 and [140]GSM4990627. [141]GSE201608 dataset: [142]GSM6068511 and [143]GSM6068512. The survival analysis was conducted in [144]GSE14814. Besides, the RNA-seq data, mutation data and clinical pathological data of LUAD samples were downloaded from the TCGA Genomic Data Commons. Differentially expressed genes (DEGs) analysis We used two methods to select DEGs associated with osimertinib resistance. The first method uses the edgeR R package to identify DEGs [[145]17]. The genes satisfying |log2(FC)|> 1 and P-value < 0.05 genes were considered to have significant differences. The second method combined the datasets with the R software package named inSilicoMerging [[146]18]. We used Empirical Bayes to eliminate batch effects [[147]19]. The DESeq2 R package was finally used to find out the DEGs of the combined dataset. When |log2(FC)|> 1 and P-value < 0.05 were satisfied, the genes were statistically significant in the combined matrix [[148]20]. Function enrichment analysis The DEGs screened from the methods above were subjected to Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and Gene Set Enrichment Analysis (GSEA). A P-value < 0.05 was considered significant. The final visualizations of function enrichment analysis were performed by the ggplot2 R package. Drug sensitivity prediction To predict potential therapeutic agents for the acquired osimertinib-resistant patients, we used the "oncoPredict" R package to predict the semi-maximum inhibitory concentration (IC[50]) of therapeutic drugs in osimertinib-sensitive and resistant cells, as well as MRAS^high/MRAS^low and HEG1^high/HEG1^low NSCLC patients [[149]21]. The package was designed by using the cell lines expression and the TCGA gene expression profiles on the Genomics of Cancer drug sensitivity in Cancer (GDSC, [150]https://www.cancerrxgene.org) to construct Ridge regression models. Cell lines and reagents NCI-H1975 cell and HEK-293T were cultured in DMEM (Cat. 11995500BT, Gibco, USA) and 1640 (Cat. C11875500BT, Gibco, USA), 10% fetal bovine serum (FBS, Vazyme, China), and 1% penicillin–streptomycin (Cat. 150063, Invitrogen, USA). HEK-293T and NCI-H1975 cells were purchased from BeNa Culture Collection (BNCC, China) and authenticated periodically (at least every 6 months) via STR profiling (Beijing Tsingke Biotech, China). NCI-H1975 cells were treated with 2 μM of osimertinib for seven days and surviving clones were selected and continually maintained in the dose of 2 μM. Osimertinib (Cat. S7297) and AZD1332 (Cat. A-495) were purchased from Selleck (USA) and Alomone Labs (Israel), respectively. AZD8186 (Cat. HY-12330), cerdiranib (Cat. HY-10205), dasatinib (Cat. HY-10181), doramapimod (Cat. HY-10320), and WIKI4 (Cat. HY-16910) were purchased from MedChem Express (USA). Crystal violet viability assay Cells were seeded in 96-well plates followed by drug treatment for 72 h. Cells were then stained with crystal violet (Cat. A600331, Sangon Biotech, China) for 30 min. The crystal violet solution was washed with running water and the plate was dried. Crystal violet was dissolved with fresh 100 mM sodium citrate solution for one hour and the absorbance was determined at 595 nm using a colorimetric plate reader (VersaMax™, Molecular Devices). Western blot (WB) Cells were washed with cold phosphate-buffered saline (PBS, pH 7.4) three times and lysed with radioimmunoprecipitation assay buffer (RIPA buffer, Cat. 89900, Thermo Fisher, USA) supplemented with protease and phosphatase inhibitor (Cat. A32959, Thermo Fisher, USA). Protein was quantified using Bradford assay (Cat. 5000001, Bio-Rad, USA). At least 40 μg of lysates were resolved by SDS-PAGE gel and transferred to 0.45 μm PVDF (Cat. IPVH00010, Millipore, USA). The blots were blocked with 5% non-fat milk dissolved in tris-buffered saline (TBS) supplemented with 0.1% Tween® 20 detergent (TBST) at room temperature for one hour and processed for the incubation of indicated primary antibodies at 4 ℃ overnight. The blots were washed with TBST and incubated with appropriated horseradish peroxidase-conjugated secondary antibodies (Cat. 31430 and 31460, Thermo Fisher, USA) at room temperature for one hour followed by visualization of immunoreactive bands by chemiluminescence (Cat. E423-01, Vazyme, China). Antibodies are as follows: MRAS (Cat. PA5-112613, Thermo Fisher, USA), HEG1 (Cat. Bs-15449R, Bioss Biotech, China), and GAPDH (Cat. 301341, Zenbio, China). Quantitative reverse transcription polymerase chain reaction (qRT-PCR) Messenger RNA (mRNA) was isolated from cells using TRIzol™ (Cat. 15596026, Invitrogen, USA), followed by cDNA synthesis using a qPCR cDNA synthesis kit (Cat. R323-01, Vazyme, China). PCR reactions were conducted using ChamQ SYBR qPCR Master Mix (Cat. Q321-02, Vazyme, China) and the Bio-Rad CFX96 deep cycler. Relative mRNA expression was calculated by the comparative CT method. Primers are listed below: Gapdh-forward: 5′-GTCTCCTCTGACTTCAACAGCG-3′, Gapdh-reverse: 5′-ACCACCCTGTTGCTGTAGCCAA-3′. MRAS-forward: 5′-CCACCATTGAAGACTCCTACCTG-3′, MRAS-reverse: 5′-ACGGAGTAGACGATGAGGAAGC-3′. HEG1-forward: 5′-CTGCCACCTTTGCTGTTCAGAG-3′, HEG1-reverse: 5′-CTGGTGTTGTCTGCGACGCATT-3′. Patients and tissue specimen The tumor slices were obtained from Sichuan Cancer Hospital & Institute, Sichuan Cancer Center with approval from the Institutional Ethics Committee (protocol code: SCCHEC-04-2022-002). All patients signed the informed consent form, and clinical information is provided in Supplementary Table 6. Immunohistochemistry (IHC) Immunohistochemical staining was performed on tumor tissue slides as previously described [[151]22]. The slides were incubated with rabbit polyclonal anti-HEG1 (1:100, Cat. Bs-15449R, Bioss, China) and anti-MRAS (1:100, Cat. PA5-112613, Thermo Fisher, USA) antibodies at 4℃ overnight. HEG1 and MRAS expression were evaluated by using the Halo data analysis system to calculate the 3’,3’-doaminobenzidine tetrahydrochloride (DBA) positive area proportion of high-power images (40X). Statistics The significance between treatment groups for the in vitro studies was determined by a Student t-test (two-tailed) or a One-way ANOVA (two-tailed) with Dunnett’s post-hoc test. The significance in IHC analysis was determined by an independent samples t-test. P values less than 0.05 were categorized as statistically significant. Error bars for all figures represent SD unless otherwise stated. Figures of in vitro experiments are representative of at least three independent experiments. Discussion EGFR TKIs are commonly administered to treat NSCLC patients with EGFR mutations [[152]23–[153]26]. However, in more than half of the patients receiving first- or second-generation EGFR TKIs, the occurrence of a new EGFR mutation, T790M, contributes to acquired resistance [[154]27]. The third-generation EGFR-TKI, osimertinib, was initially developed to target T790M mutation and has shown selective activity in targeting L858R or Exon19 del combined T790M [[155]23, [156]28–[157]30]. However, most NSCLC patients eventually become refractory to osimertinib through on-target or off-target mechanisms, which refer to the occurrence of new genetic alterations at the EGFR genes or genetic alterations and activations of other signaling pathways, respectively [[158]31]. According to studies in clinical trials and real-world registries, the resistant mechanisms vary when osimertinib is administered as first-line or second-line treatment. Studies have shown that in cases where osimertinib is administered as first- or second-line therapy, off-target mechanisms account for approximately 20% to 50%. These mechanisms include MET amplification, HER2 amplification, PIK3CA amplification, BRAF mutation, KRAS mutation, PIK3CA mutation, PTEN loss, oncogenic fusions, and histologic transformation [[159]2]. With emerging literature highlighting the impact of signaling pathways on acquired osimertinib resistance [[160]32–[161]34], we sought to determine biomarkers for osimertinib resistance. Our study analyzed six online GEO datasets and discovered that MRAS and HEG1 were significantly upregulated in osimertinib-resistant LUAD cells. Higher expression levels of MRAS and HEG1 were further confirmed in online data and tumor biopsies from LUAD patients with resistance to osimertinib in our hospital. We generated osimertinib resistant LUAD cell lines with elevated mRNA and protein levels of both MRAS and HEG1. We also proved a dose/time-dependent manner of MRAS and HEG1. Otherwise, lower expression of MRAS and HEG1 in tumor tissues than its paired paracancerous tissues in LUAD. These results above imply that MRAS and HEG1 expressions in LUAD are in osimertinib used-dependent manner until the resistance develops, which prompts us to conduct deep mechanism exploration. We still investigated the potential of MRAS and HEG1 in predicting the survival of LUAD patients. The LUAD patients with higher MRAS expression showed poorer survival probability (P < 0.05). On the other hand, the LUAD patients with higher HEG1 expression showed poorer survival probability than those with lower HEG1 expression, but the difference was not significant (P > 0.05). These indicate that MRAS might be both an effective osimertinib response and survival biomarker in LUAD. Although HEG1 is osimertinib resistance biomarker, the efficiency of HEG1 in predicting survival needs more studies to prove. It has been reported that MRAS and HEG1 are associated with cancers. MRAS belongs to the Ras subfamily of GTP-binding proteins. Its expression, gene polymorphisms, and mutation have been linked to various diseases, including coronary artery disease, painful temporomandibular disorders, Noonan syndrome, and cancers [[162]35–[163]39]. A previous study has shown that activated MRAS can initiate the epithelial-mesenchymal transition (EMT) and tumorigenesis of normal epithelial cells [[164]40]. MRAS interacts with various proteins, including Ras/Rap1 (RA)-guanine nucleotide exchange factor (GEF)-2, SCRIB, or SHOC2, to promote differentiation, aggregation, proliferation, and migration through MAPK and AKT signaling cascades [[165]41–[166]44]. MRAS also promotes estrogen-independent growth of breast cancer cells through the interaction with Rlf and the following Ral/JNK activation in an EGFR-dependent manner [[167]45]. However, MRAS coupled with G-protein gamma subunit 2 (GCN2) retards breast cancer cell proliferation by inhibiting ERK/Akt signaling [[168]46]. HEG1 regulates the concentric growth of the heart and recruits RAS-interacting protein 1 (Rasip1) to cell–cell junctions through direct binding, which maintains the stabilization of epithelial cell (EC) cell–cell junctions [[169]47, [170]48]. In hepatocellular carcinoma (HCC) cells, HEG1 promotes intrahepatic metastasis, lung metastasis, and EMT by inducing the expression and stability of β-catenin [[171]49]. The most recent studies have revealed the impact of MRAS and HEG1 on therapeutic resistance in cancers. In models of KRAS mutant lung and colorectal cancer models, KRAS G12C inhibitors suppress the Hippo/ yes-associated protein (YAP) signaling pathway by mislocalizing SCRIB in the cytoplasm. The activated YAP transports into the nucleus and initiates the transcription of MRAS. The increasing MRAS interacts with SHOC2 and PP1 and subsequently reactivates MEK/ERK signaling by dephosphorylating CRAF at Ser259, which ultimately drives adaptive resistance to KRAS G12C inhibitors [[172]39]. In HCC cells, the protein arginine methyltransferase 3 (PRMT3)/IGF2BP1 axis triggers the resistance to oxaliplatin by stabilizing the mRNA of HEG1 [[173]50]. Our study suggested that MRAS and HEG1 might be drivers of osimertinib resistance by activating signal pathways or altering the immune environment. It has been known that the regulation of the PI3K-Akt signaling pathway is a promising therapeutic tactic for osimertinib resistance, which may be the key point in HEG1-related resistance [[174]16]. An immunosuppressive environment was also identified at osimertinib resistance with decreased T cell infiltration and activation and increased macrophage infiltration and M2 polarization [[175]13]. Interestingly, our results also revealed that MRAS and HEG1 expressions interaction with M2 infiltration might result in osimertinib resistance. Moreover, it was found that MRAS expression is higher in LUAD patients with ALK mutation. A rare ALK rearrangement has been reported as a might incentive for acquired osimertinib resistance in a lung cancer patient [[176]51], which implies a positive association between ALK mutation and the upregulation of MRAS. Additionally, oncoPredict analysis showed that osimertinib-resistant LUAD with high levels of MRAS and HEG1 might respond to certain agents, including AZD1332 (a selective inhibitor of the tropomyosin receptor kinase receptors (TrKs)), dasatinib (a Src family kinases inhibitor), doramapimod (a pan-p38/MAPK inhibitor), WIKI4 (a Wnt/β-catenin inhibitor), AZD8186 (a selective PI3Kβ inhibitor), and cediranib (a selective VEGF signaling inhibitor). A study has reported that AXL/Cubomain-containing protein 1 (CDCP1)/SRC/AKT signaling axis is augmented upon osimertinib treatment and eventually confers to osimertinib resistance in LUAD cells [[177]52]. Consistently, our in vitro crystal violet cytotoxic assay showed that both parental NCI-H1975 and OR-1/2 clones, especially OR-2 clone, are comparatively susceptible to dasatinib, whose efficacy in unselected LUAD patients has been evaluated [[178]53]. Furthermore, Nie M et al. reported that acetylcholine (Ach) binds to muscarinic Ach receptors (mAchRs) and subsequently promotes the transcription of β-catenin-mediated WNT target genes, which ultimately causes cell survival and drug tolerance of drug-tolerant persister (DTP) NSCLC cells [[179]54]. In this study, OR-2 clone was more sensitive to the Wnt/β-catenin inhibitor WIKI4 than the parental NCI-H1975, whereas OR-1 clone exhibited insensitivity. RAS/MAPK signaling has been found hyperactivated in osimertinib and other EGFR TIK-resistant NSCLC cells [[180]55] and they key downstream of MRAS [[181]56]. Although IHC of MRAS and HEG1 showed that samples from LUAD patients with osimertinib resistance had higher expression levels of MRAS and HEG1 compared to samples from LUAD treatment naïve patients, the sample size is too small, and more clinical samples are needed to further verify. Additionally, the findings are mainly based on the existing data sets and few in vitro experiments, for example, overexpression of MRAS or HEG1 should be performed to further assess the association of MRAS or HEG1 with sensitivity to osimertinib in our future investigation. The cell line/patient-derived LUAD mouse models are also needed to strengthen the rationale and understanding of mechanisms and potential for targeting MRAS and HEG1. Overall, it is implied that elevating MRAS and HEG1 might predict the development of acquired osimertinib resistance, and the underlying mechanisms require further investigation. These findings suggest that certain drugs may be effective in treating lung cancer patients with high levels of specific proteins, and further research is needed to explore these treatments in more detail. Conclusion In summary, MRAS and HEG1 showed good prognostic value in predicting resistance to osimertinib and might play important roles in the resistance development in LUAD. More data from the real world are demanded to validate the accuracy and further studies are required to strengthen the rationale. Supplementary Information [182]12672_2024_1552_MOESM1_ESM.docx^ (57.6KB, docx) Supplementary Material 1. Table S8. List of HEG1 differential gene KEGG enrichment analyses. [183]12672_2024_1552_MOESM2_ESM.xlsx^ (35.4KB, xlsx) Supplementary Material 2. List of EGFR mutation status in the TCGA 454 dataset. [184]12672_2024_1552_MOESM3_ESM.tif^ (2.7MB, tif) Supplementary Material 3. Figure S1: The MRAS expression in LUAD patients with different gene mutations from the TCGA dataset; [185]12672_2024_1552_MOESM4_ESM.tif^ (2MB, tif) Supplementary Material 4. Figure S2: The HEG1 expression in LUAD patients with different gene mutations from the TCGA dataset. [186]12672_2024_1552_MOESM5_ESM.docx^ (506.5KB, docx) Supplementary Material 5. Table S1. List of genes analyzed individually for differences in the six datasets. [187]12672_2024_1552_MOESM6_ESM.docx^ (49.7KB, docx) Supplementary Material 6. Table S2. List of six data GO enrichment analyses. [188]12672_2024_1552_MOESM7_ESM.docx^ (33.7KB, docx) Supplementary Material 7. Table S3. List of six data GSEA enrichment analyses. [189]12672_2024_1552_MOESM8_ESM.docx^ (50.6KB, docx) Supplementary Material 8. Table S4. List of six data sets and concurrent analysis of variance for GO enrichment analysis. [190]12672_2024_1552_MOESM9_ESM.docx^ (48.8KB, docx) Supplementary Material 9. Table S5. List of GSEA enrichment analyses [191]12672_2024_1552_MOESM10_ESM.docx^ (19.9KB, docx) Supplementary Material 10. Table S6. Clinical information of NSCLC patients received tumor surgery/biopsies [192]12672_2024_1552_MOESM11_ESM.docx^ (78.4KB, docx) Supplementary Material 11. Table S7. List of MRAS differential gene KEGG enrichment analyses. Author contributions ML, BT, and LH conducted the experiments and drafted the manuscript. RX and PHH participated in the analysis and interpretation of data. LH, CX and QL designed the work. All authors read and approved the final manuscript. Funding This research was funded by Sichuan Province Science and Technology Support Program (No.2022NSFSC0691) and National Natural Science Foundation of China (No. 922591002). Data availability Data is provided within the manuscript or supplementary information files. Declarations Ethics approval and consent to participate The study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee for Medical Research and New Medical Technology of Sichuan Cancer Hospital (protocol code: SCCHEC-04-2022-002). Competing interests The authors declare no competing interests. Footnotes Publisher's Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Contributor Information Lanlin Hu, Email: hulanlin@jflab.ac.cn. Qiang Li, Email: liqiang@sichuancancer.org. References