International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Lung cancer is a globally prevalent malignant tumor and one of the most common causes of cancer-associated mortality. Lung cancer is classified into two main types: Non-small cell lung cancer (NSCLC) and small cell lung cancer, with NSCLC comprising ~85% of all cases (1). Among the different subtypes of NSCLC, lung adenocarcinoma (LUAD) has the highest incidence and prevalence. However, despite significant advances in surgery, chemotherapy and targeted therapy, the prognosis for patients with LUAD remains poor (2). Therefore, it is particularly urgent to develop reasonable treatment strategies to improve the therapeutic effect of lung adenocarcinoma. The invasion and metastasis of tumors is a complex regulatory process influenced by multiple factors, such as the activation of oncogenes, the inactivation of tumor suppressor genes and mutations in signaling pathways (3,4). Discovering molecular biological markers associated with lung adenocarcinoma not only helps to deepen the understanding of the disease, but also may provide guidance for the future diagnosis and treatment of lung adenocarcinoma.
The coagulation system is an inherent defense mechanism of the human body, which can be activated through the tissue factor pathway (extrinsic pathway) or the intrinsic pathway. Tumor cells express procoagulant factors, including tissue factor, which triggers the coagulation cascade and ultimately leads to the generation of thrombin (5). The activation of this coagulation and fibrinolysis system promotes the invasion, metastasis and angiogenesis of malignant tumor cells (6). Patients with malignant tumors often exhibit a hypercoagulable state, and have a higher risk of cancer-related thrombosis than healthy individuals (7), including deep vein thrombosis and pulmonary thromboembolism, which significantly reduces the survival rate and prognosis of patients (8). A large amount of experimental evidence supports that patients with malignant tumors often remain in a state of chronic hypercoagulability and hyperfibrinolysis, which is the main cause of the increased risk of thrombosis and bleeding (6). In particular, the prognosis of patients with cancer is closely related to the effectiveness of anticoagulant therapy. Specifically, patients with a better prognosis have an extended survival period after receiving anticoagulant treatment (9). The association between biomarkers related to coagulation disorders and the prognosis of various cancer types has previously been reported (10). The role of the coagulation cascade in the formation of the tumor microenvironment (TME) has also been emphasized in research (11). Although the associations between the coagulation system and multiple types of tumors, such as cutaneous melanoma (12), gastric cancer (13) and hepatocellular carcinoma (14), have been confirmed, the precise role of the coagulation system in LUAD has yet to be elucidated.
The present study aimed to develop a specific coagulation-related gene (CRG) prognostic model for LUAD, and to offer novel insights and approaches for the prognostic assessment, immune analysis and therapy of this disease. This model is intended to facilitate the investigation of tumor prognosis, immune cell infiltration, genetic mutations, tumor mutation burden (TMB) and drug responses.
LUAD transcriptome data and associated clinical information were sourced from The Cancer Genome Atlas (TCGA) official data portal (https://gdc portal.nci.nih.gov/). The dataset included 582 samples, comprising 524 tumor samples and 58 normal tissue samples. Following initial data screening, samples with missing or zero prognostic information were excluded, resulting in the selection of 507 tumor samples and 58 normal tissue samples for detailed analysis.
A total of 138 CRGs were extracted from the Molecular Signatures database (MsigDB; http://www.gsea-msigdb.org/gsea/msigdb) (Table SI). Differentially expressed genes (DEGs) were identified using the limma package (version 3.54.2) in R (https://cran.r-project.org) with gene set normalization applied. The screening criteria were set to an absolute value of the log2 fold change (|log2FC|)>1.5 and adjusted P<0.05. The DEGs were identified by comparing normal lung tissue samples with LUAD tissue samples from TCGA database. The DEGs were visualized using the ggplot2 package (version 3.4.2) and plotted as volcano plots. Comparative analysis was performed to identify coagulation-related DEGs specific to LUAD.
Following the identification of differentially expressed CRGs, enrichment analysis was performed using the clusterProfiler package (version 4.6.2). The analyses included three Gene Ontology (GO) categories, namely biological process (BP), cellular component (CC) and molecular function (MF), as well as Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. The aim of performing these analyses was to elucidate the molecular mechanisms associated with CRGs. The criteria for statistical significance were P<0.05 and q<0.05 in both the GO and KEGG analyses. Additionally, gene set enrichment analysis (GSEA) was conducted to assess model genes in high- and low-risk groups based on risk scores, with statistical significance defined as P<0.05 and false discovery rate (FDR) <0.05.
The survival package (version 3.5.5) was used to identify CRGs significantly associated with overall survival (OS; P<0.05). Univariate Cox regression analyses were performed to calculate the hazard ratio (HR). The data from TCGA were randomly divided into two essentially equal-sized groups: Training set (n=254) and test set (n=253). Least absolute shrinkage and selection operator (LASSO) regression analyses were then conducted using the glmnet package (version 4.1.7) to refine the gene selection. A risk model was developed using the training set data and validated using the overall and test set data. The risk score formula constructed was as follows:
The coefficient Coefi and expression level Expri represent the Cox regression coefficient and normalized expression level for each mRNA, respectively. Based on the risk score, samples were divided into high- and low-risk groups, with this stratification applied to the overall, test and training sets. To compare OS rates, Kaplan-Meier (KM) curves were generated using the survival package and survminer package (version 0.4.9). An analysis of progression-free survival (PFS) was also performed. In addition, gene expression differences between the high- and low-risk groups were visualized using the pheatmap package (version 1.0.12) Receiver operating characteristic (ROC) curves were constructed using the survival package, survminer package (version 0.4.9) and timeROC package (version 0.4) to evaluate the accuracy of the prognostic model and the 1-, 3- and 5-year survival rates. The protein expression of the four genes included in the prognostic model was assessed in normal and LUAD tissues using immunohistochemistry (IHC) data from the Human Protein Atlas (HPA; http://www.proteinatlas.org/). Finally, principal component analysis (PCA) was performed using the ggplot2 package in R to analyze the distribution differences between high- and low-risk groups, thereby assessing the accuracy and applicability of the model.
Univariate and multivariate Cox regression analyses were performed using the survival package (version 3.5.5), incorporating age, sex, tumor stage and TNM staging to compute risk scores as independent risk factors. Subsequently, a nomogram was constructed using the rms package (version 6.7.0), survcomp package (version 1.48.0), regplot package (version 1.1) and survival package. The accuracy of the model was further validated using calibration curves.
Initially, immune, ESTIMATE and stromal scoring was conducted for high- and low-risk groups of LUAD using the limma package and estimate package (version 1.0.13). CIBERSORT analysis was then performed using the limma package, with immune cell differences between the two groups visualized using the limma, reshape2 package (version 1.4.4) and ggpubr package (version 0.6.0). To assess immune function differences between the two groups, the limma GSVA package (version 1.46.0) and GSEABase package (version 1.60.0) were used, with results visualized using the reshape2 and ggpubr packages. Additionally, gene mutation information was obtained from TCGA, and the mutation frequencies of the most prevalent genes in the high- and low-risk groups were visualized using the maftools package (version 2.14.0). TMB was then compared between the two groups using the ggpubr and limma packages. The analysis was performed using ‘futime’ as the time variable and ‘fustat’ as the event variable. The variable TMB was used to determine the cut off value; patients with a TMB value lower than the cut off were assigned to the low TMB group, while those with a TMB value higher than the cut off were assigned to the high TMB group. The survival rates of these two groups are compared to evaluate their association with risk scores. P<0.05 was considered to indicate statistical significance.
The limma and ggpubr packages were used to visualize differences in TIDE scores between the high- and low-risk groups. Subsequently, drug sensitivity analysis was performed on the two groups using the limma package, oncoPredict package (version 0.2) and parallel package (version 4.2.2). The results of the drug sensitivity analysis were then visualized as half-maximal inhibitory concentration (IC50) values using the ggpubr and ggplot2 packages.
Three pairs of LUAD samples and adjacent non-cancerous tissues were collected from patients who underwent curative resection between November 2024 and January 2025 at Jinan Central Hospital (Jinan, China), including 2 female and 1 male patient, aged between 50–70 years. All tissue samples were verified by histopathological examination and stored in liquid nitrogen for subsequent reverse transcription-quantitative PCR (RT-qPCR) analysis. The inclusion criteria were as follows: i) Diagnosed with LUAD through pathological examination; ii) had not undergone any form of tumor treatment prior to surgery; and iii) had a complete clinicopathological record. The study excluded patients meeting the following criteria: i) Age <18 or >80 years; ii) pregnant or breastfeeding; iii) had autoimmune diseases or other types of malignancies; or iv) had received any antitumor therapy. The study protocol was approved by the Ethics Committee of Jinan Central Hospital (20241120026). All procedures were conducted in accordance with applicable guidelines and regulations. All patients signed the informed consent form.
The A549 human LUAD and BEAS-2B normal human lung epithelial cell lines were obtained from Wuhan Pricella Biotechnology Co., Ltd. The cells were cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin, and maintained at 37°C with 5% CO2. The medium was changed every 2 to 3 days. Total RNA was extracted from the cells and tissues using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and reverse transcribed into cDNA using a reverse transcription kit (Takara Biotechnology Co., Ltd.). The mRNA expression was quantified using RT-qPCR analysis using 10.0 µl SYBR™ Green (Hunan Accurate Bio-Medical Technology Co., Ltd.), 0.5 µl primer, 2 µl cDNA and 7.0 µl diethylpyrocarbonate-treated water. The thermocycling conditions were as follows: Initial denaturation at 95°C for 30 sec, followed by 40 cycles of 95°C for 5 sec, 55°C for 30 sec and 72°C for 30 sec. β-actin was used as an internal control. The mRNA expression levels were quantified using the 2−ΔΔCq method and normalized against the expression levels of β-actin (15). Each experiment was repeated three times. The sequences of the primers used are listed in Table I.
All statistical analyses were performed using R software (version 4.2.2) and GraphPad_Prism (version 8.0.2; Dotmatics). The Shapiro-Wilk test was used to assess whether the data in the high- and low-risk groups followed a normal distribution. The Mann-Whitney U test (Wilcoxon rank-sum test) was used when comparing the two risk groups, as well as unpaired LUAD and normal tissue from TCGA. Wilcoxon signed-rank analysis was used to compare paired LUAD and normal tissues from TCGA. Differences between paired primary tissues and between cell lines were analyzed using paired and unpaired t-tests, respectively. The log-rank test was employed to identify differences between KM curves. Univariate and multivariate Cox regression analyses were performed to identify prognostic risk factors associated with LUAD. P<0.05 was considered to indicate a statistically significant difference.
Fig. 1 provides an overview of the workflow for the present study. Gene expression data, clinical information and mutation data for patients with LUAD were collected from TCGA. By comparing the expression of 138 known CRGs between LUAD samples and adjacent normal tissue, 51 differentially expressed CRGs were identified (FDR <0.05, |log2 FC|>1.5), including 37 upregulated genes and 14 downregulated genes. Heatmaps and volcano plots were subsequently generated to visualize the expression patterns of these genes in normal and LUAD tissues (Fig. 2A and B). Further analysis verified that all 51 DEGs were associated with the coagulation process (Fig. 2C).
To explore the functions and potential mechanisms of the 51 DEGs, GO analysis was performed. The results reveal a significant enrichment of these CRGs in certain BPs, including ‘blood coagulation’, ‘coagulation’, ‘hemostasis’, ‘wound healing’ and ‘regulation of body fluid levels’. At the CC level, these genes are predominantly enriched in ‘collagen-containing extracellular matrix’, ‘secretory granule lumen’, ‘cytoplasmic vesicle lumen’ and ‘vesicle lumen’. In addition, MF analysis demonstrated that these genes are closely associated with ‘serine-type endopeptidase activity’, ‘serine-type peptidase activity’, ‘serine hydrolase activity’ and ‘endopeptidase activity’ (Fig. 2D). KEGG analysis revealed that these genes are significantly enriched in the ‘complement and coagulation cascades’ pathway (Fig. 2E). These findings reinforce the close association between coagulation processes and the development and progression of LUAD. Subsequently, a risk model based on the coagulation genes was created, to further confirm the association between LUAD and coagulation pathways.
As noted above, 51 DEGs were identified through differential expression analysis. Subsequently, univariate Cox regression analysis was performed, which revealed that 13 genes were significantly associated with OS. Of these, matrix metalloproteinase (MMP) 1 (HR, 1.105; P=0.044), MMP10 (HR, 1.119; P=0.040), cathepsin V (CTSV; HR=1.251; P=0.003), thrombin/coagulation factor II (F2; HR=1.324; P<0.001), FGA (HR, 1.073; P=0.017), KLK8 (HR, 1.212; P=0.027), GDA (HR, 1.233; P=0.011), HRG (HR, 1.618; P=0.025), HNF4A (HR, 1.190; P=0.020), F2RL2 (HR, 1.237; P=0.012), PROZ (HR, 1.430; P=0.037) and APOC3 (HR, 1.393; P=0.006) were classified as high-risk genes, while DPP4 (HR, 0.887; P=0.045) was classified as a low-risk gene (Fig. 3A). Subsequently, 507 eligible LUAD samples from TCGA were randomly divided into training (n=254) and test (n=253) sets in a 1:1 ratio (Table II). Based on analyses of age, sex, tumor stage and TNM staging, no significant differences between the training and test sets were identified. Within the training set, LASSO regression analysis was employed to identify the most prognostically relevant genes, which led to the construction of a four-gene prognostic model comprising the MMP1, MMP10, CTSV and F2 genes (Fig. 3B and C). The risk score formula for this model was as follows: Total risk score=(0.0837569167784308 × MMP1) + (0.0878985239886888 × MMP10) + (0.159310542027166 × CTSV + (0.258137401792528 × F2).
Table II.Clinical characteristics of the three sets of data randomly generated from The Cancer Genome Atlas database. |
Individuals were classified on the basis of the risk score calculated using this formula, with the average score as the cutoff. Individuals with scores above the average were categorized into the high-risk group, whereas those below the average were placed in the low-risk group. In the total set, 253 individuals were placed in the high-risk group, and 254 were categorized in the low-risk group. In the training set, the high- and low-risk groups each comprised 127 cases, whereas the test set contained 126 high-risk and 127 low-risk cases.
The Shapiro-Wilk test was employed to evaluate the normality of the data in the high- and low-risk groups. This test indicated that both datasets deviate significantly from the normal distribution (high risk, W=0.6951, P<2.2×10−16; low risk, W=0.96479, P=6.825×10−6). Since the data did not meet the prerequisite of a normal distribution required for ANOVA, a non-parametric test method was used, namely Wilcoxon's rank-sum test. The Wilcoxon test result (W=64,262, P<2.2×10−16) indicated that there was a significant difference in the distribution of risk scores between the high- and low-risk groups.
KM curves were subsequently generated to compare the OS and PFS between the high- and low-risk groups in the total, training and test sets. This analysis revealed that the OS of the high-risk group was significantly lower compared with that of the low-risk group in the total, training and test sets (Fig. 4A, C and E). Similarly, the PFS rates were significantly shorter in the high-risk group compared with the low-risk group in all three sets (Fig. 4B, D and F). There were significant statistical differences (P<0.05). Prognostic validation using heatmaps identified MMP1, MMP10, CTSV and F2 as consistently high-risk genes across the total set (Fig. 5A). Additionally, the associations between risk scores and survival times between the high- and low-risk groups in the total set were examined, which revealed a significant reduction in survival time for patients with high risk scores (Fig. 5B). Further analysis of the OS risk score distribution in the total set corroborated the accuracy of this model (Fig. 5C). Similar results were observed for validation of the training and test sets (Fig. 5D-I).
Univariate and multivariate Cox regression analyses were performed to determine whether each risk factor functioned as an independent prognostic indicator, distinct from other clinical characteristics. Initially, univariate Cox regression analysis was performed on the total set. The results revealed that age, sex, tumor stage, T stage, M stage, N stage and the risk score were significantly associated with prognosis. Specifically, tumor stage (HR, 1.595; P<0.001), T stage (HR, 1.618; P<0.001), N stage (HR, 1.732; P<0.001) and risk score (HR, 1.517; P<0.001) were significantly associated with prognosis, and M stage (HR, 1.858; P=0.034) was marginally significant (Fig. 6A). Subsequently, multivariate Cox regression analysis was performed on the total set to assess the independence of these factors. The results indicated that T stage (HR, 1.362; P=0.010) and risk score (HR, 1.415; P<0.001) were independent prognostic indicators. By contrast, tumor stage (HR, 1.246; P=0.301), sex (HR, 0.924; P=0.651), M stage (HR, 1.011; P=0.984) and N stage (HR, 1.251; P=0.216) did not have significant independent prognostic value (Fig. 6B). To evaluate the accuracy of the risk model, validation analysis was subsequently performed using ROC curves. The area under the ROC curve (AUC) values for 1-, 3- and 5-year survival were 0.687, 0.634 and 0.622 respectively, demonstrating the predictive capability of the model (Fig. 6C). In addition, ROC curve analysis for different clinical characteristics revealed that the predictive performance of the risk score (AUC, 0.687) was greater than that of age (AUC, 0.490), sex (AUC, 0.550), T stage (AUC, 0.652), M stage (AUC, 0.501) and N stage (AUC, 0.666) (Fig. 6D). Taken together, these findings confirm that the risk score model was capable of outperforming other clinical characteristics in terms of prognostic accuracy.
The present study employed PCA to further assess the accuracy of the prognostic model. The distribution of samples from patients with LUAD between the high- and low-risk groups was compared at the genome-wide level (Fig. 6E), in CRGs (Fig. 6F) and in genes included in the prognostic model (Fig. 6G). The results revealed significant differences in the genome-wide distributions, with the genes involved in the prognostic model exhibiting the highest level of discrimination. Additionally, a nomogram for patients with LUAD was constructed (Fig. 6H), which incorporated variables including age, sex, the TNM, T, N and M stages, and risk score. This nomogram was used to predict the 1-, 3- and 5-year survival rates of the patients. Calibration curves were then constructed (Fig. 6I), which demonstrated that the nomogram possessed high predictive accuracy.
To investigate the associations between various patient characteristics and risk scores in greater depth, heat maps were constructed to assess the associations between gene expression data derived from the model and various clinical and pathological characteristics (Fig. 7A). This analysis reaffirmed MMP1, MMP10, CTSV and F2 as high-risk genes, and highlighted significant correlations between the risk score and various clinical characteristics, particularly N stage (P<0.01), tumor stage (P<0.01) and survival time (P<0.01). Furthermore, KM analyses were performed between the high- and low-risk groups within clinical characteristic sub-groups. In patients aged <65 or ≥65 years, of either sex, or with tumor stage I–II, T1-T2, T3-T4, N0 or M0, the prognostic accuracy of the risk model in terms of distinguishing between the high- and low-risk groups was significant (Fig. 7B-M).
Based on the results obtained, the prognostic model constructed from the genes MMP1, MMP10, CTSV and F2 demonstrated high accuracy in terms of distinguishing between high- and low-risk groups for patients with LUAD. GO enrichment analysis revealed that these genes are primarily associated with ‘sulfur compound binding’, ‘glycosaminoglycan binding’, ‘serine hydrolase activity’, ‘serine-type peptidase activity’ and ‘serine-type endopeptidase activity’ at the BP level. At the CC level, these genes were found to be significantly enriched in ‘collagen-containing extracellular matrix’, ‘apical part of cell’, ‘vesicle lumen’, ‘cytoplasmic vesicle lumen’ and ‘secretory granule lumen’. The analysis of MF identified associations with ‘epidermis development’, ‘negative regulation of proteolysis’, ‘skin development’ and ‘antimicrobial humoral response’ Fig. 8A and B). Furthermore, KEGG pathway enrichment analysis revealed that these genes are primarily associated with ‘neuroactive ligand-receptor interaction’ (Fig. 8C and D). To further differentiate between the high- and low-risk groups, GSEA was performed. This led to the identification of five significant pathways based on P-value ranking. The GSEA analysis revealed that the high-risk group was significantly enriched in the pathways ‘cell cycle’, ‘neuroactive ligand-receptor interaction’, ‘pentose and gluconate interconversion’, ‘porphyrin and chlorophyll metabolism’ and ‘starch and sucrose metabolism’. By contrast, the low-risk group was enriched in the pathways ‘allograft rejection’, ‘asthma’, ‘autoimmune thyroid disease’, ‘ribosome’ and ‘systemic lupus erythematosus’ (Fig. 8E and F).
In terms of the TME, immune, ESTIMATE and stromal scores were analyzed. The results revealed no significant differences in TME scores between the high- and low-risk groups (Fig. 9A). However, further analysis detected significant differences in immune cell infiltration between the two groups. The low-risk group exhibited significantly higher proportions of resting CD4+ memory T cells, monocytes, M2 macrophages, resting dendritic cells and resting mast cells. By contrast, the high-risk group exhibited significantly higher proportions of activated CD4+ memory T cells, resting natural killer (NK) cells, M0 macrophages, M1 macrophages, activated mast cells and neutrophils (Fig. 9B and C). In addition, an analysis of immune function revealed significant differences in the expression of immune checkpoint molecules: The low-risk group was found to have higher proportions of B cells, human leukocyte antigens, interstitial dendritic cells, mast cells, helper T cells and type II interferon (IFN) responses. By contrast, the high-risk group had higher proportions of chemokine receptors, macrophages, MHC I molecules, NK cells, parainflammatory cells and type I IFN responses (Fig. 9D).
Somatic mutation data were initially collected from TCGA database for patients with LUAD. Among the 171 samples in the high-risk group, 158 samples (92.4%) exhibited somatic mutations, whereas 137 of 154 samples (88.96%) in the low-risk group had somatic mutations. Further analysis revealed that the incidence of gene mutations in the high-risk group was higher compared with that in the low-risk group. The top 15 genes with the highest mutation rates were tumor protein P53 (TP53), titin (TTN), mucin 16 (MUC16), CSMD3, RYR2, LRP1B, ZFHX4, USH2A, KRAS, XIRP2, FLG, SPTA1, NAV3, ZNF536 and COL11A1 (Fig. 10A and B). TMB analysis further showed that the TMB was higher in the high-risk group compared with the low-risk group (Fig. 10C). According to the TMB value (cut off, 7.63), patients were divided into low- and high-TMB groups. KM survival analysis demonstrated that the OS rate of the high-TMB group was significantly higher compared with that of the low-TMB group (Fig. 10D). Comparative analysis for different combinations of risk and TMB levels revealed that the high risk + low TMB group had the poorest prognosis, whereas the low risk + high TMB group had the best prognosis, with statistically significant differences observed among the groups (Fig. 10E). To assess immune escape, TIDE scores were calculated, where a higher TIDE score is indicative of a greater likelihood of immune escape. The results obtained showed that the TIDE score of the high-risk group was significantly higher compared with that of the low-risk group (Fig. 10F). Further research, however, is necessary to understand the differences in drug treatment responses between the high- and low-risk groups.
Differences in drug sensitivity between the high- and low-risk patient groups were investigated. Analysis of the drug-sensitivity data revealed significant differences in IC50 values between the two groups. Specifically, the high-risk group demonstrated greater sensitivity to the following drugs: BI-2536 (P=1.1×10−4; Fig. 11A), dasatinib (P=3.3×10−6; Fig. 11B), PD0325901 (P=7.3×10−8; Fig. 11C), SCH772984 (P=3.4×10−16; Fig. 11D) and trametinib (P=5.2×10−7; Fig. 11E). By contrast, the low-risk group exhibited higher sensitivity to BMS-754807 (P=9.99×10−8; Fig. 11F), doramapimod (P=4.0×10−10; Fig. 11G), JAK1-8709 (P=5.9×10−4; Fig. 11H), ribociclib (P=2.5×10−5; Fig. 11I) and SB216763 (P=1.4×10−5; Fig. 11J).
Subsequently, samples from TCGA database were utilized to assess the expression levels of MMP1, MMP10, CTSV and F2 in normal and LUAD tissues. Analysis using the Mann-Whitney U test revealed that the expression of these genes in LUAD tissue was significantly higher compared with that in normal lung tissue (Fig. 12A-D). Wilcoxon signed-rank analysis further confirmed significant differences in the expression of these genes between the two types of tissues (Fig. 12E-H), supporting the high-risk status of these genes. To validate these findings, IHC results from the HPA database were compared between LUAD and normal lung tissues. The protein expression levels of MMP10, CTSV and F2 were observed to be markedly higher in LUAD tissues compared with normal tissues (Fig. 13A). The HPA database did not include IHC data for MMP1, so its protein expression level could not be assessed. Additionally, RT-qPCR experiments were performed to further verify the accuracy of the CRG diagnostic model. The mRNA expression levels of MMP10, CTSV, MMP1 and F2 were found to be upregulated in primary tumor tissues compared with normal tissue (Fig. 13B). Furthermore, the mRNA levels of MMP10, CTSV and F2 were found to be significantly higher in the LUAD cell line A549 compared with the BEAS-2B cell line (Fig. 13C). Taken together, these findings corroborate the bioinformatics analysis results from TCGA database.
The coagulation system maintains a delicate balance between clotting and bleeding under physiological conditions, although this balance is disrupted in disease states (16). The systemic activation of hemostasis and thrombosis in patients with malignancies has been shown to be closely associated with tumor occurrence, progression and metastasis (17,18). A review of numerous studies revealed that approximately half of patients with malignancies exhibit coagulation abnormalities, and this proportion rises to as high as 90% in patients with metastasis (19). There is a close interplay between tumor activation and the coagulation system, which is crucial for advancing our understanding of cancer biology and cancer-associated thrombosis, particularly the role of tissue factor. The coagulation system plays an important role in tumor formation, cell migration, vascular invasion, extravasation and distant metastasis in LUAD (20). Therefore, coagulation-associated biomarkers hold great potential for prognostication.
In the present study, four key prognostic CRGs, namely MMP1, MMP10, CTSV and F2, were identified and used to construct a prognostic signature for patients with LUAD. This predictive model may serve as a valuable tool in the differential diagnosis of lung cancer and its mimickers, such as those reported by Neacşu et al (21). However, only a handful of studies have highlighted the roles played by these four genes during the progression of LUAD. MMP1, which was recognized as a high-risk gene in the present study, is mainly located in the mitochondria, where it suppresses mitochondrial respiration, increases hypoxia-inducible factor-1 expression, and reduces the production of reactive oxygen species (22). MMP1 has been shown to promote the progression of several types of tumors, including cutaneous squamous cell carcinoma, hepatocellular carcinoma and colorectal cancer (23–25). In addition, Guo et al (26) found that wogonin inhibits lung cancer progression by regulating MMP1 and the PI3K/Akt signaling pathway. MMP10 (27), another high-risk gene, is involved in various pathological and physiological processes, including colitis-associated cancer, hepatocellular carcinoma and breast cancer (28–30). In addition, previous evidence has shown that MMP10 regulates NSCLC radiosensitivity via the DNA damage repair pathway (31). Therefore, MMP10 may play an important role in NSCLC and could help determine indications for radiotherapy. The third high-risk gene, CTSV, is a cysteine protease, with an active form present in the cytoplasm, nucleus and plasma membrane. It contributes to the degradation of specific components of the extracellular matrix, and has been implicated in the progression of various types of cancer (32–34). In addition, Zhu et al (35) found that CSTV promotes the development of lung cancer by modulating the immunosuppressive microenvironment and cleaving adhesion molecules. The fourth high-risk gene, F2, encodes the serine protease thrombin. Elevated levels of thrombin not only promote blood coagulation but also facilitate tumor growth and metastasis. Therefore, thrombin and its precursor factors have emerged as potential targets for the treatment of malignant tumors and tumor-associated thrombosis (36). Furthermore, F2 has also been found to be associated with immune infiltration and poor prognosis in various types of tumors, including LUAD and breast cancer (37). The present study indicates that the four genes are all upregulated in LUAD tissues and function as tumor oncogenes, which is consistent with previous findings. Furthermore, an innovative predictive model incorporating these four genes has been developed in the present study to assess the prognosis of LUAD. This model provides valuable insights for the prognosis assessment and treatment of LUAD, and potentially other tumor types.
Alterations in the TME are critical in LUAD progression (38). The quantity and characteristics of tumor-infiltrating lymphocytes serve as important predictors of prognosis and treatment response in cancers such as oral squamous cell carcinoma and epithelial ovarian cancer (39–41). In the present study, significant differences in immune cell composition were observed between the high- and low-risk groups. Specifically, the low-risk group exhibited higher proportions of resting CD4+ memory T cells, monocytes, M2 macrophages, resting dendritic cells and resting mast cells. By contrast, the high-risk group exhibited higher proportions of activated CD4+ memory T cells, resting NK cells, M0 macrophages, M1 macrophages, activated mast cells and neutrophils. A number of studies have investigated the antitumor functions of CD4+ T memory cells (42–44), and their presence has been significantly associated with an improved prognosis in various types of cancer (45–47). The role of NK cells in antitumor activity, exerted through their cytotoxic effects, has been established (48). However, M0 macrophages have been identified as independent predictive factors of poor outcomes in patients with pancreatic ductal adenocarcinoma (49). Previous studies have reported similar findings to those of the present study, supporting the accuracy of the current model (50,51). In the present study, the low-risk group exhibited enhanced immune cell infiltration and immune function activation, indicative of a more responsive and active antitumor immune system. By contrast, the high-risk group was indicated to have a more immunosuppressive TME, leading to a weaker immune response and potential resistance to immunotherapy. These differences in immune profiles and TME characteristics highlight the greater potential of the low-risk group for improved treatment outcomes, particularly with immunotherapy. These insights into the associations between risk groups, TME and immune function are valuable for understanding LUAD immunology and may assist in the development of personalized treatment strategies.
In the present study, the mutation rate in the high-risk group was significantly higher compared with that in the low-risk group. Notably, the mutation frequencies of TP53, TTN and MUC16 were significantly different between the two groups. Mutations in the TP53 tumor suppressor gene are the most common in human malignancies, particularly in NSCLC (52). In the present study, the analysis of 227 patients with LUAD revealed that differences in mutation rates between the high- and low-risk groups were primarily attributable to TP53 gene mutations. These mutations are typically associated with extensive tumor invasion and poor prognosis (53). Notably, several drugs have been confirmed to treat malignancies by inhibiting the TP53 signaling pathway (54). Similarly, TTN plays a crucial role in the development and progression of various tumors. In LUAD, the long non-coding RNA TTN-AS1 has been shown to promote cell invasion and migration (55). In the present study, the mutation rate of MUC16 was significantly higher in the low-risk group than in the high-risk group, suggesting that MUC16 may serve as a protective mutation in LUAD, a conclusion supported by findings in other tumor types. For example, in ovarian cancer, high expression of MUC16 has been demonstrated to have a significant antitumor effect (56). In addition, MUC16 is considered a potential therapeutic target in pancreatic cancer (57). An observational study of patients with LUAD revealed that MUC16 is significantly associated with a higher TMB and improved clinical prognosis (58), findings that are consistent with those of the present study. However, the specific mechanism of action of MUC16 requires further investigation. Genetic mutations serve as critical targets for the treatment of malignancies. In the present study, mutations in TP53, TTN and MUC16 were particularly important when comparing the high- and low-risk LUAD groups. Specifically, TP53 and TTN mutations play important roles in tumor progression and invasion in LUAD, whereas MUC16 may act as a protective mutation, although this warrants further investigation. Collectively, these findings offer novel insights for targeted therapies in patients with LUAD, and potential strategies for improving patient prognosis.
Immune checkpoint inhibitors (ICIs) have emerged as efficacious immunotherapies for different types of cancer. By targeting molecules such as programmed death 1 (PD-1)/PD-ligand 1 (PD-L1) and cytotoxic T-lymphocyte associated protein 4, they boost antitumor responses and counteract tumor immune evasion (59,60). TMB is a key predictive factor of ICI response (61–63), with higher TMB being associated with improved efficacy of PD-1/PD-L1 inhibitors (64). An association between high TMB and improved ICI treatment outcomes has also been observed (65). In addition, the TIDE score has been shown to be useful for assessing the response to immunotherapy and the immune evasion capacity of tumors (66). In the present study, the TIDE score in the high-risk group was significantly higher compared with that in the low-risk group, suggesting a stronger tendency for immune evasion in high-risk patients, which may affect treatment outcomes. In addition, the patients with LUAD and high TMB exhibited longer OS times. However, the cross-validation of TMB with risk scores revealed that patients in the high-risk group with low TMB had significantly poorer OS outcomes. These findings underscore the importance of considering TMB and its interaction with risk scores when evaluating the immune response and predicting treatment outcomes in LUAD. This information is vital for guiding personalized treatment decisions, and for optimizing patient prognosis in the context of immunotherapy.
In the analysis of drug sensitivity, the response of patients with LUAD in different risk groups to anticancer agents was evaluated, which provides insights into personalized treatment strategies. The results demonstrated that the IC50 values of BI-2536 (67), dasatinib (68), PD0325901 (69), SCH772984 (70) and trametinib (71) were significantly lower in the high-risk group than in the low-risk group, suggesting that patients in the high-risk group may exhibit more favorable therapeutic responses to these agents. By contrast, BMS-754807 (72), doramapimod (73), JAK1_8709 (74), ribociclib (75) and SB216763 (76) exhibited superior efficacy in the low-risk group. These findings provide novel insights into the pharmacotherapy for patients with high- and low-risk LUAD. By matching the most suitable anticancer drugs with different patient types, the present study has the potential to facilitate more precise and effective treatment strategies. Although these drugs are suggested to have considerable therapeutic potential, the specific mechanisms underlying their action and actual efficacy in LUAD require further investigation.
The present study had certain limitations. The data were mainly derived from TCGA and lacked validation with external datasets, which may introduce some bias. In addition, the data did not follow a normal distribution. In subsequent research, a more detailed analysis of data distribution characteristics will be conducted, to enhance the scientific rigor of the research methods and the reliability of the results. Furthermore, although RT-qPCR analysis confirmed that MMP1, MMP10, F2 and CTSV are highly expressed in LUAD tissues, the sample size was limited. In future studies, it is planned to increase the sample size, conduct more comprehensive clinical validations, and perform in vivo and in vitro experiments to further evaluate the performance of the present model as an independent predictor of survival in real-world clinical settings.
In summary, the present study developed a new CRG-based model, by integrating bioinformatics analyses of immune-related indicators and tumor mutations to predict potential drug sensitivities across different patient subgroups. This model serves as a novel index for predicting treatment efficacy and prognosis in patients with LUAD. Future validation of the model using primary data is essential to confirm its reliability, accuracy and practicality in a clinical setting. This will involve conducting large-scale, multi-center clinical trials to test the performance of the model in diverse patient populations and real-world scenarios, and indicate its potential to guide individualized treatment strategies.
Not applicable.
This study was financially supported by the Shandong Province Medical and Health Development Plan (grant no. 202304020860) and Jinan Municipal Health Commission Science and Technology Development Plan Project (grant no. 2024302003).
The data generated in the present study may be requested from the corresponding author.
LZ and XS conceived and designed the research. JL and XG prepared the manuscript and figures. LL performed the acquisition and analysis of bioinformatic data. XG, JL, YH and HZ performed experiments, LZ and XS revised the manuscript. LZ and XS confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.
Human lung cancer tissues and paired adjacent noncancerous lung tissues were obtained from three patients undergoing lung resection. The study was approved by the Institutional Review Board (IRB) of Jinan Central Hospital (approval no. 20241120026). All subjects signed an IRB-approved informed consent form prior to enrollment in the study.
Not applicable.
The authors declare that they have no competing interests.
|
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ Jr, Wu YL and Paz-Ares L: Lung cancer: Current therapies and new targeted treatments. Lancet. 389:299–311. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang M, Yang C, Dong W, Zhao Y, Chen N and Gao C: Expression patterns and prognostic role of m6A RNA methylation regulators in Non-small Cell Lung Cancer. Cell Mol Biol (Noisy-le-grand). 70:67–72. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng X, Qiu L, Huang Y, Cheng R, Huang S, Xu K, Cai W, Deng Y, Wang W, Zhong X, et al: Exploring the molecular and Immune-landscape of lung cancer associated with cystic airspaces. Mol Immunol. 168:75–88. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Hamza MS and Mousa SA: Cancer-Associated Thrombosis: Risk factors, molecular mechanisms, future management. Clin Appl Thromb Hemost. 26:10760296209542822020. View Article : Google Scholar : PubMed/NCBI | |
|
Korte W: Changes of the coagulation and fibrinolysis system in malignancy: Their possible impact on future diagnostic and therapeutic procedures. Clin Chem Lab Med. 38:679–692. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Mulder FI, Horváth-Puhó E, van Es N, van Laarhoven HWM, Pedersen L, Moik F, Ay C, Büller HR and Sørensen HT: Venous thromboembolism in cancer patients: A Population-based cohort study. Blood. 137:1959–1969. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Tatsumi K: The pathogenesis of Cancer-associated thrombosis. Int J Hematol. 119:495–504. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Tieken C and Versteeg HH: Anticoagulants versus cancer. Thromb Res. 140 (Suppl 1):S148–S153. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Kawai K and Watanabe T: Colorectal cancer and hypercoagulability. Surg Today. 44:797–803. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Saidak Z, Soudet S, Lottin M, Salle V, Sevestre MA, Clatot F and Galmiche A: A pan-cancer analysis of the human tumor coagulome and its link to the tumor immune microenvironment. Cancer Immunol Immunother. 70:923–933. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Song B, Chi H, Peng G, Song Y, Cui Z, Zhu Y, Chen G, Wu J, Liu W, Dong C, et al: Characterization of coagulation-related gene signature to predict prognosis and tumor immune microenvironment in skin cutaneous melanoma. Front Oncol. 12:9752552022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang B, Zou D, Wang N, Wang H, Zhang T, Gao L, Ma C, Zheng P, Gu B, Li X, et al: Construction and validation of a novel Coagulation-related 7-gene prognostic signature for gastric cancer. Front Genet. 13:9576552022. View Article : Google Scholar : PubMed/NCBI | |
|
Yang WX, Gao HW, Cui JB, Zhang AA, Wang FF, Xie JQ, Lu MH and You CG: Development and validation of a coagulation-related genes prognostic model for hepatocellular carcinoma. BMC Bioinformatics. 24:892023. View Article : Google Scholar : PubMed/NCBI | |
|
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Palta S, Saroa R and Palta A: Overview of the coagulation system. Indian J Anaesth. 58:515–523. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Langer F and Bokemeyer C: Crosstalk between cancer and haemostasis. Implications for cancer biology and cancer-associated thrombosis with focus on tissue factor. Hamostaseologie. 32:95–104. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Lima LG and Monteiro RQ: Activation of blood coagulation in cancer: Implications for tumour progression. Biosci Rep. 33:2013. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y and Wei S: Advances on mechanisms of coagulation with non-small cell lung cancer. Zhongguo Fei Ai Za Zhi. 16:676–680. 2013.(In Chinese). PubMed/NCBI | |
|
Du H and Chen J: Occurrence of venous thromboembolism in patients with lung cancer and its anticoagulant therapy. Zhongguo Fei Ai Za Zhi. 21:784–789. 2018.(In Chinese). PubMed/NCBI | |
|
Neacşu F, Vârban A, Simion G, Şurghie R, Pătraşcu OM, Sajin M, Dumitru M and Vrînceanu D: Lung cancer mimickers-a case series of seven patients and review of the literature. Rom J Morphol Embryol. 62:697–704. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Herrera I, Cisneros J, Maldonado M, Ramírez R, Ortiz-Quintero B, Anso E, Chandel NS, Selman M and Pardo A: Matrix metalloproteinase (MMP)-1 induces lung alveolar epithelial cell migration and proliferation, protects from apoptosis, and represses mitochondrial oxygen consumption. J Biol Chem. 288:25964–25975. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang S, Liu H, Zhang J, Zhang F, Fan J and Liu Y: MMP1 regulated by NEAT1/miR-361-5p axis facilitates the proliferation and migration of cutaneous squamous cell carcinoma via the activation of Wnt pathway. Cancer Biol Ther. 22:381–391. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu H, Lan T, Li H, Xu L, Chen X, Liao H, Chen X, Du J, Cai Y, Wang J, et al: Circular RNA circDLC1 inhibits MMP1-mediated liver cancer progression via interaction with HuR. Theranostics. 11:1396–1411. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang K, Zheng J, Yu J, Wu Y, Guo J, Xu Z and Sun X: Knockdown of MMP-1 inhibits the progression of colorectal cancer by suppressing the PI3K/Akt/c-myc signaling pathway and EMT. Oncol Rep. 43:1103–1112. 2020.PubMed/NCBI | |
|
Guo J, Jin G, Hu Y, Zhao Z, Nan F, Hu X, Hu Y and Han Q: Wogonin restrains the malignant progression of lung cancer through modulating MMP1 and PI3K/AKT signaling pathway. Protein Pept Lett. 30:25–34. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Rempe RG, Hartz AMS and Bauer B: Matrix metalloproteinases in the brain and Blood-brain barrier: Versatile breakers and makers. J Cereb Blood Flow Metab. 36:1481–1507. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Y, Zhou Y, Chen J, Yang J, Yuan Y and Wu W: Exosomal lncRNA SNHG12 promotes angiogenesis and breast cancer progression. Breast Cancer. 31:607–620. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Faraj Shaglouf LH, Ranjpour M, Wajid S and Jain SK: Elevated expression of cellular SYNE1, MMP10, and GTPase1 and their regulatory role in hepatocellular carcinoma progression. Protoplasma. 257:157–167. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
He L, Kang Q, Chan KI, Zhang Y, Zhong Z and Tan W: The immunomodulatory role of matrix metalloproteinases in colitis-associated cancer. Front Immunol. 13:10939902022. View Article : Google Scholar : PubMed/NCBI | |
|
Bi Y, Cao K, Wang Y, Yang W, Ma N, Lei X and Chen Y: Radiosensitivity in non-small-cell lung cancer by MMP10 through the DNA damage repair pathway. J Oncol. 2023:56368522023. View Article : Google Scholar : PubMed/NCBI | |
|
Lin CL, Hung TW, Ying TH, Lin CJ, Hsieh YH and Chen CM: Praeruptorin B mitigates the metastatic ability of human renal carcinoma cells through targeting CTSC and CTSV Expression. Int J Mol Sci. 21:29192020. View Article : Google Scholar : PubMed/NCBI | |
|
Liu J, Zhang W, Wang Z, Wang Y, Li T, Wang Y, Ding J and Ning B: Cathepsin V is correlated with the prognosis and tumor microenvironment in liver cancer. Mol Carcinog. 63:400–416. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Sereesongsaeng N, Burrows JF, Scott CJ, Brix K and Burden RE: Cathepsin V regulates cell cycle progression and histone stability in the nucleus of breast cancer cells. Front Pharmacol. 14:12714352023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu L, Zeng Q, Wang J, Deng F and Jin S: Cathepsin V drives lung cancer progression by shaping the immunosuppressive environment and adhesion molecules cleavage. Aging (Albany NY). 15:13961–13979. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu L, Li J, Fan C, Wen M, Li C, Sun W and Wang W: Construction of a new Immune-related competing endogenous rna network with prognostic value in lung adenocarcinoma. Mol Biotechnol. 66:300–310. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Xu S, Tuo QZ, Meng J, Wu XL, Li CL and Lei P: Thrombin induces ferroptosis in triple-negative breast cancer through the cPLA2α/ACSL4 signaling pathway. Transl Oncol. 39:1018172024. View Article : Google Scholar : PubMed/NCBI | |
|
Wu J, Li L, Zhang H, Zhao Y, Zhang H, Wu S and Xu B: A risk model developed based on tumor microenvironment predicts overall survival and associates with tumor immunity of patients with lung adenocarcinoma. Oncogene. 40:4413–4424. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hwang C, Lee SJ, Lee JH, Kim KH, Suh DS, Kwon BS and Choi KU: Stromal tumor-infiltrating lymphocytes evaluated on H&E-stained slides are an independent prognostic factor in epithelial ovarian cancer and ovarian serous carcinoma. Oncol Lett. 17:4557–4565. 2019.PubMed/NCBI | |
|
Paijens ST, Vledder A, de Bruyn M and Nijman HW: Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 18:842–859. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Shaban M, Khurram SA, Fraz MM, Alsubaie N, Masood I, Mushtaq S, Hassan M, Loya A and Rajpoot NM: A Novel digital score for abundance of tumour infiltrating lymphocytes predicts disease free survival in oral squamous cell carcinoma. Sci Rep. 9:133412019. View Article : Google Scholar : PubMed/NCBI | |
|
Choi A, Jung YW and Choi H: The extrinsic factors important to the homeostasis of Allergen-specific memory CD4 T cells. Front Immunol. 13:10808552022. View Article : Google Scholar : PubMed/NCBI | |
|
Kawabe T and Sher A: Memory-phenotype CD4+ T cells: A naturally arising T lymphocyte population possessing innate immune function. Int Immunol. 34:189–196. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Krueger PD, Osum KC and Jenkins MK: CD4+ memory T-cell formation during type 1 immune responses. Cold Spring Harb Perspect Biol. 13:a0381412021. View Article : Google Scholar : PubMed/NCBI | |
|
Okła K, Farber DL and Zou W: Tissue-resident memory T cells in tumor immunity and immunotherapy. J Exp Med. 218:e202016052021. View Article : Google Scholar : PubMed/NCBI | |
|
Park SL, Gebhardt T and Mackay LK: Tissue-resident memory T cells in cancer immunosurveillance. Trends Immunol. 40:735–747. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Zhu Z, Modrak S and Little A: Tissue-resident memory CD4+ T cells play a dominant role in the initiation of antitumor immunity. J Immunol. 208:2837–2846. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Parrish-Novak J, Dillon SR, Nelson A, Hammond A, Sprecher C, Gross JA, Johnston J, Madden K, Xu W, West J, et al: Interleukin 21 and its receptor are involved in NK cell expansion and regulation of lymphocyte function. Nature. 408:57–63. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Xu C, Sui S, Shang Y, Yu Z, Han J, Zhang G, Ntim M, Hu M, Gong P, Chen H and Zhang X: The landscape of immune cell infiltration and its clinical implications of pancreatic ductal adenocarcinoma. J Adv Res. 24:139–148. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chang J, Wu H, Wu J, Liu M, Zhang W, Hu Y, Zhang X, Xu J, Li L, Yu P and Zhu J: Constructing a novel mitochondrial-related gene signature for evaluating the tumor immune microenvironment and predicting survival in stomach adenocarcinoma. J Transl Med. 21:1912023. View Article : Google Scholar : PubMed/NCBI | |
|
Sun X, Li J, Gao X, Huang Y, Pang Z, Lv L, Li H, Liu H and Zhu L: Disulfidptosis-related lncRNA prognosis model to predict survival therapeutic response prediction in lung adenocarcinoma. Oncol Lett. 28:3422024. View Article : Google Scholar : PubMed/NCBI | |
|
Mogi A and Kuwano H: TP53 mutations in nonsmall cell lung cancer. J Biomed Biotechnol. 2011:5839292011.PubMed/NCBI | |
|
Perez-Rivas LG, Simon J, Albani A, Tang S, Roeber S, Assié G, Deutschbein T, Fassnacht M, Gadelha MR, Hermus AR, et al: TP53 mutations in functional corticotroph tumors are linked to invasion and worse clinical outcome. Acta Neuropathol Commun. 10:1392022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen H, Deng J, Hou TW and Shan YQ: Villosol reverses 5-FU resistance in colorectal cancer by inhibiting the CDKN2A gene regulated TP53-PI3K/Akt signaling axis. J Ethnopharmacol. 325:1179072024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhong Y, Wang J, Lv W, Xu J, Mei S and Shan A: LncRNA TTN-AS1 drives invasion and migration of lung adenocarcinoma cells via modulation of miR-4677-3p/ZEB1 axis. J Cell Biochem. 120:17131–17141. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Mack KN, Samuels ZV, Carter LM, Viray TD, Mandleywala K, Brooks CL, Hollingsworth MA, Radhakrishnan P and Lewis JS: Interrogating the theranostic capacity of a MUC16-Targeted antibody for ovarian cancer. J Nucl Med. 65:580–585. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Aguilar EN, Sagar S, Murray BR, Rajesh C, Lei EK, Michaud SA, Goodlett DR, Caffrey TC, Grandgenett PM, Swanson B, et al: Structural basis for multivalent MUC16 recognition and robust Anti-pancreatic cancer activity of humanized antibody AR9.6. Mol Cancer Ther. 23:836–853. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu T, Wu L, Liu J, Chen H, Zhu B, Qiao D, Zhu Y, Liu T, Chen Q and Hu A: Comprehensive characterization of MUC16 mutations in lung adenocarcinoma for immunotherapies and prognosis: An observational study. Medicine (Baltimore). 102:e354812023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu C, Zheng S, Wang Z, Wang S, Wang X, Yang L, Xu H, Cao Z, Feng X, Xue Q, et al: KRAS-G12D mutation drives immune suppression and the primary resistance of anti-PD-1/PD-L1 immunotherapy in non-small cell lung cancer. Cancer Commun (Lond). 42:828–847. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wen Y, Tang F, Tu C, Hornicek F, Duan Z and Min L: Immune checkpoints in osteosarcoma: Recent advances and therapeutic potential. Cancer Lett. 547:2158872022. View Article : Google Scholar : PubMed/NCBI | |
|
Jardim DL, Goodman A, de Melo Gagliato D and Kurzrock R: The challenges of tumor mutational burden as an immunotherapy biomarker. Cancer Cell. 39:154–173. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu L, Bai X, Wang J, Tang XR, Wu DH, Du SS, Du XJ, Zhang YW, Zhu HB, Fang Y, et al: Combination of TMB and CNA stratifies prognostic and predictive responses to immunotherapy across metastatic cancer. Clin Cancer Res. 25:7413–7423. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, Barron DA, Zehir A, Jordan EJ, Omuro A, et al: Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 51:202–206. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Ricciuti B, Wang X, Alessi JV, Rizvi H, Mahadevan NR, Li YY, Polio A, Lindsay J, Umeton R, Sinha R, et al: Association of high tumor mutation burden in Non-small cell lung cancers with increased immune infiltration and improved clinical outcomes of PD-L1 blockade across PD-L1 expression levels. JAMA Oncol. 8:1160–1168. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Cao D, Xu H, Xu X, Guo T and Ge W: High tumor mutation burden predicts better efficacy of immunotherapy: A pooled analysis of 103078 cancer patients. Oncoimmunology. 8:e16292582019. View Article : Google Scholar : PubMed/NCBI | |
|
Ding D, Wang L, Zhang Y, Shi K and Shen Y: Machine learning developed a programmed cell death signature for predicting prognosis and immunotherapy benefits in lung adenocarcinoma. Transl Oncol. 38:1017842023. View Article : Google Scholar : PubMed/NCBI | |
|
Müller-Tidow C, Bug G, Lübbert M, Krämer A, Krauter J, Valent P, Nachbaur D, Berdel WE, Ottmann OG, Fritsch H, et al: A randomized, open-label, phase I/II trial to investigate the maximum tolerated dose of the Polo-like kinase inhibitor BI 2536 in elderly patients with refractory/relapsed acute myeloid leukaemia. Br J Haematol. 163:214–222. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Liu L, Zhang B, Wu X, Cheng G, Han X, Xin X, Qin C, Yang L, Huo M and Yin L: Bioresponsive nanocomplex integrating cancer-associated fibroblast deactivation and immunogenic chemotherapy for rebuilding immune-excluded tumors. Nanomedicine. 58:1027432024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Yu T, Gao G, Xu J, Lin R, Pan Z, Liu J and Feng W: Cell division cycle 42 effector protein 4 inhibits prostate cancer progression by suppressing ERK signaling pathway. Biomol Biomed. 24:840–847. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Mi K, Zeng L, Chen Y, Ning J, Zhang S, Zhao P and Yang S: DHX38 enhances proliferation, metastasis, and EMT progression in NSCLC through the G3BP1-mediated MAPK pathway. Cell Signal. 113:1109622024. View Article : Google Scholar : PubMed/NCBI | |
|
Jeon Y, Park S, Lee SH, Kim TH, Kim SW, Ahn MJ, Jung HA and Chung JH: Combination of dabrafenib and trametinib in patients with metastatic BRAFV600E-Mutated thyroid cancer. Cancer Res Treat. 56:1270–1276. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng B, Liao X, Liu L, Zhang C, Ruan H and Yang B: Thyroid hormone mediates cardioprotection against postinfarction remodeling and dysfunction through the IGF-1/PI3K/AKT signaling pathway. Life Sci. 267:1189772021. View Article : Google Scholar : PubMed/NCBI | |
|
Shi X, Han X and Cao Y, Li C and Cao Y: ZCCHC14 regulates proliferation and invasion of non-small cell lung cancer through the MAPK-P38 signalling pathway. J Cell Mol Med. 25:1406–1414. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ling Y, Li J and Zhou L: Smoking-related epigenetic modifications are associated with the prognosis and chemotherapeutics of patients with bladder cancer. Int J Immunopathol Pharmacol. 37:39463202311667742023. View Article : Google Scholar : PubMed/NCBI | |
|
Prahallad A, Weiss A, Voshol H, Kerr G, Sprouffske K, Yuan T, Ruddy D, Meistertzheim M, Kazic-Legueux M, Kottarathil T, et al: CRISPR screening identifies mechanisms of resistance to KRASG12C and SHP2 inhibitor combinations in Non-Small cell lung cancer. Cancer Res. 83:4130–4141. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Phyu SM, Tseng CC and Smith TAD: CDP-choline accumulation in breast and colorectal cancer cells treated with a GSK-3-targeting inhibitor. Magma. 32:227–235. 2019. View Article : Google Scholar : PubMed/NCBI |