Open Access

Integrated analysis of single‑cell and bulk RNA sequencing data to construct a risk assessment model based on plasma cell immune‑related genes for predicting patient prognosis and therapeutic response in lung adenocarcinoma

  • Authors:
    • Weijun Zhou
    • Zhuozheng Hu
    • Jiajun Wu
    • Qinghua Liu
    • Zhangning Jie
    • Hui Sun
    • Wenxiong Zhang
  • View Affiliations

  • Published online on: April 7, 2025     https://doi.org/10.3892/ol.2025.15017
  • Article Number: 271
  • Copyright: © Zhou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Plasma cells serve a crucial role in the human immune system and are important in tumor progression. However, the specific role of plasma cell immune‑related genes (PCIGs) in tumor progression remains unclear. Therefore, the present study aimed to establish a risk assessment model for patients with lung adenocarcinoma (LUAD) based on PCIGs. The data used in the present study were obtained from The Cancer Genome Atlas and the Gene Expression Omnibus databases. After identifying nine PCIGs, a risk assessment model was constructed and a nomogram was developed for predicting patient prognosis. To explore the molecular mechanism and clinical significance, gene set enrichment analysis (GSEA), tumor mutational burden (TMB) analysis, tumor microenvironment (TME) analysis and drug sensitivity prediction were performed. Furthermore, the accuracy of the model was validated using reverse transcription‑­quantitative PCR (RT‑qPCR). The present study constructed a risk assessment model consisting of nine PCIGs. Kaplan‑Meier survival curves indicated a worse prognosis in the high‑risk subgroup (risk score ≥0.982) compared with that in the low‑risk subgroup. The nomogram exhibited predictive value for survival prediction (area under the curve=0.727). GSEA enrichment analysis revealed enrichment of the focal adhesion and extracellular matrix‑receptor interaction pathways in the high‑risk group. Moreover, the high‑risk group exhibited a higher TMB, as demonstrated by the TME analysis showing lower ESTIMATE scores. Drug sensitivity prediction facilitated potential drug selection. Subsequently, differential gene expression was validated in multiple LUAD cell lines using RT‑qPCR. In conclusion, the risk assessment model based on nine PCIGs may be used to predict the prognosis and drug selection in patients with LUAD.

Introduction

Lung cancer is one of the most common cancers worldwide. The main types of lung cancer are lung adenocarcinoma (LUAD), lung squamous cell carcinoma and small cell lung cancer (1). Among them, LUAD is the most common histological subtype, accounting for ~40% of the global incidence of lung cancer (2). Despite advancements in therapeutic techniques and the identification of tumor therapeutic targets, such as immune checkpoint inhibitors (ICIs) [for example, programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1)] (3), which have improved patient prognosis, lung cancer remains the primary cause of cancer-related mortality worldwide, accounting for ~1.76 million deaths annually, which represents 18.4% of all cancer-related deaths. This high mortality rate is largely attributed to drug resistance and distant metastasis (4). The tumor (T)-node (N)-metastasis (M) system is currently widely utilized to evaluate the degree of tumor development (5). However, the TNM system does not accurately predict the prognosis of patients with lung cancer (6). Therefore, it is particularly important to identify a new biomarker to construct a risk assessment model for predicting the prognosis and therapeutic response of patients with LUAD.

B cells undergo differentiation into plasma cells upon activation, with plasma cells producing and releasing significant quantities of antigen-specific immunoglobulins (7). This process is crucial for the adaptive immune response and serves a significant role in humoral immunity. In recent years, plasma cells have been reported to be related to tumor progression; for example, multiple myeloma results from the malignant transformation of plasma cells or their precursors (8). In addition, plasma cell leukemia (PCL) is a rare cancer caused by the uncontrolled proliferation of plasma cells in the peripheral blood and bone marrow (9). Chaudhary et al (9) reported that tumor protein 53 (TP53), mitogen-activated protein kinase 1, suppressor of cytokine signaling 1, methyl-CpG binding domain protein 3 and YES proto-oncogene 1, Src family tyrosine kinase are signature central genes that may lead to poor prognosis in PCL. Considerable progress has been made in cancer immunotherapy research regarding the response of the immune system to cancer (10). Hao et al (11) used the plasma cell signature as a potential biomarker to predict the overall survival benefit and efficacy of a PD-1/PD-L1 blockade in patients with LUAD. However, no studies have used plasma cell signatures to predict the prognosis of patients with LUAD, to the best of our knowledge.

In recent years, the utilization of RNA sequencing has greatly expanded in cancer research due to its broad applicability (12). Traditional RNA sequencing can only detect the average expression level of all cells in a sample and cannot discern changes in individual cells. However, the advent of single-cell sequencing technology has addressed this limitation, enabling the detection of transcriptomes in different cell types (13). Utilizing this technology, Wang et al (14) employed single-cell RNA sequencing (scRNA-seq), along with lipidomic techniques, to investigate dysregulated lipid metabolism in lung cancer, offering a potential avenue for early detection. Similarly, Zhu et al (15) used scRNA-seq and spatial transcriptomics to elucidate specific cellular information and spatial architecture of cancer cells and tumor microenvironment (TME) subpopulations, thus advancing precision medicine in understanding the invasive process of LUAD from adenocarcinoma in situ to invasive adenocarcinoma cancer. Therefore, the present study aimed to develop a risk assessment model for patients with LUAD based on plasma cell immune-related genes (PCIGs) by integrating scRNA-seq and bulk RNA sequencing.

The present study developed a risk assessment model to evaluate the prognosis of patients with LUAD and its clinical significance. Several analyses, including survival analysis, enrichment analysis, tumor mutational burden (TMB) analysis, TME differential analysis, drug sensitivity prediction and clinical correlation analysis, were performed.

Materials and methods

Data sources and access

A total of 1,288 samples were collected, including 600 LUAD samples obtained from The Cancer Genome Atlas (TCGA) database (TCGA-LUAD; www.portal.gdc.cancer.gov/). Tumor transcriptome data, clinical information and tumor somatic cell mutation data were extracted from the TCGA database. Additionally, data from the GSE72094 cohort (n=442) and GSE31210 cohort (n=246) from the Gene Expression Omnibus (GEO) database (www.ncbi.nlm.nih.gov/geo/) were used (Table I). The GSE31210 cohort served as an independent external dataset to validate the reproducibility of the risk model. Furthermore, immune-related genes were retrieved from ImmPort (www.immport.org/home) and InnateDB (www.innatedb.ca/), and the two sets of genetic information were merged.

Table I.

Clinical information of the patients in The Cancer Genome Atlas, Gene Expression Omnibus and external groups.

Table I.

Clinical information of the patients in The Cancer Genome Atlas, Gene Expression Omnibus and external groups.

A, TCGA cohort (n=522)

Characteristicn (%)
Age
  <65 years223 (42.72)
  >65 years280 (53.64)
  Unknown19 (3.64)
Status
  Alive334 (63.98)
  Dead188 (36.02)
Sex
  Female280 (53.64)
  Male242 (46.36)
Stage
  I279 (53.45)
  II124 (23.75)
  III85 (16.28)
  IV26 (4.98)
  Unknown8 (1.53)
T stage
  T1172 (32.95)
  T2281 (53.83)
  T347 (9.00)
  T419 (3.64)
  Unknown3 (0.57)
M stage
  M0353 (67.62)
  M125 (4.79)
  Unknown144 (27.59)
N stage
  N0335 (64.18)
  N198 (18.77)
  N275 (14.37)
  N32 (0.38)
  Unknown12 (2.30)

B, GEO cohort (n=442)

Characteristicn (%)

Age
  <65 years127 (28.73)
  >65 years294 (66.52)
  Unknown21 (4.750)
Status
  Alive298 (67.42)
  Dead122 (27.60)
  Unknown22 (4.98)
Sex
  Female240 (54.30)
  Male202 (45.70)
Stage
  I265 (59.95)
  II69 (15.61)
  III63 (14.25)
  IV17 (3.84)
  Unknown28 (6.33)

C, External cohort (n=246)

Characteristicn (%)

Age
  <65 years178 (72.36)
  >65 years68 (27.64)
Status
  Alive174 (70.73)
  Dead52 (21.14)
  Unknown20 (8.13)
Sex
  Female130 (52.85)
  Male116 (47.15)
Stage
  I168 (68.29)
  II58 (23.58)
  III0 (0.00)
  IV0 (0.00)
  Unknown20 (8.13)

[i] TCGA, The Cancer Genome Atlas; GEO, Gene Expression Omnibus; T stage, Tumor stage; N stage, Node stage; M stage, metastasis stage.

Analysis of single-cell data

The LUAD cell cluster, cell annotation and differentially expressed gene (DEG) data for each cluster were obtained from the Tumor Immune Single-cell Hub 2 (TISCH2) database (www.tisch.comp-genomics.org/), which was derived from the GSE131907 cohort (16) and a total of 203,298 cells from 44 patients. The Seurat package was used for clustering analysis using the FindClusters function, and cell distribution was visualized in a two-dimensional space using the RunTSNE function. R software (version 4.3.2; http://www.r-project.org/) was used to screen for marker genes in plasma cells.

Identification of PCIGs

PCIGs were identified as genes commonly expressed among immune-related genes and plasma cell marker genes. The R package ‘VennDiagram’ was utilized to determine the intersection, and a Venn diagram was generated to visualize the PCIGs.

Enrichment analysis and protein-protein interaction (PPI) networks

Considering the aforementioned PCIGs, an exploration of potential molecular mechanisms was initiated using Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. These analyses were performed utilizing the R packages ‘clusterProfiler’ and ‘org.Hs.eg.db’. GO and KEGG terms exhibiting a significance level of P<0.05 were visually represented using the ‘circlize’ R package. To further assess the protein-level mechanisms and associations, the STRING database (www.cn.string-db.org/) was used to elucidate the PPI relationships among the PCIGs. Subsequently, these interactions were depicted through a network diagram. Furthermore, R software was utilized to construct a histogram illustrating the core genes identified in the analysis.

Identification of prognostic genes

The R packages ‘limma’ and ‘sva’ were used to determine the intersection of TCGA cohort and the GEO cohort to obtain the expression information of the intersecting genes in the two cohorts. Subsequently, the genes commonly expressed between the PCIGs and the intersection genes were identified. Univariate Cox hazard analysis was performed to identify prognosis-related genes using the R packages ‘survival’ and ‘survminer’ (P<0.05). After identifying prognosis-related genes, the genes were visualized by generating forest plots.

Construction and validation of the model

Initially, the TCGA cohort was designated as the training cohort and the GEO cohort as the testing cohort. Least absolute shrinkage and selection operator (LASSO) regression and multivariate Cox regression analysis were performed to construct a risk assessment model. LASSO regression analysis is a method used to analyze high-dimensional data and is often applied to construct regression models (17). The R package ‘glmnet’ was utilized to effectively reduce the number of genes in the final risk model. The risk scores for both the training and test cohorts were derived based on the model format. The risk score calculation followed the following formula: Risk score=coefficient (gene 1) × expression (gene 1) + coefficient (gene 2) × expression (gene 2) + coefficient (gene 3) × expression (gene 3) + … + coefficient (gene n) × expression (gene n). Risk scores exceeding the median were classified as high risk, whilst those below the median were classified as low risk. Following this classification, univariate and multivariate independent prognostic analyses were performed to assess the independence of the model compared with other clinical features.

Subsequently, the R packages ‘survminer’ and ‘survival’ were used to construct Kaplan-Meier survival curves for the training and test cohorts, which depended on survival duration and survival status. In addition, the R package ‘timeROC’ was utilized to generate a receiver operating characteristic (ROC) curve to evaluate the efficacy of risk scores in predicting 1-, 3- and 5-year survival in patients with LUAD. Based on this, a nomogram was developed to further facilitate survival prediction., considering factors such as age, risk, sex and disease stage. Furthermore, a ROC curve based on the nomogram, decision curve analysis (DCA) and calibration curve were generated to further confirm the predictive accuracy of the nomogram (18). Additionally, the GSE31210 cohort was used as an independent external cohort to validate the prognostic model.

Finally, Kaplan-Meier curves were constructed for 9 PCIGs to evaluate the prognosis of the high- and low-risk groups under different gene subgroups.

Gene Set Enrichment Analysis (GSEA). GSEA was used to evaluate the enriched pathways, and the R packages ‘limma’, ‘org.Hs.eg.db’, ‘clusterProfiler’ and ‘enrichplot’ were used to generate GSEA enrichment maps. The five most significant pathways were then delineated in the high- and low-risk groups (19).

TME analysis

The R package ‘ESTIMATE’ was used to analyze the stromal score, immune score and ESTIMATE score. Following this, a comparison was made between the high- and low-risk groups to identify any differences in these scores. Subsequently, the R packages ‘GSVA’ and ‘GSEABase’ were utilized to perform single-sample GSEA. This analysis enabled the elucidation of the differences in the infiltration of major immune cells between the high- and low-risk groups. The same analysis was then used to evaluate the variances in immune-related functions.

TMB

To assess the gene mutations in tumor cells in each sample, TMB data for LUAD was obtained from TCGA database (www.portal.gdc.cancer.gov/). The data was manipulated using the ‘TCGAbiolinks’ package and waterfall plots were constructed using the R programming language ‘maftools’, after which the TMB was calculated (20,21).

Treatment response prediction

The association between the risk score and ICI-related gene expression levels were evaluated, and the R package ‘ggplot2’ was used for visualization. According to the tumor expression profiles downloaded from the database (http://tide.dfci.harvard.edu/), the Tumor Immune Dysfunction and Exclusion (TIDE) score could be used as a transcriptomic biomarker to predict the response to immune checkpoint blockade and to explore the potential clinical efficacy of immunotherapy. Moreover, to assess the sensitivity of several risk groups to chemotherapy drugs, the R package ‘OncoPredict’ was utilized to forecast the disparity in the treatment outcomes of chemotherapy drugs among high- and low-risk patients (22).

Clinical correlation analysis

The clinicopathological features of patients were then obtained from TCGA cohort to assess the linkages between clinical features and the risk score, and a heatmap was generated utilizing the R package ‘ComplexHeatmap’. Moreover, the R packages ‘reshape2’, ‘tidyverse’, ‘ggplot2’, ‘RColorBrewer’ and ‘grid’ were used to construct a cyclic graph of clinical correlations. This enabled the evaluation of disparities in clinical characteristics between the high- and low-risk groups. Subsequently, the associations between each clinical characteristic and the risk score were assessed individually.

Validation by reverse transcription-quantitative PCR (RT-qPCR)

TRIzol™ reagent (Thermo Fisher Scientific, Inc.) was used to extract total RNA from normal human lung epithelial BEAS-2B cells and human LUAD H1373 and H1563 cell lines. Reverse transcription was performed using the PrimeScript™ RT Reagent Kit (Takara Bio, Inc.) at 42°C for 30 min, followed by inactivation at 85°C for 5 min. SYBR Green Master Mix (GeneCopoeia, Inc.) was used for qPCR. PCR amplification was initiated with an initial denaturation step at 95°C for 5 min, followed by 40 cycles consisting of 95°C for 15 sec (denaturation), 60°C for 30 sec (annealing) and 72°C for 30 sec (extension). A final extension step was performed at 72°C for 5 min. mRNA expression levels were normalized to GAPDH mRNA levels and the 2−ΔΔCq method (23) was used for calculating the relative expression of mRNAs. All primers were purchased from Takara Biomedical Technology (Beijing) Co., Ltd.; Takara Bio Inc., and Table SI presents the forward and reverse primers used for RT-qPCR, including PCIGs and primers for normalization control (National Center for Biotechnology Information reference sequence, NM_002046.7).

Immunohistochemical analysis

Finally, immunohistochemical images of selected PCIGs were obtained from the Human Protein Atlas (HPA) database (www.proteinatlas.org/) (24), which is a comprehensive resource that provides high-resolution images and detailed data on protein expression in normal tissues, cancer tissues and cell lines. Based on the obtained images, the differences in the protein expression levels of selected PCIGs between lung cancer tissues and normal lung tissues were evaluated by assessing staining intensity and positive cell proportion provided in the HPA database.

Statistical analysis

Statistical analyses were performed using R, version 4.3.2. The Kruskal-Wallis test was used for comparing multiple groups, followed by Dunn's test for post hoc comparisons, with Bonferroni correction when applicable. For comparisons between two groups, the Wilcoxon rank-sum test was employed. Pearson correlation was used for evaluating the correlation between continuous variables. The Log-rank test was used to evaluate survival differences using Kaplan-Meier curves, with statistical significance defined as P<0.05. LASSO regression and Cox regression analyses were utilized to develop the prediction model. All data are presented as mean ± standard deviation. For each experiment, three replicates were performed per sample to ensure reproducibility. P<0.05 was considered to indicate a statistically significant difference.

Results

Identification of PCIGs

Fig. 1 presents the flow chart of the present study. Human immune-related genes were obtained from the ‘ImmPort’ and ‘InnateDB’ databases, and the data sets were merged, identifying 2,533 genes. The gene expression profiles of 203,298 cells from 44 LUAD samples were subsequently obtained from the TISCH database (GSE131907). The cell clustering and annotation results revealed that cells with similar differential gene expression patterns were grouped into distinct clusters (Fig. 2A and B). scRNA-seq data was then utilized to perform cell clustering based on differential gene expression profiles. A total of 25 clusters were identified, and each cluster was named according to the expression of specific marker genes. For example, clusters 16 and 17 were identified as plasma cells, whilst clusters 3 and 4 were identified as macrophages. Notably, clusters 16 and 17, characterized as plasma cells, exhibited distinct profiles of DEGs (Table SII). To further elucidate the cellular origins of the DEGs identified in the GSE131907 dataset, in the detailed annotations of the relevant plasma cells were summarized to clarify their contributions to the gene expression profiles analyzed in the present study (Table SIII). Additionally, cluster 0 was identified as B cells, and 737 marker genes were obtained for the plasma cells. The study involved the analysis of the overlap between plasma cell marker genes and human immune-related genes, and 175 common DEGs were identified (Table SIV). A Venn diagram was generated to visualize the distribution of these genes (Fig. 2C), and these genes are referred to as PCIGs.

Enrichment analysis and PPI network

GO enrichment analysis of the DEGs revealed that biological processes were involved mainly in microtubule-based movement, humoral immune response and defense response to bacteria. The most representative cellular component terms were the collagen-containing extracellular matrix (ECM), immunoglobulin complex and external side of the plasma membrane. Among the molecular functions, signaling receptor activator activity, receptor ligand activity and glycosaminoglycan binding were the main categories (Fig. S1A and B). According to the KEGG analysis, the model may be associated with complement and coagulation cascades, arachidonic acid metabolism, hematopoietic cell lineage and linoleic acid metabolism (Fig. S1C and D). Based on the data and images acquired from the STRING database, the PPIs of the PCIGs were analyzed (Fig. S2A), and the core gene histogram demonstrated that the IL-1B gene had the highest number of adjacent nodes compared to the other top 30 core genes (Fig. S2B).

Identification of plasma cell prognosis-related genes

The intersection of the TCGA and GEO cohorts (GSE72094) were utilized to acquire expression data for the common genes present in both cohorts. Subsequently, genes that were commonly expressed between the PCIGs and the intersection genes were identified. Among all PCIGs sources, including primary tumors, lymph nodes, brain metastases, pleural effusions, and normal lung tissues and lymph nodes, co-expressed genes were extracted from the TCGA and GEO cohorts. In the GEO cohort, all tested genes were derived from tumor tissues of LUAD, whilst in the TCGA cohort, normal tissue samples were omitted. Therefore, the final screened PCIGs consisted only of genes differentially expressed in tumor tissues. A univariate Cox hazard analysis was then performed based on the survival data of the aforementioned co-expressed genes to identify the genes associated with prognosis (Table SV and Fig. S2C).

Construction and validation of the prognostic model

After LASSO regression analysis (Fig. 2D and E) and multivariate Cox regression analysis, 9 PCIGs were identified, namely, galectin-3 (LGALS3), WAP four-disulfide core domain 2 (WFDC2), leukocyte specific transcript 1 (LST1), X-box binding protein 1 (XBP1), ribosomal protein S19 (RPS19), superoxide dismutase 1 (SOD1), integrin β1 (ITGB1), CCAAT enhancer binding protein β (CEBPB) and C-C motif chemokine ligand 20 (CCL20) (Table SVI). The high-risk group was defined as individuals with risk scores above the median, whilst the low-risk group was defined as individuals with risk scores below the median (Table II). The risk assessment model was subsequently subjected to univariate and multivariate independent prognostic analysis (Table SVII). The results demonstrated that the risk score could predict patient prognosis independent of other clinical characteristics (Fig. 2F and G).

Table II.

Clinical information of the patients with lung adenocarcinoma in the low- and high-risk groups.

Table II.

Clinical information of the patients with lung adenocarcinoma in the low- and high-risk groups.

A, Low risk group (n=254)

Characteristicn (%)
Age
  ≤65 years120 (47.24)
  >65 years128 (50.39)
  Unknown6 (2.36)
Sex
  Female155 (61.02)
  Male99 (38.98)
Status
  Alive187 (73.62)
  Dead67 (26.38)
Stage
  I163 (64.17)
  II52 (20.47)
  III27 (10.63)
  IV9 (3.54)
  Unknown3 (1.18)
T stage
  T1102 (40.16)
  T2131 (51.57)
  T316 (6.30)
  T43 (1.18)
  Unknown2 (0.79)
N stage
  N0178 (70.08)
  N141 (16.14)
  N225 (9.84)
  N30 (0)
  Unknown10 (3.94)
M stage
  M0168 (66.14)
  M18 (3.15)
  Unknown78 (30.71)

B, High risk group (n=253)

Characteristicn (%)

Age
  ≤65 years119 (47.04)
  >65 years130 (51.38)
  Unknown4 (1.58)
Sex
  Female117 (46.25)
  Male136 (53.75)
Status
  Alive137 (54.15)
  Dead116 (45.85)
Stage
  I109 (43.08)
  II68 (26.88)
  III54 (21.34)
  IV17 (6.72)
  Unknown5 (1.98)
T stage
  T167 (26.48)
  T2140 (55.34)
  T339 (15.42)
  T416 (6.32)
  Unknown1 (0.40)
N stage
  N0149 (58.89)
  N154 (21.34)
  N246 (18.18)
  N32 (0.79)
  Unknown2 (0.79)
M stage
  M0170 (67.19)
  M117 (6.72)
  Unknown66 (26.09)

[i] T stage, Tumor stage; N stage, Node stage; M stage, metastasis stage.

Considering the TCGA cohort as the training cohort and the GEO cohort as the test cohort, the high-risk group in the training cohort exhibited significantly worse survival than that of the low-risk group. This trend was also observed in the test cohort (Fig. 3A and B). The predictive capability of the model for patient survival in LUAD was also assessed. The risk score demonstrated a strong performance in predicting the 1-, 3- and 5-year survival rates of patients with LUAD. Specifically, in the training cohort, the area under the curve (AUC) values were 0.740, 0.715 and 0.678 at 1, 3 and 5 years, respectively. In the test cohort, the AUC values were 0.655, 0.598 and 0.748 at 1, 3 and 5 years, respectively (Fig. 3D and E). Similar results were observed for the GSE31210 cohort (Fig. 3C and F). Risk plots were used to illustrate the survival status of patients in the training and test cohorts (Fig. S3). To evaluate the predictive value of the model, a nomogram based on age, sex, stage and risk score was constructed (Fig. 4A), which showed predictive value for predicting the survival of patients with LUAD (AUC=0.727; Fig. 4B). Additionally, to confirm the precision of the predictive impact of the model, DCA and calibration curve assessments were performed. The DCA results indicated that the risk score served as a reliable predictive tool (Fig. 4C), independent of other clinical characteristics, thereby supporting its effectiveness. A calibration curve analysis combined with the risk score demonstrated a greater C-index of 0.698 (Fig. 4D) compared with the C-index of 0.6758 without the risk score (Fig. 4E). These findings indicated that the model improves the prognosis of patients with LUAD.

Additionally, survival curves were generated for each prognostic-related gene (Table SVIII). According to the survival curve analysis, the high-risk group exhibited significantly worse survival outcomes for the genes CCL20, CEBPB, ITGB1, LGALS3, RPS19 and SOD1 compared with that of the low-risk group. Conversely, the low-risk group demonstrated significantly worse survival outcomes for the genes LST1, WFDC2 and XBP1 (Fig. S4). Moreover, it was demonstrated that the survival curves for CCL20, CEBPB and LST1 intersected in the later stages. Consequently, a two-stage test was performed, and the results revealed that the P-values for all three genes were <0.05 (Table SIX), leading to the rejection of the null hypothesis (namely, there is no significant effect or relationship between gene expression and survival time). These findings indicate that, according to statistical tests, gene expression significantly impacts survival time. Furthermore, based on the PCIGs used to construct the model, a mechanistic map of the role of several PCIGs in the progression and obstruction of LUAD was drawn (Fig. 5).

GSEA

GSEA revealed that the cell cycle, ECM-receptor interaction, focal adhesion, pathways in cancer and spliceosome pathways were notably enriched in the high-risk group (Fig. 6A), whilst the B-cell receptor signaling pathway, complement activation, immunoglobulin complex, immunoglobulin complex circulating, immunoglobulin receptor binding were notably enriched in the low-risk group (Fig. 6B).

Figure 6.

GSEA, TME and TMB analyses in different risk groups. GSEA was used to assess the biological processes and pathways enriched in the (A) high- and (B) low- risk groups. (C) Box plots showing the fraction of different immune cells in the high- and low-risk groups. (D) Heatmap illustrating the relative proportion (%) of immune cells between the high- and low-risk groups. (E) Immune cells infiltration score and immune-related function in the low-and high-risk groups estimated by single sample GSEA. (F) Violin plots of differences in immune, stromal and ESTIMATE scores. Waterfall plots summarizing the gene mutation landscape in the (G) high- and (H) low-risk groups. (I) TMB differences between the high-and low-risk patients. *P<0.05, **P<0.01 and ***P<0.001 vs. low-risk. GSEA, gene set enrichment analysis; TME, tumor microenvironment; TMB, tumor mutation burden; KEGG, Kyoto Encyclopedia of Genes and Genomes; GO, Gene Ontology; aDCs, activated dendritic cells; CCR, C-C motif chemokine receptor; HLA, human leukocyte antigen; Tfh, follicular helper T cells; TIL, tumor-infiltrating lymphocyte; TP53, tumor protein p53; TTN, titin; MUC16, mucin 16; CSMD3, CUB and Sushi multiple domains 3; RYR2, ryanodine receptor 2; LRP1B, LDL receptor related protein 1B; ZFHX4, zinc finger homeobox 4; USH2A, usherin 2A; KRAS, Kirsten rat sarcoma viral oncogene homolog; XIRP2, Xin actin binding repeat containing 2; FLG, filaggrin; SPTA1, spectrin α erythrocytic 1; NAV3, neuron navigator 3; ZNF536, zinc finger protein 536; COL11A1, collagen type XI α1 chain; ANK2, ankyrin 2; FAT3, FAT atypical cadherin 3; PCLO, piccolo presynaptic cytomatrix protein; APOB, apolipoprotein B; iDCs, immature dendritic cells.

TME analysis

Analysis of immune cell infiltration revealed that the high-risk group exhibited greater invasion of T cells activated by CD4 memory cells, resting natural killer cells, M0 macrophages, M1 macrophages and activated mast cells than the low-risk group, whilst resting dendritic cells and resting mast cells showed greater invasion in the low-risk group than in the high-risk group (Fig. 6C and D). Subsequently, variations in immune-related functions were assessed across different risk groups. Significant differences were observed in activated dendritic cells, B cells, human leukocyte antigen, immature dendritic cells, mast cells, neutrophils, T helper cells, tumor-infiltrating lymphocytes and the type II interferon response, all of which were enriched in the low-risk group (Fig. 6E). In addition, TME analysis suggested that the stromal score, immune score and ESTIMATE score were significantly greater in the low-risk group than in the high-risk group (Fig. 6F), suggesting that there may be a higher presence of tumor cells in the high-risk group.

TMB

Based on the somatic mutation data for LUAD tumors from the TCGA database, a waterfall plot of the top 20 mutated genes in the different groups was generated. In the high-risk group, the five most common mutated genes were TP53 (52%), titin (TTN; 52%), mucin-16 (MUC16; 42%), CUB and sushi multiple domains 3 (CSMD3; 44%) and ryanodine receptor 2 (RYR2; 37%). TP53 (39%), TTN (35%), MUC16 (38%), CSMD3 (32%) and RYR2 (35%) were prone to mutation in low-risk patients (Fig. 6G-H). Calculation of the TMB demonstrated that there was a significant positive correlation between the TMB and the risk score (Fig. 6I).

Treatment response prediction

The association between the risk score and ICI-related gene expression was evaluated. The findings indicated a significant association: A high risk score was significantly associated with the upregulation of CD276 compared with a low risk score; however, there was no significant difference in the risk score according to the PD-1, programmed cell death protein 1 or cytotoxic T-lymphocyte associated protein 4 expression level (Fig. 7A). Subsequently, according to the results of the TIDE analysis, patients in the low-risk subgroup had significantly higher TIDE scores than those in the high-risk subgroup (Fig. 7B). The ‘oncoPredict’ R package was then used to evaluate the sensitivity of patients with LUAD in different risk groups to chemotherapy drugs. The results revealed that the high-risk group was significantly more sensitive to doramapimod (p38 MAPK inhibitor), ribociclib (CDK4/6 inhibitor), BMS-754807 (IGF-1R inhibitor) and SB216763 (GSK3β inhibitor) than the low-risk group, whilst the low-risk group was significantly more sensitive to SCH772984 (ERK inhibitor), 5-Fluorouracil (antimetabolite drug), cisplatin (platinum based chemotherapy agent) and MK-1775 (Potent Wee1 inhibitor) than the high-risk group (Fig. 7C and Table SX).

Clinical correlation analysis

The results of the clinical correlation analysis indicated that male patients, as well as those with worse T and N stages, had significantly higher risk scores compared with female patients and those with lower T and N stages (Fig. 8A). Additionally, a cyclic graph was constructed to evaluate clinical correlation. The findings also indicated that patients with higher risk scores had significantly more advanced grade, T stages and N stages compared with those with lower risk scores (Fig. 8B). Boxplots revealed similar results, with increasing risk score associated with a significant increase in the lung cancer stage (Fig. S5).

In vitro experimental validation of the risk models

The protein expression levels of LGALS3 (https://www.proteinatlas.org/ENSG00000131981-LGALS3), WFDC2 (https://www.proteinatlas.org/ENSG00000101443-WFDC2), SOD1 (https://www.proteinatlas.org/ENSG00000142168-SOD1), ITGB1 (https://www.proteinatlas.org/ENSG00000150093-ITGB1) and CEBPB (https://www.proteinatlas.org/ENSG00000172216-CEBPB) were compared between LUAD and normal tissues based on immunohistochemical staining images obtained from the HPA database (Fig. S6A). Furthermore, RT-qPCR revealed that LST1 and XBP1 were highly expressed in normal human lung cell lines (P<0.05); LGALS3, RPS19, SOD1, ITGB1 and CEBPB were highly expressed in both the H1373 and H1563 cell lines, with CEBPB showing the highest significance (P<0.001), and CCL20 was highly expressed in only the H1563 cell line (P<0.05) (Fig. S6B and C).

Discussion

Drug resistance and distant metastasis are major contributors to the status of LUAD as a leading cause of cancer-related deaths worldwide. The TNM system remains the most widely used classification method for lung cancer staging. While it provides valuable prognostic information, its accuracy in predicting patient outcomes for LUAD is limited, highlighting the need for continuous refinement and complementary approaches (25). Recently, researchers have explored the potential of single-cell sequencing as an emerging tool to enhance prognosis prediction and therapeutic response assessment (26). Therefore, the aim of the present study was to develop a risk assessment model for patients with LUAD using PCIGs through the integration of scRNA-seq and bulk RNA sequencing. Additionally, the present study aimed to assess the potential molecular mechanisms and clinical applications of the model. According to the model, Kaplan-Meier survival curves revealed a worse prognosis in the high-risk group compared with the low-risk group, a finding validated in the external cohort. The nomogram demonstrated the effectiveness of the model in predicting the prognosis of patients with LUAD. Furthermore, GSEA results indicated enrichment of the focal adhesion and ECM receptor interaction pathways in the high-risk group. TMB analysis also revealed a higher TMB in the high-risk group.

The immune system serves an important role in the progression of tumors, and immune cell signatures are increasingly favored by researchers. As important immune cells in the human body, plasma cells also serve an important role in tumor progression (27). The present study acquired data from the TCGA, GEO and TISCH databases, and univariate Cox hazard analysis was performed to identify prognosis-related genes. LASSO regression and multivariate Cox regression analysis were subsequently performed to develop a risk assessment model. Individuals classified as high risk had a notably lower survival rate than those in the low-risk group. The nomogram showed good predictive value for predicting patient prognosis, indicating that the model can predict the prognosis of patients with LUAD independently and effectively. Similar models based on different immune cells have also been proposed in other studies and have demonstrated predictive value for the prognosis of LUAD. For example, Song et al (28) constructed a model based on natural killer cell marker genes to predict the prognosis of patients with LUAD using integrated analysis of scRNA-seq and bulk RNA sequencing. LGALS3, WFDC2, LST1, XBP1, RPS19, SOD1, ITGB1, CEBPB and CCL20 were identified as signature genes to construct the prognostic assessment model in the present study, which effectively predicted the prognosis of patients with LUAD. LGALS3 is an important regulator of LUAD progression. Vuong et al (29) reported that an orally active LGALS3 antagonist effectively blocked the growth and spread of LUAD in a novel study. Furthermore, XBP1 is closely related to tumorigenesis and tumor progression (30) and serves an antitumorigenic role in LUAD through alternative splicing. The adaptation of plasma cells may serve a role in this process (31). As a major antioxidant enzyme, SOD1 is essential for the growth of non-small cell lung cancer (NSCLC), as knockdown or pharmacological inhibition of SOD1 potently inhibits the growth of NSCLC cell lines driven by the oncogenes KRAS and EGFR (32,33). The accumulation of the basic leucine zipper family member CEBPB potentially controls the expression of the nuclear factor erythroid 2-related factor 2 gene, which may serve a role in the progression of NSCLC (34). Furthermore, CEBPB can promote the production of IL-6 in the immune response center in pneumonia, thus aggravating the inflammatory response of pulmonary epithelial cells (35). CCL20 serve a role in several oncogenic processes, and Fan et al (36) reported that downregulated expression of CCL20 can repress EMT signaling pathways in LUAD cells and restrain tumor growth. The remaining PCIGs have also been confirmed to be related to tumor progression (3739).

Furthermore, GSEA and TMB analysis were implemented to elucidate the potential underlying mechanisms involved. The high-risk group displayed notable upregulation in pathways related to ECM-receptor interactions and focal adhesion. This finding suggests that the poor prognosis of patients in the high-risk group may be related to ECM-receptor interactions, which significantly contribute to several tumor progression processes. The involvement of ECM-receptor interactions in other types of cancers has been demonstrated (40). Furthermore, Anagnostou et al (41) reported that a higher concentration of neoepitopes resulting from somatic mutations could amplify the clinical advantages derived from immune checkpoint blockade. This finding suggested that immunotherapy may be more effective in individuals in the high-risk group. The present study also demonstrated a greater occurrence of TP53 mutation in the high-risk group. Previous research suggests that there is a notable link between TP53 mutation and the treatment and prognosis of lung cancer (42).

Subsequently, the present study evaluated the capacity of the model to predict treatment response, and it was demonstrated that the expression of CD276 was positively associated with the risk score. B7-H3 (CD276) has been reported to be upregulated in several types of human cancer cells (43). In patients with NSCLC, B7-H3 has been identified as potentially functioning in conjunction with other factors to aid in the immune evasion of tumor cells (44). Furthermore, B7-H3 may also serve a role in conferring resistance to anticancer drugs through different mechanisms. Enoblituzumab (MGA271), a humanized monoclonal antibody that targets B7-H3, was reported to have satisfactory safety and demonstrated antitumor efficacy in patients with NSCLC (45). Subsequently, sensitivity to chemical drugs between the high- and low-risk groups was evaluated. The high-risk group was more sensitive to doramapimod, which is a p38 MAPK inhibitor that has shown specific sensitivity to cancer cell lines and organoids of cervical squamous cell carcinoma (46). The aforementioned results suggest that the model in the present study may be used guide immunotherapy and chemotherapy for patients with LUAD.

The present study marks the inaugural attempt to develop a prognostic signature for patients with LUAD, utilizing PCIGs and integrating both scRNA-seq and bulk RNA sequencing data. This approach addresses a crucial gap, as bulk RNA sequencing cannot capture the nuances observed at the single-cell level (13). The present study still has some limitations. First, it relies on publicly available datasets, which may introduce biases related to data processing, sample heterogeneity and batch effects. Additionally, while external validation and RT-qPCR were performed, further experimental validation is required. Specifically, western blotting or immunohistochemistry analyses in LUAD tissue samples and cell lines should be conducted to confirm protein expression and functional roles. Moreover, advanced spatial transcriptomics techniques could provide deeper insights into the spatial distribution of plasma cell immune-related genes within the tumor microenvironment.

From a clinical perspective, despite the promising predictive potential of the PCIG-based risk assessment model, its real-world applicability remains to be fully established. Well-designed prospective clinical studies are needed to assess its predictive accuracy for patient prognosis and therapeutic response in independent cohorts. Furthermore, the model's generalizability to other malignancies has not been explored, necessitating further validation across different tumor types to evaluate its broader applicability. Addressing these limitations through additional experimental and clinical research will be essential to strengthen the robustness and translational value of the present study findings. In conclusion, the model based on nine PCIGs and the nomogram demonstrated high efficacy for predicting patient prognosis. These findings may be associated with the enrichment of ECM-receptor interactions and focal adhesion pathways. The evaluation of ICIs and sensitivity to chemotherapeutic drugs may lead to potential protocols for drug selection in treatments for patients with LUAD. Nevertheless, well-designed prospective clinical trials and basic experiments are still needed to confirm the results of the present study.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by National Natural Science Foundation of China (grant no. 81560345).

Availability of data and materials

The TCGA datasets analyzed in this study are publicly available at https://www.cancer.gov/about-nci/organization/ccg/research/structural-genomics/tcga, while the GEO datasets can be accessed at https://www.ncbi.nlm.nih.gov/geo/ under accession numbers GSE72094, GSE31210, and GSE131907. The remaining data generated in the present study may be requested from the corresponding author.

Authors' contributions

HS had full access to all the data in the manuscript and takes responsibility for its integrity and accuracy. WZho, ZH, JW, QL, ZJ, HS and WZha contributed to the concept and design of the study. WZho, ZH, JW and HS performed the experiments, while WZho, ZH, JW, QL, ZJ, HS and WZha were involved in data acquisition, analysis and interpretation. Statistical analysis was conducted by WZho, ZH, JW and HS. The manuscript was drafted by WZho, HS and WZha, with critical revisions provided by WZho, HS and WZha. Supervision was carried out by WZho, HS and WZha. WZho and ZJ confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

DEG

differentially expressed gene

GEO

Gene Expression Omnibus

GSEA

gene set enrichment analysis

GO

Gene Ontology

ICI

immune checkpoint inhibitor

KEGG

Kyoto Encyclopedia of Genes and Genomes

LUAD

lung adenocarcinoma

LASSO

least absolute shrinkage and selection operator

NSCLC

non-small cell lung cancer

PD-1

programmed death 1

PD-L1

programmed death ligand 1

PCIG

plasma cell immune-related gene

scRNA-seq

single-cell RNA-sequencing

TME

tumor microenvironment

TNM

tumor-node-metastasis

TCGA

The Cancer Genome Atlas

TIDE

Tumor Immune Dysfunction and Exclusion

TISCH

Tumor Immune Single-cell Hub

TMB

tumor mutation burden

References

1 

Bade BC and Dela Cruz CS: Lung cancer 2020: Epidemiology, etiology, and prevention. Clin Chest Med. 41:1–24. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Duma N, Santana-Davila R and Molina JR: Non-small cell lung cancer: Epidemiology, screening, diagnosis, and treatment. Mayo Clin Proc. 94:1623–1640. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Yi M, Li A, Zhou L, Chu Q, Luo S and Wu K: Immune signature-based risk stratification and prediction of immune checkpoint inhibitor's efficacy for lung adenocarcinoma. Cancer Immunol Immunother. 70:1705–1719. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Germain C, Gnjatic S, Tamzalit F, Knockaert S, Remark R, Goc J, Lepelley A, Becht E, Katsahian S, Bizouard G, et al: Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer. Am J Respir Crit Care Med. 189:832–844. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Ettinger DS, Wood DE, Aisner DL, Akerley W, Bauman JR, Bharat A, Bruno DS, Chang JY, Chirieac LR, D'Amico TA, et al: Non-small cell lung cancer, version 3.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 20:497–530. 2022. View Article : Google Scholar : PubMed/NCBI

6 

Nesbitt JC, Putnam JB Jr, Walsh GL, Roth JA and Mountain CF: Survival in early-stage non-small cell lung cancer. Ann Thorac Surg. 60:466–472. 1995. View Article : Google Scholar : PubMed/NCBI

7 

Chen-Kiang S: Cell-cycle control of plasma cell differentiation and tumorigenesis. Immunol Rev. 194:39–47. 2003. View Article : Google Scholar : PubMed/NCBI

8 

O'Connor BP, Gleeson MW, Noelle RJ and Erickson LD: The rise and fall of long-lived humoral immunity: Terminal differentiation of plasma cells in health and disease. Immunol Rev. 194:61–76. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Chaudhary RK, Patil P, Ananthesh L, Srinivasa MG, Mateti UV, Shetty V and Khanal P: Identification of signature genes and drug candidates for primary plasma cell leukemia: An integrated system biology approach. Comput Biol Med. 162:1070902023. View Article : Google Scholar : PubMed/NCBI

10 

Lahiri A, Maji A, Potdar PD, Singh N, Parikh P, Bisht B, Mukherjee A and Paul MK: Lung cancer immunotherapy: Progress, pitfalls, and promises. Mol Cancer. 22:402023. View Article : Google Scholar : PubMed/NCBI

11 

Hao D, Han G, Sinjab A, Gomez-Bolanos LI, Lazcano R, Serrano A, Hernandez SD, Dai E, Cao X, Hu J, et al: The single-cell immunogenomic landscape of B and plasma cells in early-stage lung adenocarcinoma. Cancer Discov. 12:2626–2645. 2022. View Article : Google Scholar : PubMed/NCBI

12 

Hong M, Tao S, Zhang L, Diao LT, Huang X, Huang S, Huang S, Xie SJ, Xiao ZD and Zhang H: RNA sequencing: New technologies and applications in cancer research. J Hematol Oncol. 13:1662020. View Article : Google Scholar : PubMed/NCBI

13 

Suvà ML and Tirosh I: Single-Cell RNA sequencing in cancer: Lessons learned and emerging challenges. Mol Cell. 75:7–12. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Wang G, Qiu M, Xing X, Zhou J, Yao H, Li M, Yin R, Hou Y, Li Y, Pan S, et al: Lung cancer scRNA-seq and lipidomics reveal aberrant lipid metabolism for early-stage diagnosis. Sci Transl Med. 14:eabk27562022. View Article : Google Scholar : PubMed/NCBI

15 

Zhu J, Fan Y, Xiong Y, Wang W, Chen J, Xia Y, Lei J, Gong L, Sun S and Jiang T: Delineating the dynamic evolution from preneoplasia to invasive lung adenocarcinoma by integrating single-cell RNA sequencing and spatial transcriptomics. Exp Mol Med. 54:2060–2076. 2022. View Article : Google Scholar : PubMed/NCBI

16 

Kim N, Kim HK, Lee K, Hong Y, Cho JH, Choi JW, Lee JI, Suh YL, Ku BM, Eum HH, et al: Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nat Commun. 11:22852020. View Article : Google Scholar : PubMed/NCBI

17 

Zhang J, Liu X, Huang Z, Wu C, Zhang F, Han A, Stalin A, Lu S, Guo S, Huang J, et al: T cell-related prognostic risk model and tumor immune environment modulation in lung adenocarcinoma based on single-cell and bulk RNA sequencing. Comput Biol Med. 152:1064602023. View Article : Google Scholar : PubMed/NCBI

18 

Chen Q, Wang S and Lang JH: Development and validation of nomogram with tumor microenvironment-related genes and clinical factors for predicting overall survival of endometrial cancer. J Cancer. 12:3530–3538. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Yoshihara K, Shahmoradgoli M, Martínez E, Vegesna R, Kim H, Torres-Garcia W, Treviño V, Shen H, Laird PW, Levine DA, et al: Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 4:26122013. View Article : Google Scholar : PubMed/NCBI

20 

Mayakonda A, Lin DC, Assenov Y, Plass C and Koeffler HP: Maftools: Efficient and comprehensive analysis of somatic variants in cancer. Genome Res. 28:1747–1756. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, Sabedot TS, Malta TM, Pagnotta SM, Castiglioni I, et al: TCGAbiolinks: An R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 44:e712016. View Article : Google Scholar : PubMed/NCBI

22 

Maeser D, Gruener RF and Huang RS: OncoPredict: An R package for predicting in vivo or cancer patient drug response and biomarkers from cell line screening data. Brief Bioinform. 22:bbab2602021. View Article : Google Scholar : PubMed/NCBI

23 

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

24 

Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A, et al: Proteomics. Tissue-based map of the human proteome. Science. 347:12604192015. View Article : Google Scholar : PubMed/NCBI

25 

Asamura H, Nishimura KK, Giroux DJ, Chansky K, Hoering A, Rusch V and Rami-Porta R; Members of the IASLC Staging and Prognostic Factors Committee and of the Advisory Boards, and Participating Institutions, : IASLC lung cancer staging project: The new database to inform revisions in the ninth edition of the TNM classification of lung cancer. J Thorac Oncol. 18:564–575. 2023. View Article : Google Scholar : PubMed/NCBI

26 

Lei Y, Tang R, Xu J, Wang W, Zhang B, Liu J, Yu X and Shi S: Applications of single-cell sequencing in cancer research: Progress and perspectives. J Hematol Oncol. 14:912021. View Article : Google Scholar : PubMed/NCBI

27 

Wouters MCA and Nelson BH: Prognostic significance of tumor-infiltrating B cells and plasma cells in human cancer. Clin Cancer Res. 24:6125–6135. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Song P, Li W, Guo L, Ying J, Gao S and He J: Identification and validation of a novel signature based on NK cell marker genes to predict prognosis and immunotherapy response in lung adenocarcinoma by integrated analysis of single-cell and bulk RNA-sequencing. Front Immunol. 13:8507452022. View Article : Google Scholar : PubMed/NCBI

29 

Vuong L, Kouverianou E, Rooney CM, McHugh BJ, Howie SEM, Gregory CD, Forbes SJ, Henderson NC, Zetterberg FR, Nilsson UJ, et al: An orally active galectin-3 antagonist inhibits lung adenocarcinoma growth and augments response to PD-L1 blockade. Cancer Res. 79:1480–1492. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Chen S, Chen J, Hua X, Sun Y, Cui R, Sha J and Zhu X: The emerging role of XBP1 in cancer. Biomed Pharmacother. 127:1100692020. View Article : Google Scholar : PubMed/NCBI

31 

Zhong Z, Wang J, Han Q, Lin H, Luo H, Guo D, Jiang Y and Liu A: XBP1 impacts lung adenocarcinoma progression by promoting plasma cell adaptation to the tumor microenvironment. Front Genet. 13:9695362022. View Article : Google Scholar : PubMed/NCBI

32 

Glasauer A, Sena LA, Diebold LP, Mazar AP and Chandel NS: Targeting SOD1 reduces experimental non-small-cell lung cancer. J Clin Invest. 124:117–128. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Somwar R, Erdjument-Bromage H, Larsson E, Shum D, Lockwood WW, Yang G, Sander C, Ouerfelli O, Tempst PJ, Djaballah H and Varmus HE: Superoxide dismutase 1 (SOD1) is a target for a small molecule identified in a screen for inhibitors of the growth of lung adenocarcinoma cell lines. Proc Natl Acad Sci USA. 108:16375–16380. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Okazaki K, Anzawa H, Liu Z, Ota N, Kitamura H, Onodera Y, Alam MM, Matsumaru D, Suzuki T, Katsuoka F, et al: Enhancer remodeling promotes tumor-initiating activity in NRF2-activated non-small cell lung cancers. Nat Commun. 11:59112020. View Article : Google Scholar : PubMed/NCBI

35 

Lorenz J, Zahlten J, Pollok I, Lippmann J, Scharf S, N'Guessan PD, Opitz B, Flieger A, Suttorp N, Hippenstiel S and Schmeck B: Legionella pneumophila-induced IκBζ-dependent expression of interleukin-6 in lung epithelium. Eur Respir J. 37:648–657. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Fan T, Li S, Xiao C, Tian H, Zheng Y, Liu Y, Li C and He J: CCL20 promotes lung adenocarcinoma progression by driving epithelial-mesenchymal transition. Int J Biol Sci. 18:4275–4288. 2022. View Article : Google Scholar : PubMed/NCBI

37 

Li J, Li J, Hao H, Lu F, Wang J, Ma M, Jia B, Zhuo M, Wang J, Chi Y, et al: Secreted proteins MDK, WFDC2, and CXCL14 as candidate biomarkers for early diagnosis of lung adenocarcinoma. BMC Cancer. 23:1102023. View Article : Google Scholar : PubMed/NCBI

38 

Li J, Xiao X, Ou Y, Cao L, Guo M, Qi C, Wang Z, Liu Y, Shuai Q, Wang H, et al: USP51/PD-L1/ITGB1-deployed juxtacrine interaction plays a cell-intrinsic role in promoting chemoresistant phenotypes in non-small cell lung cancer. Cancer Commun (Lond). 43:765–787. 2023. View Article : Google Scholar : PubMed/NCBI

39 

Chen KC, Hsu WH, Ho JY, Lin CW, Chu CY, Kandaswami CC, Lee MT and Cheng CH: Flavonoids luteolin and quercetin inhibit RPS19 and contributes to metastasis of cancer cells through c-Myc reduction. J Food Drug Anal. 26:1180–1191. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Andersen MK, Rise K, Giskeødegård GF, Richardsen E, Bertilsson H, Størkersen Ø, Bathen TF, Rye M and Tessem MB: Integrative metabolic and transcriptomic profiling of prostate cancer tissue containing reactive stroma. Sci Rep. 8:142692018. View Article : Google Scholar : PubMed/NCBI

41 

Anagnostou V, Smith KN, Forde PM, Niknafs N, Bhattacharya R, White J, Zhang T, Adleff V, Phallen J, Wali N, et al: Evolution of neoantigen landscape during immune checkpoint blockade in non-small cell lung cancer. Cancer Discov. 7:264–276. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Steels E, Paesmans M, Berghmans T, Branle F, Lemaitre F, Mascaux C, Meert AP, Vallot F, Lafitte JJ and Sculier JP: Role of p53 as a prognostic factor for survival in lung cancer: A systematic review of the literature with a meta-analysis. Eur Respir J. 18:705–719. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Seliger B and Quandt D: The expression, function, and clinical relevance of B7 family members in cancer. Cancer Immunol Immunother. 61:1327–1341. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Jin Y, Zhang P, Li J, Zhao J, Liu C, Yang F, Yang D, Gao A, Lin W, Ma X and Sun Y: B7-H3 in combination with regulatory T cell is associated with tumor progression in primary human non-small cell lung cancer. Int J Clin Exp Pathol. 8:13987–13995. 2015.PubMed/NCBI

45 

Aggarwal C, Prawira A, Antonia S, Rahma O, Tolcher A, Cohen RB, Lou Y, Hauke R, Vogelzang N, Zandberg D, et al: Dual checkpoint targeting of B7-H3 and PD-1 with enoblituzumab and pembrolizumab in advanced solid tumors: Interim results from a multicenter phase I/II trial. J Immunother Cancer. 10:e0044242022. View Article : Google Scholar : PubMed/NCBI

46 

He D, Zhao XQ, Chen XG, Fang Y, Singh S, Talele TT, Qiu HJ, Liang YJ, Wang XK, Zhang GQ, et al: BIRB796, the inhibitor of p38 mitogen-activated protein kinase, enhances the efficacy of chemotherapeutic agents in ABCB1 overexpression cells. PLoS One. 8:e541812013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2025
Volume 29 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou W, Hu Z, Wu J, Liu Q, Jie Z, Sun H and Zhang W: Integrated analysis of single‑cell and bulk RNA sequencing data to construct a risk assessment model based on plasma cell immune‑related genes for predicting patient prognosis and therapeutic response in lung adenocarcinoma. Oncol Lett 29: 271, 2025.
APA
Zhou, W., Hu, Z., Wu, J., Liu, Q., Jie, Z., Sun, H., & Zhang, W. (2025). Integrated analysis of single‑cell and bulk RNA sequencing data to construct a risk assessment model based on plasma cell immune‑related genes for predicting patient prognosis and therapeutic response in lung adenocarcinoma. Oncology Letters, 29, 271. https://doi.org/10.3892/ol.2025.15017
MLA
Zhou, W., Hu, Z., Wu, J., Liu, Q., Jie, Z., Sun, H., Zhang, W."Integrated analysis of single‑cell and bulk RNA sequencing data to construct a risk assessment model based on plasma cell immune‑related genes for predicting patient prognosis and therapeutic response in lung adenocarcinoma". Oncology Letters 29.6 (2025): 271.
Chicago
Zhou, W., Hu, Z., Wu, J., Liu, Q., Jie, Z., Sun, H., Zhang, W."Integrated analysis of single‑cell and bulk RNA sequencing data to construct a risk assessment model based on plasma cell immune‑related genes for predicting patient prognosis and therapeutic response in lung adenocarcinoma". Oncology Letters 29, no. 6 (2025): 271. https://doi.org/10.3892/ol.2025.15017