Abstract Background The prognosis of lung adenocarcinoma (LUAD) is poor. Infection with coronavirus disease 2019 (COVID-19) may further worsen the outcome of LUAD. This study utilized the immune model and the COVID-19 receptor signal to identify the potential immune structure affecting the prognosis of COVID-19 and LUAD. Methods A prognostic model was established and verified. The correlation between immune cells and risk score was examined through a variety of immune calculation methods. Gene set variation analysis (GSVA) was used to explore the correlation between the immune signaling pathway, risk model, COVID-19 binding receptor (CO19ORS) signal, and different clinicopathological factors. Results The analysis showed that the prognosis of patients was better in the low-risk group versus the high-risk group. The tertiary lymphoid structure dominated by T and B cells (TLS1) can improve the prognosis of patients in the low-risk group. Interestingly, the CO19ORS was enriched only in females and aged >65 years. The age group >65 years is closely related to the tertiary lymphatic structure of the newborn (TLS2), while the female sex is closely related to the TLS2 and TLS1 signature. The two groups exhibited a high level of inflammation-related signal distribution. In the near future, I will collect LUAD and COVID-19 related organizations to verify the changes of 8 risk protein. Conclusion TLS1 structure may improve the prognosis of patients with LUAD and SARS-COV-2 (Severe acute respiratory syndrome coronavirus 2). This unexpected discovery provides new insight into the comprehensive treatment of patients with LUAD and SARS-COV-2. Keywords: LUAD, SARS-COV-2, TLS1, Immune prognostic model, COVID-19 binding receptor, GSVA 1. Introduction According to the latest data published by the International Cancer Research Center in 2020, lung cancer is one of the most common malignancies worldwide. Moreover, it is associated with the highest rates of morbidity and mortality among all cancers [[33]1]. Non-small cell lung cancer (NSCLC) is the primary type of lung cancer. LUAD is a predominant histological type of NSCLC. Owing to the early development of metastatic disease, and despite the use of molecular pathology testing and targeted therapies, the prognosis of patients with LUAD is poor (average 5-year survival rate: <20%) [[34][2], [35][3], [36][4]]. Since 2019, the coronavirus disease 2019 (COVID-19) pandemic poses a serious threat to the health and life of individuals worldwide. The infection is caused by a severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) and is mainly characterized by fever and inflammation of the lungs [[37]5,[38]6]. Therefore, it is important to consider the impact of infection with SARS-COV-2 on the prognosis of patients with LUAD. Furthermore, accumulating evidence suggests that the immune microenvironment significantly influences the progression of tumors and infection with SARS-COV-2, as well as the effectiveness of immunotherapy [[39][7], [40][8], [41][9], [42][10]]. Currently, the therapeutic targeting of programmed cell death 1 (PD1) and programmed cell death 1 ligand 1 (PDL1) reduces tumor volume and improves the prognosis in multiple types of cancer [[43][11], [44][12], [45][13]]. However, these treatments do not play a determinative role in immunotherapy for NSCLC, with effective treatment observed in only a limited subset of patients [[46]14]. Recent studies found that tertiary lymphoid structure (TLS) is an aggregate of immune T and B cells responding to immune stimulation. Mature TLS are often linked to B cell development, and the presence of TLS in tumors is also associated with prolonged survival in patients with various types of cancer [[47]15]. Moreover, the potential impact of TLS on SARS-COV-2 has not been investigated thus far. Therefore, the evaluation of multiple immune mechanisms and development of new immune-related therapeutic targets is necessary to improve the prognosis of patients with LUAD and COVID-19. In this study, we used a variety of databases to detect significant alterations in the expression of immune function genes, and established a risk model closely related to the prognosis of patients with LUAD. 2. Materials and methods 2.1. Data source and preprocessing The RNA-sequencing data of 535 tumors and 59 normal samples, and the corresponding data from a clinical environment, were downloaded from the Cancer Genome Atlas (TCGA) database. The clinical features of interest were age, sex, tumor stage, TNM classification, survival time, and vital status. Transcriptome profiling of 364 patients with LUAD was performed using the [48]GSE42127, [49]GSE50081, and [50]GSE115002 data sets of the Gene Expression Omnibus (GEO) database. The [51]GSE42127 and [52]GSE50081 data sets were used for validation, while [53]GSE115002 was used to identify significant differences in gene expression using the GEO2R online tools. In addition, the proteomic data of 102 normal and 109 tumor samples were obtained from the Clinical Proteomic Tumor Analysis Consortium (CPTAC) database. 2.2. Screening of immune-related differentially expressed genes (IMDEGS) According to the limma package and Wilcoxon test of R (R Vision 4.0.3), the differentially expressed genes (DEGs) were analyzed. The screening thresholds for TCGA and GEO databases were |log fold change [FC]| >1 and false discovery rate <0.05 or P-values <0.05. Additionally, P-values <0.05 and |log FC| >0.5 were used as the identification criteria for significant proteins in the CPTAC database. Firstly, 1811 immune-related genes were obtained from the tumor immune evaluation resource (TIMER) website. Next, the intersection of significant genes and proteins in multiple databases and among immune-related genes was determined. Finally, 81 IMDEGS were obtained. 2.3. Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) analyses We used the Database for Annotation, Visualization, and Integrated Discovery (DAVID) to perform GO and KEGG pathway analyses online. These analyses were conducted to further investigate the molecular function, cellular composition, biological process, and signaling pathway of these IMDEGS. 2.4. Construction of the immune-related prognostic model Initially, we used the “survival” package in R to identify IMDEGS related to prognosis. Subsequently, we implemented least absolute shrinkage and selection operator (LASSO) regression analysis based on the “glmnet” and “survival” packages for dimensionality reduction. We also calculated the analysis risk coefficient through multivariate Cox regression analysis. The risk score was computed using the following formula: risk score = (expr IMDEG1 × Coef IMDEG1) + (expr IMDEG2 × Coef IMDEG2) + … + (expr IMDEGN × Coef IMDEGN), where Coef is the coefficient obtained through the LASSO Cox regression, and expr is the expression of the signature genes. The receiver operating characteristic (ROC) curve in the “survival Roc” package was used to analyze the clinical optimal risk threshold, which was used as the basis for the classification of patients into high- and low-risk groups. 2.5. Correlations between risk models and clinical features Based on the “limma” package, differences in the risk scores of groups with different clinicopathological factors were analyzed. The “survival” package was used to investigate differences in survival between the high- and low-risk groups according to different clinicopathological factors. During the same period, univariate and multivariate Cox regression and ROC curves were used to analyze the impact and value of the risk score and multiple clinical indicators on the prognosis of patients. The calibration chart and consistency index (C-index) were determined using the “RMS” package in R to calibrate and evaluate the nomogram of various combined models. 2.6. Immune landscape exploration The immune cell infiltration for each sample in the TCGA-LUAD data set was assessed through various calculation methods, including TIMER [[54]16], CIBERSORT, CIBERSORT-ABS, QUANTISEQ, MCPCOUNTER, XCELL, and EPIC. In addition, Spearman correlation (R) and “limma” packages were used to examine the association between risk score and immune infiltration. 2.7. HALLMARK enrichment and GO analysis in different risk groups Cancer HALLMARK enrichment and GO analysis was performed in different risk groups by gene set enrichment analysis (GSEA; [55]http://www.gsea-msigdb.org/gsea/index.jsp) and using DAVID tools ([56]https://david.ncifcrf.GOv/home.jsp). A P-value <0.05 denoted statistically significant differences. Signal gene set related to thymocyte selection associated high mobility group box protein (TOX), TLS1-2, and CD8 + T cell depletion. Newborn TLS related 12 chemokines (TLS2_Signature), T, and B cells clustered in TLS-related nine genes (TLS1_Signal) [[57][17], [58][18], [59][19]]. The “ACUTE_VS_CHRONIC_LCMV_PRIMARY_IARY_INF_CD8_TCELL_UP” gene set can be considered as CD8 + T cell depletion related signal gene set, one hundred previously reported TOX-related signal genes were used and well verified in lung squamous cell carcinoma [[60][20], [61][21], [62][22]]. Seven genes were associated with COVID-19 binding receptor (CO19ORS) signaling [[63][23], [64][24], [65][25], [66][26]]. 2.8. Gene set variation analysis (GSVA) Significant differences in the enrichment of signaling pathways in the high- and low-risk groups were analyzed using the GSVA package and multiple immune-related gene sets. The screening criteria were |log FC| >0.3 and P-values <0.05. 3. Results 3.1. Identification of immune-related core genes RNA sequencing data of 535 LUAD tissues (tumor group) from TCGA database ([67]https://portal.gdc.cancer.gov/) and 59 non-cancerous tissues (healthy group) were used to perform differential expression analysis. Through this analysis, we obtained 6776 DEGs ([68]Fig. 1a). Thereafter, data from the [69]GSE115002 data set and CPTAC database were analyzed for DEGs and proteins; we obtained 5479 genes and 2234 proteins ([70]Fig. 1b and c). Approximately 1811 immune-related genes were downloaded from the TIMER database; the DEGs and proteins detected in TCGA, CPTAC, and GEO databases overlapped. Finally, the analysis yielded 81 immune-related core genes ([71]Fig. 1c). Fig. 1. [72]Fig. 1 [73]Open in a new tab Identification of the differentially expressed immune genes (IMDEGS) between the three databases. Volcanic diagram of DEGs based on the comparison of Tumour/Normal group from TCGA database (a–b). Differential protein expression levels in different groups of the CPTAC database were determined by volcano map (b). Differential genes of multiple databases (TCGA, [74]GSE115002 and CPTAC) and the immune-related genes were taken intersection (c). d-e, IMDEGs were submitted to GO (d) and KEGG (e) enrichment. 3.2. GO enrichment and KEGG pathway analyses GO and KEGG pathway analyses were performed using the DAVID website to further recognize the feature of IMDEGS; the results were visualized using R. In the GO analysis of IMDEGS, the results revealed enrichments in biological process (immunoreactivity, positive regulation of angiogenesis, hypoxia, inflammatory, immune response, positive regulation of endothelial cell proliferation, negative chemotaxis, chemokine mediated signaling pathway, and chemotaxis), cellular composition (extracellular space, extracellular domain, extracellular body, protein, and extracellular matrix), and molecular function (retinoic acid-binding, chemokine activity, retinal binding, retinol-binding, transporter activity, heparin-binding, growth factor activity, signal receptor binding, and cytokine activity) ([75]Fig. 1d). The KEGG analysis demonstrated that IMDEGS were mainly enriched in axon guidance, rheumatoid arthritis, cytokine receptor interaction, chemokine signaling pathway, phagosome, and the tumor necrosis factor (TNF) signaling pathway. The results showed that IMDEGS may be closely associated with immune reactions ([76]Fig. 1e). 3.3. Construction and analysis of the immune-related prognostic model Univariate Cox regression analysis was performed to investigate 26 prognostic IMDEGS ([77]Fig. 2a). Risk genes and coefficients were determined using LASSO regression analysis and multivariate Cox regression; finally, eight risk genes were obtained ([78]Fig. 2b and c). The expression levels of these eight genes were used to construct the immune-related prognostic model. The prognostic risk score was calculated using the following formula: risk score = (−0.185 × expr LIF receptor subunit alpha [LIFR]) + (−0.197 × expr Fetal growth restriction [FGR]) + (−0.127 × expr WAP four-disulfide core domain 2 [WFDC2]) + (0.164 × expr semaphorin 4B [SEMA4B]) + (0.728 × expr Fibroblast growth factor 2 [FGF2]) + (0.151 × expr C-C motif chemokine ligand 20 [CCL20]) + (−0.211 × expr growth differentiation factor 10 [GDF10]). Fig. 2. [79]Fig. 2 [80]Fig. 2 [81]Open in a new tab The technological process of constructing the immune-related risk model. The prognosis-related IMDEGS were performed by univariate analysis (a). The figures of the LASSO method (most minor absolute shrinkage and selection operator) and coefficient profiles (b). Multivariate Cox regression analyses were performed on the forest plots (c). Optimal thresholds points were obtained by the operating characteristic (ROC) curve (d). the distribution of the risk score (e). The survival status of patients was plotted in different risk groups and scores (f–h). The analysis of the time-dependent ROC curve was performed for patients in 1,2,3 years (i). Kaplan-Meier curves of the correlative models of immune in TCGA (training set) (j), [82]GSE42127 and [83]GSE50081 (testing set 1, 2) (k–l), The distribution of different genes in high/low-risk groups was displayed as box-and-whisker plot (m). Association between eight prognosis-related genes with risk score (n). According to the analysis performed using the “survival rock” package in R ([84]Fig. 2d) the best-predicted risk coefficient of the risk model was 1.025; this coefficient was used as the basis for the classification of patients into high- and low-risk groups. Also, our data showed that the mortality rate was higher in the high-risk group than the low-risk group ([85]Fig. 2e–h). Kaplan–Meier survival curves for TCGA training data set indicated that the survival rate was higher in the low-risk group versus the high-risk group ([86]Fig. 2i). In the training set, the areas under the curve (AUCs) of the time-dependent ROC curve were 0.697, 0.712, and 0.716 for 1, 2, and 3-year OS predictions, respectively ([87]Fig. 2j). These findings confirmed that the immune-related risk model was sufficiently accurate to predict the prognosis of patients with LUAD. After performing a GEO data set analysis, we also conducted a validation study using the validation groups established by the same risk model and scoring formula to validate the stability of the model. The predictive efficacy of the model was validated using validation set1 and set2 by Kaplan–Meier analysis ([88]Fig. 2k and l). The expression levels of LIFR, FGR, WFDC2, CD79A, and GDF10 was higher in the low-risk groups than the high-risk group. There was a negative association with the risk score, contrary to the expression of SEMA4B and CCL20 in different risk groups and risk scores ([89]Fig. 2m, n). 3.4. Associations between risk score and clinicopathological factors The interrelationships between the risk score and clinical features of patients with LUAD were evaluated. The risk score was associated with sex (P = 0.003), clinical stage (P = 7.7e-07), T classification (P = 0.0016), and N classification (P = 0.00012) ([90]Fig. 3a–f). Meanwhile, according to the survival analysis of different risk groups based on the clinical characteristics, we found that the high-risk group (age: >65, ≤65 years; sex: male and female; stage: I–II, M0, T1–2, T3–4, and N1-2) was characterized by a low survival rate ([91]Fig. 3g). Fig. 3. [92]Fig. 3 [93]Fig. 3 [94]Open in a new tab The correlation of the various clinical features with the Risk Score. Spearman was performed the association between risk score and different clinical parts (a–f). The difference of survival between high and low-risk groups in other clinical characteristics (g). Univariate and multivariate Cox regression models identified that TMN stage and risk scores were significantly associated with prognosis ([95]Fig. 4a and b). The ROC curve results showed that the predictive performance of the model for the rate of overall survival was excellent. [96]Fig. 4c shows the AUC values of the various clinical features and risk scores: age (AUC = 0.471), sex (AUC = 0.558), TMN stage (AUC = 0.669), T classification (AUC = 0.576), M classification (AUC = 0.496), N classification (AUC = 0.632), and risk score (AUC = 0.697). Subsequently, we constructed a nomogram using the risk scores and TMN stages for the prediction of the patient ([97]Fig. 4d, e). The C-index of the nomogram was 0.69 (95% confidence interval: 0.6468–0.73312), and a calibration plot showed optimal agreement compared with the actual observation. Fig. 4. [98]Fig. 4 [99]Open in a new tab Effect of risk model and various clinicopathological features on the prognosis of patients. The immune-related prognostic model and Clinical features were analyzed via the Univariate, multivariate Cox, and receiver operating curve (ROC) (a–c). Based on the immune-related model and more prominent clinical factors, a nomogram was built for predicting 1-, 2- and 3-year survival rates of LUAD, and the prognostic accuracy of the nomogram was shown by calibration plots (d–e). 3.5. Composition of immune cells in the risk model We estimated the immune cell composition of TCGA-LUAD through multiple calculation methods from the TIMER website ([100]Fig. 5a) to analyze the association between risk score and immune cells. The risk score exhibited a generally negative correlation with B cells and CD8 T cells. There were significant differences in B, CD8, and myeloid dendritic cells between the low- and high-risk groups; B cell infiltration was the most prominent among them ([101]Fig. 5b–e). The distribution differences in the immune score, matrix score, and risk score, were calculated by x cell under different risk groups. Based on the data, we concluded that risk score had a significant negative association with the immune score and stroma score, which were higher in the low-risk group versus the high-risk group ([102]Fig. 5f). Fig. 5. [103]Fig. 5 [104]Fig. 5 [105]Open in a new tab Research of different risk groups of immune infiltration. Spearman correlation analysis was computed between risk scores and immune infiltration, using multiple calculation methods from TIMER Database (a–e). Distribution of the microenvironment score and composition of immune cells in different risk groups (f). (_A: TIMER,_B: CIBERSORT, _C: CIBERSORT-ABS, _D: QUANTISEQ, _E: MCPCOUNTER, _G: XCELL, _H: EPIC). 3.6. Distribution of immune-related genes in different risk groups The genes related to B cells (CD20, CD23, marginal zone B and B1 cell specific protein [MZB1], joining chain of multimeric IgA and IgM [JCHAIN], immunoglobulin lambda-like [IGLL0]), CD8 cells (interleukin 2 [IL2], interleukin 4 [IL4], CD8, C-X-C motif chemokine receptor 4 [CXCR4], C-X3-C motif chemokine receptor 1 [CX3CR1]), CD4 T cells (CD4), follicular dendritic cells (CD21), and primary T cells (TCF7) were highly expressed in the low-risk group ([106]Fig. 6a). We also found that hypoxia inducible factor 1 subunit alpha (HIF1A) was highly expressed in the high-risk group and positively associated with the risk score ([107]Fig. 6 b). CD8 cells, B cells, TOX, and TLS1 signatures were higher in the low-risk group versus the high-risk group. These signals were negatively correlated with the risk score ([108]Fig. 6c, d). Fig. 6. [109]Fig. 6 [110]Open in a new tab Distribution level of tertiary lymphatic structure (TLS2, TLS1) in different risk groups. (a–b) The difference of the T, B, and dendritic cell-related genes in the high/low-risk group. (c) Based on GSVA analysis and GO data set in GSEA, (d) the distribution levels of CD8 cells, B cells, TLS1, TLS2, CD8+T cell Exhaustion, and Tox-related signal pathways in high and low-risk groups and their correlation with the riskscore.(GO_1:B_CELL_HOMEOSTASIS,GO_2:B_CELL_PROLIFERATION,GO_3:B_CEL L_MEDIATED_IMMUNITY,GO_4:B_CELL_DIFFERENTIATION,GO_5:B_CELL_ACTIVATION, GO_6:B_CELL_RECEPTOR_SIGNALING_PATHWAY,GO_7:B_CELL_ACTIVATION_INVOLVED_ IN_IMMUNE_RESPONSE,GO_8:CD8_POSITIVE_ALPHA_BETA_T_CELL_ACTIVATION,GO_9: CD8_POSITIVE_ALPHA_BETA_T_CELL_DIFFERENTIATION). 3.7. GO analysis of different risk groups The high-risk group exhibited enrichments in the immune-related pathways of cellular signal transduction, the defense response of Gram-negative bacteria, chemotaxis, bacterial defense response, neutrophil chemotaxis, and hypoxia response ([111]Fig. 7a). The low-risk group exhibited enrichments in the immune-related pathways of the antibacterial immune response, humoral reaction, complement activation, receptor-mediated endocytosis, classical pathway, leukotriene biosynthesis process, B cell receptor signaling pathway, and positive regulation of B cell activation ([112]Fig. 7b). Fig. 7. [113]Fig. 7 [114]Open in a new tab Enrichment analysis of highly expressed genes in different risk groups. GO analysis of the highly expressed genes of high/low-risk groups, respectively (a–b). KEGG enrichment analysis of upregulated genes of the high and low-risk groups, respectively (c–d). 3.8. Analysis of the KEGG pathways by GSEA Thereafter, we conducted a specific pathway enrichment analysis using GSEA for different risk groups according to the KEGG data set. The low-risk group was enriched in arachidonic acid metabolism, linolenic acid metabolism, and production of intestinal immune network ([115]Fig. 7d). The high-risk group was enriched in DNA replication, p53 signaling pathway, cell cycle, glycolysis, galactose metabolism, gluconeogenesis, oocyte meiosis, and renal cell carcinoma ([116]Fig. 7c). 3.9. Association of CO19RS signaling with immunity Immune and stromal scores, TLS2, TLS1, and TOX signals were highly distributed in the high-CO19ORS expression group, and were directly proportional to the expression of CO19ORS ([117]Fig. 8a–d). For the clinicopathological factors, the distribution level of the CO19ORS signal in the age and sex groups differed; the highest concentration was observed in females and aged >65 years ([118]Fig. 8e and f). The expression of CO19ORS-related genes (AXL, epidermal growth factor receptor [EGFR], and neuropilin 1 [NRP1]) was higher in females versus males. The levels of other related genes (AXL receptor tyrosine kinase [AXL], angiotensin converting enzyme 2 [ACE2], and CD209) were higher in patients aged >65 years versus those aged ≤65 years ([119]Fig. 8k, l). Fig. 8. [120]Fig. 8 [121]Fig. 8 [122]Open in a new tab Correlation between COVID-19 binding receptor, immune response and different pathological parameters. (a) Distribution of immune and matrix scores in high and low-COVID-19 receptor group. (b) Correlation between COVID-19 binding receptor and immunity and matrix score. (c) Distribution of immune-related signals and risk scores in high and low COVID-19 binding receptor groups. (d) Correlation between immune-related signals, risk score, and high and low COVID-19 combined receptor score. e-j Distribution level of COVID-19 binding receptors under different clinical and pathological factors. (e) age. (f) gender. (g) M. (h) N. (I) stage. (j) T. k-l, Expression levels of COVID-19 binding receptors in different age and gender groups, (k) gender. (l) age. 3.10. Differences in the enrichment of TOX, TLS2, and TLS1 signatures between groups The highest TLS2, TLS1, and TOX signals were observed in those aged >65 years with high expression of CO19ORS (Group 1) and females with high expression of CO19ORS (Group 3) ([123]Fig. 9a–d). In Group 1, the TLS2 and TOX signals were proportional to the CO19ORS signal ([124]Fig. 9e). In those aged ≤65 years with low expression of CO19ORS (Group 2), the CO19ORS signal was directly proportional to the TOX signal ([125]Fig. 9f). In Group 3 and Group 4 (males with low expression of CO19ORS), there was a positive correlation between the signals of the CO19ORS, TLS2, TLS1, and TOX ([126]Fig. 9g and h). Fig. 9. [127]Fig. 9 [128]Open in a new tab Using GSVA analysis, the enrichment levels of TOX, TLS1, and TLS2 signals in different groups Age>65-high-COVID-19 binding receptor (Group1) V Age ≤ 65-Low-COVID-19 binding receptor(Group2), and FEMALE-high-COVID-19 binding receptor(Group3) V MALE-Low-COVID-19 binding receptor (Group4), a-b Group1v2. (a) heatmap. (b) Deviation diagram. c-d Group3v4. (c) heatmap. (d) Deviation diagram. e-h, The correlation degree between TLS2, TOX, TLS1 and COVID-19 binding receiver signals under different groups. (e) Group1. (f) Group2. (g) Group3. (h) Group4. 3.11. Inflammatory signaling pathways Based on the GSVA analysis and HALLMARK data set, we found that inflammation-related pathways, including the IL6/JAK/STAT3 signaling and inflammatory response, were highly expressed in Group 1 ([129]Fig. 10a–c) and Group 3 ([130]Fig. 10d–f), compared with Groups 2 and 4. Fig. 10. [131]Fig. 10 [132]Open in a new tab Based on GSVA analysis, HALLMARK-related signal pathways are enriched in different groups Group1) V Group2, Group3 V Group4. a-c, Inflammation related signaling pathways were distributed at Group1. (a) Volcan, (b) Heatmap, (c) Deviation diagram. d-f, Inflammation related signaling pathways were distributed at Group3. (d) Volcan, (e) Heatmap, (f) Deviation diagram. 4. Discussion Lung adenocarcinoma is the most common pathological subtype of NSCLC (30–40%) [[133]27]. NSCLC poses a significant public health challenge, and is the leading cause of cancer-related death worldwide. The study and development of various therapeutic have suggested that the immune damage is decisive factors in the development of cancer. With the application of cancer immunotherapy, it has been shown that some immunomodulators also play an increasingly important role in the treatment of LUAD [[134][28], [135][29], [136][30], [137][31], [138][32]]. SARS-COV-2 current causes severe lung disease globally [[139]7]. This ongoing phenomenon undoubtedly complicates the treatment of LUAD. Therefore, we strive to improve the prognosis of patients with COVID-19 and LUAD through the development of new immunotherapies. In this study, we obtained 81 core IMDEGS based on the intersection of immune-related genes and DEGs reported in TCGA, GEO, and CPTAC databases. The IMDEGS exhibited enrichments in immune response, positive regulation of angiogenesis, hypoxia response, inflammatory response, negative chemotaxis, positive regulation of endothelial cell proliferation, chemokine mediated signaling pathway, chemotaxis, etc. According to the KEGG pathway analysis, the IMDEGS were predominantly enriched in rheumatoid arthritis, cytokine receptor interaction, phagosome, TNF signaling pathway, and the chemokine signaling pathway. The results showed that IMDEGS may be closely related to immune response. Eight genes significantly associated with prognosis were selected to create an immune-related prognostic model. As demonstrated by univariate and multivariate Cox regression analyses, as well as LASSO regression analysis, the model showed good prognostic accuracy. Moreover, we found that the high-risk group had a lower overall survival rate versus the low-risk group; of note, the classification into risk groups was based on the optimal threshold of the training (TCGA) and the validation ([140]GSE42127, [141]GSE50081) data sets. In addition, the relationship and prognostic value of the risk score with different clinical indicators was well verified. The nomogram – a statistical model based on the risk model and stage (the most prominent prognostic factors) – provided a more individualized prediction. In summary, the results demonstrated that the established immune-related model was stable and accurate. Meanwhile, we analyzed differences in the biological functions between the different risk groups. In the high-risk group, enrichment was noted in C-C motif chemokine receptor 6 (CCR6) chemokine receptor binding, Gram-negative bacteria defense response, bacterial defense response, neutrophil chemotaxis, and hypoxia response. In the low-risk group, enrichment was observed in the signal of the receptor-mediated endocytosis, positive regulation and phagocytosis of B cell activation recognized leukotriene biosynthesis, immune response, complement activation, classical pathway, B cell receptor signaling pathway, antibacterial humoral response, and immunoglobulin receptor binding. Through the GO analysis of different risk groups, we established that the related paths of B cell and humoral immunity were ubiquitous in the low-risk group. We also found that the hypoxic environment may inhibit B cell and humoral response in the low-risk group. The high expression group significantly enriched hypoxia related pathways and genes. Recent studies have also suggested that hypoxia may affect the development of B cells and humoral response [[142]33]. Using the TIMER database, we examined the correlation between immune cell infiltration and risk score in LUAD. We found that B cells, CD8 cells, and TLS1-related signals were highly enriched in the low-risk group. Moreover, immune genes related to B cells, CD4 cells, CD8 cells, and dendrites were also highly expressed in the low-risk group, indicating that the low-risk group also had TLSs [[143][34], [144][35], [145][36], [146][37]]. Recent studies have found that extensive B cell infiltration, or accompanied by the TLS system of the CD20 + B cells in sarcoma, melanoma, renal cell carcinoma, and lung cancer contribute to the prognosis of patients [[147]19,[148]35,[149]38,[150]39]. Previous studies have shown that CD8 T cells play an essential role in the anti-tumor immune response and are positively correlated with the overall survival rate of patients with various malignant tumors (including lung cancer) [[151]40,[152]41]. Interestingly, the TLS1, TOX and CD8 T cell depletion signaling pathways were highly enriched in the low-risk group. T cell depletion leads to T cell dysfunction, which occurs in the tumor microenvironment. This process is often responsible for the inability of the immune system to eliminate the tumor [[153]42,[154]43]. Several investigations have reported that TOX regulates the critical factor of CD8 cell depletion [[155][44], [156][45], [157][46]]. This finding suggests that TLS1 counteracts the impact of CD8 cell depletion on the prognosis of patients. Among the eight genes included in the risk model, LIFR, FGR, WFDC2, GDF10, and CD79A were upregulated. In the low-risk group, SEMA4B and CCL20 were downregulated. FGF2 did not show statistically significant differences in any group. As a serum tumor marker, the upregulation of LIFR can inhibit the proliferation and invasion of lung cancer cells, and promote their apoptosis [[158]47]. WFDC2 plays essential roles in the early diagnosis of lung cancer and monitoring of treatment efficacy, and is significantly correlated with the TNM stage and prognosis of LUAD. Overexpression of WFDC2 can repress the metastatic potential of prostate cancer by inhibiting the EGFR signal [[159]48,[160]49]. SEMA4B restrains the growth of NSCLC in vitro, ex vivo, inhibited by hypoxia and HIF1A, promoting the invasion of NSCLC [[161][50], [162][51], [163][52]]. The downregulation of FGF2 inhibits the invasion of breast cancer, osteosarcoma, and LUAD [[164][53], [165][54], [166][55], [167][56]]. The overexpression of CCL20 promotes the progression of lung cancer and breast epithelial cells. Furthermore, it recruits regulatory T cells through forkhead box O1/CCAAT enhancer-binding protein/nuclear factor-kB (FOXO1/CEPB/NF-kB) signal transduction to promote drug resistance in colorectal cancer cells [[168][57], [169][58], [170][59]]. The downregulation of GDF10 promotes the proliferation of prostate cancer and the invasion of hepatocellular carcinoma [[171]60,[172]61]. It has been shown that high CD79A expression was beneficial to the prognosis of cervical cancer [[173]62]. In addition, FGR expression is associated with decreased cytotoxic T-lymphocyte infiltration and poor prognosis in colorectal cancer. Other studies have demonstrated that FGR plays a role in the myeloid cell-mediated inflammatory response in vivo [[174]63,[175]64]. Ordinary model construction is based on gene sequencing; immune-related genes are evaluated based on the intersection of differential genes and proteins. The survival results obtained from multiple data sets were consistent. Additionally, the model was associated with a good ROC curve and C-index, which confirmed its stability and accuracy. We downloaded the immune invasion data through various immune calculation methods to ensure the accuracy of immune cell scores for different risk groups. Moreover, the enrichment analysis of a variety of immune-related gene sets further verified the results of immune cell infiltration. The gene set of CO19ORS signal is composed of several potential and cellular receptors that overlap with those of COVID-19, including ACE2, AXL, EGFR, LDLR, NRP1, CD209, and CLEC4M [[176][23], [177][24], [178][25], [179][26]]. The signal was mainly noted in females and patients aged >65 years. There was a marked difference in the CO19ORS between the age and sex groups. Females and patients aged >65 years were linked to high enrichment in COVID-19 receptor signals. It is likely that such patients are susceptible to COVID-19. Also, there was a significant correlation between the COVID-19 signaling pathway and immune score, TLS2, TLS1, and TOX signals. These results suggested that females and patients aged >65 years may exhibit higher immunoreactivity. In Group 1, the receptor signal was positively correlated with the TOX and TLS2 signatures. In Group 2, the receptor was only related to the TOX signal. Inflammatory signals were mainly concentrated in Group 1 compared with Group 2. In recent reports, the mortality and reinfection rates were higher in patients with COVID-19 aged >65 years versus those aged ≤65 years [[180]65,[181]66]. In Groups 3 and 4, the TOX, TLS2, and TLS1 signatures were positively correlated with the COVID-19 receptor signaling pathway. In Groups 3 and 4, inflammatory-related signals were mainly concentrated in Group 3. In numerous reports, SARS-COV-2 was associated with mild symptoms and a low mortality rate in females versus males [[182][67], [183][68], [184][69], [185][70], [186][71], [187][72]]. Hence, TLS1 may play an important role in the prognosis of female patients. The adaptive immune response dominated by T and B cells plays a positive role in resisting SARS-COV-2 infection [[188][73], [189][74], [190][75]]. The inflammatory reaction often increases the damage caused by SARS-COV-2 to patients [[191]76,[192]77]. Meanwhile, TLS1 is closely related to B cells and CD8 cells in TLS [[193]18,[194]19]. TLS2 is closely related to newborn TLS, and is accompanied by inflammatory reaction [[195]17,[196]78,[197]79]. Therefore, in Group 1, the close relationship of TLS2 with the CO19RS may increase the damage induced by SARS-COV-2 in patients aged >65 years. In Group 2, the close relationship of TLS1 with the CO19RS may reduce the inflammation-related damage and prevent the damage caused by SARS-COV-2 infection. 5. Conclusion According to the present immune-related model, TLS1 may improve the prognosis of patients with LUAD. The differences observed in the correlation and enrichment level of CO19ORS, TLS2, and TLS1 in different age and sex groups further imply that TLS1 is associated with resistance to SARS-COV-2 infection. Therefore, sufficient construction of T cell- and B cell-based TLSs may improve the prognosis of patients with LUAD and COVID-19. Author contribution statement Kang Sun: Performed the experiments; Wrote the paper. Zhiqiang Zhang; Dongqin Wang; Hongyu Ma: Analyzed and interpreted the data. Jing Zhang: Conceived and designed the experiments. Chaoqun Lian: Conceived and designed the experiments; Contributed reagents, materials, analysis tools or data. Funding statement Chaoqun Lian was supported by the Key Natural Science Project of Anhui Provincial Education Department (KJ2021A0773), National Innovation Program for College Students (202210367076), Anhui Provincial Undergraduate Innovative Training Program (S202110367030). Jing Zhang was supported by the Key Natural Science Project of Anhui Provincial Education Department (KJ2020A0578). Dongqin Wang was supported by the Program for Graduate Research of Bengbu Medical College (Byycxz22016). Data availability statement The authors do not have permission to share data. Declaration of interest's statement The authors declare no conflict of interest Contributor Information Jing Zhang, Email: jade.zhangjing@bbmc.edu.cn. Chaoqun Lian, Email: lianchaoqun@bbmc.edu.cn. Abbreviation COVID-19 Coronavirus disease 2019 CO19ORS COVID-19 binding receptor CPTAC Clinical Proteomic Tumor Analysis Consortium DEGs Differentially expressed genes GEO Gene Expression Omnibus GO Gene Ontology Group 1 Aged >65 years with high expression of CO19ORS Group 2 Aged ≤65 years with high expression of CO19ORS Group 3 Female with high expression of CO19ORS Group 4 Male with low expression of CO19ORS IMDEGS Immune-related differentially expressed genes KEGG Kyoto Encyclopedia of Genes and Genomes LUAD Lung adenocarcinoma NSCLC Non-small cell lung cancer SARS-COV-2 Severe acute respiratory syndrome-coronavirus-2 TCGA The Cancer Genome Atlas TIMER Tumor Immune Estimation Resource TLS Tertiary lymphoid structures TLS1 Tertiary lymphoid structure signal dominated by T and B cells TLS2 Tertiary lymphatic structure of the newborn References