-
Six IATGs (EGFR, ANXA5, CLEC4M, CD209, UVRAG, and CACNA1C) were retrieved from the VThunter database (URL: https://db.cngb.org/VThunter/). Selected “Orthomyxoviridae” from the “Virus Family” dropdown menu and “Influenza A virus” from the “Virus” dropdown menu, then the target genes of Influenza A virus were shown in the “Target Gene” dropdown menu. The VThunter database is an up-to-date and accessible database specifically created to examine and analyze the manifestations of viral receptors in the tissues of various animal species at the single-cell level. This database identified 107 viral receptors in 142 viral species and acquired accurate expression profiles using 285 scRNA-seq datasets, which cover 2,100,962 cells from 47 distinct animal species[23].
-
We procured human kidney renal cell carcinoma cell lines (786-O, ACHN, and Caki-1) and HK-2, a proximal tubular cell line derived from the normal kidney from the American Type Culture Collection (ATCC, VA, USA). The primers were synthesized by Sangon Biotech (Shanghai, China). Cells were cultured and collected for qRT-PCR analysis as described previously[24,25]. Briefly, 50,000 cells from each of the cell lines indicated above were plated in 6-well plates. Total RNA was collected and isolated from cell cultures after the cells reached confluence using the TRIzol reagent (Invitrogen, NY, USA). iScript cDNA synthesis reagent (Bio-Rad, Hercules, CA, USA) was used to synthesize cDNA. β-actin was utilized as an internal control. The primer sequences are listed below.
-
Public databases were accessed to acquire tumor sample-related data from The Cancer Genome Atlas (TCGA) (https://portal.gdc.cancer.gov/)[26], and mRNA sequencing, clinical, single-nucleotide variants (SNV), copy number variants (CNV), and methylation data from the GSCA database (http://bioinfo.life.hust.edu.cn/GSCA)[27]. The details have been described previously[24-25]. Reverse phase protein array (RPPA) data retrieved from The Cancer Proteome Atlas (TCPA) database were used for pathway analysis[28]. The correlation between gene expression and drug sensitivity was based on the Genomics of Drug Sensitivity in Cancer (GDSC) database[29]. Samples collected from thirty-three cancer types were included in pan-cancer analysis. Detailed cancer types and cases are listed in Supplementary Table S1 (available in www.besjournal.com).
Table 1. Primers
Gene Forward sequence 5'-3' Reverse sequence 5'-3' EGFR TTGCCGCAAAGTGTGTAACG GTCACCCCTAAATGCCACCG CACNA1C AATCGCCTATGGACTCCTCTT GCGCCTTCACATCAAATCCG CLEC4M GAGTAACCGCTTCTCCTGGATG CGCACAGTCTTCATTCCCGCTA ANXA5 AACCCTCTCGGCTTTATGATGC CGCTGGTAGTACCCTGAAGTG CD209 TCAAGCAGTATTGGAACAGAGGA CAGGAGGCTGCGGACTTTTT UVRAG CTTGGGTCAGCAGATTCATGC CATCGTAAGAATTGCGAACACAG Table S1. Cancer types and cases
Cancer type (Abbreviation) Cases Adrenocortical carcinoma (ACC) 92 Breast cancer (BRCA) 1,218 Bladder uroepithelial carcinoma (BLCA) 411 Cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) 310 Cholangiocarcinoma (CHOL) 45 Colon adenocarcinoma (COAD) 329 Lymphoid neoplasm diffuse large B-cell lymphoma (DLBC) 48 Head and neck squamous cell carcinoma (HNSC) 566 Esophageal carcinoma (ESCA) 196 Glioblastoma multiforme (GBM) 174 Kidney chromophobe (KICH) 91 Kidney renal clear cell carcinoma (KIRC) 606 Kidney renal papillary cell carcinoma (KIRP) 323 Acute myeloid leukemia (LAML) 173 Brain lower grade glioma (LGG) 534 Liver hepatocellular carcinoma (LIHC) 359 Lung adenocarcinoma (LUAD) 576 Thyroid cancer (THCA) 572 Thymoma (THYM) 122 Uterine corpus endometrial carcinoma (UCEC) 201 Uterine carcinosarcoma (UCS) 57 Uveal melanoma (UVM) 80 -
Only fourteen cancer types were included (COAD, ESCA, LUSC, KIRC, HNSC, PRAD, BRCA, BLCA, THCA, STAD, KIRP, LUAD, LIHC, and KICH) in the mRNA expression analysis. The criterion was that the paired tumor and normal samples collected from the list of cancer types were more than ten. The mRNA expression values in TCGA are expressed as normalized RSEM values. Fold change was calculated as mean (tumor)/mean (normal), as described previously[24,25]. Moreover, the tumor samples were classified into two groups (high and low) based on the median values and further analyzed for the correlation between expression and survival.
-
SNV and clinical survival data from thirty-three cancers were extracted from TCGA database. Using the unique barcoding of each specimen, SNV and clinical survival data were merged. Mutated tumor specimens were identified based on the presence of certain mutated genes. For the survival analysis, at least two groups with two or more samples were included. The survival rate of the R package was used to match the survival time and status. Differences in survival between the wild-type and mutant groups were determined using the Cox proportional hazards model and log-rank test. Eight mutation types were included in the analysis: deleterious mutations, missense mutations, nonsense mutations, frame-shift insertions, splice-sites, frame-shift deletions, in-frame deletions, and in-frame insertions.
-
CNV data from thirty-three cancer types were collected from TCGA and analyzed using GISTICS 2.0[30]. The GISTIC database was used to identify significantly altered regions of amplification or deletion in the patient cohorts. This study explored the copy number level of each gene in the gene set in each pan-cancer cancer based on the GISTIC score derived from GISTIC and then summarized the four types of GISTIC scores: homozygous deletion, heterozygous deletion, heterozygous amplification, and homozygous amplification. Spearman’s correlation analysis was performed by merging the mRNA expression data with raw CNV data[31]. The false discovery rate (FDR) was used to adjust the P-value. A log-rank test was performed to examine the differences in survival between groups. SNV data and clinical survival data were merged using specimen barcoding. For the survival analysis, at least two groups with two or more samples were included. The R package for survival was used to fit survival time and status within each group. A log-rank test was performed to test survival differences between the groups.
-
Methylation analysis was performed based on the chosen fourteen cancer types with more than ten paired tumors and adjacent normal tissues. Differences in methylation levels between tumor and normal samples were determined using Student’s t-test. Spearman analysis was used to determine the correlation between the mRNA expression and methylation levels of the genes. Median methylation data were used for survival analysis after categorizing tumor samples into two groups (hypermethylated and hypomethylated). The FDR was also used to adjust the P-value.
-
Calculations were performed on ten cancer-related cell signaling pathways for thirty-three cancer types, including the TSC-mTOR pathway, receptor tyrosine kinases (RTKs), Ras/Raf/MAPK pathway, PI3K-AKT pathway, hormone estrogen receptors (ERs), androgen receptor (AR), EMT, DNA damage, cell cycle, and apoptotic pathways[32]. The activity scores of the listed pathways and gene expression between pathways (activation and repression) were determined using the median pathway scores[33].
First, all included data were grouped into high and low expression groups based on median gene expression values. A t-test was used to determine the difference in pathway activity scores (PAS) between the groups, and FDR was used to adjust the P-value. When PAS of the group with gene A in high expression was greater than PAS of the other group with gene A in low expression, gene A might activate a verified signaling pathway; otherwise, it might have a repressive effect on this pathway[33].
-
Gene set enrichment analysis (GSEA) calculations were performed using the R package fgsea[34]. By using the normalizing entrichment scores (NES), GSEA considered the differences in the sizes of the IATG gene sets and correlations within the expression datasets. NES was used to compare the results of the analyses across gene sets.
-
The IC50 values of 265 selected compounds with PubChem ID and the corresponding gene expression data in the GDSC2 dataset were collected from 860 tumor cell lines from the Genomics of Drug Sensitivity in Cancer database (GDSC; URL: https://www.cancerrxgene.org/). Compounds without valid PubChem ID were excluded from the study. These compounds are cytotoxic chemotherapeutics and targeted therapeutics that are acquired from commercial sources, academic researchers, and biopharmaceutical companies. The pathways targeted by these compounds included ABL signaling, apoptosis regulation, cell cycle, chromatin histone acetylation/methylation, cytoskeleton, kinases, DNA replication, EGFR signaling, ERK/MAPK signaling, genome integrity, hormone-related pathways, IGF1R signaling, immune response, JNK and p38 signaling, metabolism, mitosis, and other unclassified pathways. Pearson correlation analysis was performed to determine the relationship between the mRNA expression of the gene and the IC50 of the drug. The P-value was adjusted using FDR. A positive correlation implies that high gene expression suggests drug resistance.
-
The ImmuCellAI algorithm was used to calculate the infiltration of 24 immune cell lines and expressed as a correlation coefficient[35]. The association of immune cell infiltration with the GSVA scores of IATGs was analyzed using Spearman’s correlation with P-value adjusted by FDR. A set of marker genes for three immune-related pathways, including chemotactic cytokines, the MHC class I antigen presentation pathway, and immunostimulators, was obtained from the TISIDB database[36]. The relationship between IATGs and the three immune-related pathways was analyzed using the GEPIA2 database (Pearson’s coefficient)[37].
-
RNA-sequencing expression profiles and corresponding clinical information for IATGs in KIRC were downloaded from the TCGA dataset, while those for normal control group were downloaded from GTEx database (https://gtexportal.org/home/)[38]. The test for differential expression of genes was performed using the Wilcoxon rank sum test.
-
Unless otherwise stated, all statistical analyses were performed using the GraphPad Prism (version 8.0.1) and R software (version 4.0.2). Correlation analysis was conducted using the Spearman’s correlation test. Survival risk and HR were calculated using the Cox proportional risk model. The “survival” R program was used to examine the two groups’ survival time and survival status. The log-rank test was used for comparative analysis. The rank sum test was used to identify data from both groups, and a P-value < 0.05 or FDR ≤ 0.05 was considered statistically significant. Genes and cancer types with a P-value of less than 0.05 were shown. The significance of the differences between the two subgroups was evaluated using the Mann–Whitney U test (n < 5). One-way ANOVA and Bonferroni’s post hoc tests were used to perform multiple comparisons. P < 0.05 was deemed statistically significant. Independent qRT-PCR analyses were performed in triplicates.
doi: 10.3967/bes2024.094
Multi-omics Approach Reveals Influenza-A Virus Target Genes Associated Genomic, Clinical and Immunological Characteristics in Cancers
-
Abstract:
Objective To examine the precise function of influenza A virus target genes (IATGs) in malignancy. Methods Using multi-omics data from the TCGA and TCPA datasets, 33 tumor types were evaluated for IATGs. IATG expression in cancer cells was analyzed using transcriptome analysis. Copy number variation (CNV) was assessed using GISTICS 2.0. Spearman’s analysis was used to correlate mRNA expression with methylation levels. GSEA was used for the enrichment analysis. Pearson’s correlation analysis was used to examine the association between IATG mRNA expression and IC50. The ImmuCellAI algorithm was used to calculate the infiltration scores of 24 immune cell types. Results In 13 solid tumors, IATG mRNA levels were atypically expressed. Except for UCS, UVM, KICH, PCPG, THCA, CHOL, LAMI, and MESO, most cancers contained somatic IATG mutations. The main types of CNVs in IATGs are heterozygous amplifications and deletions. In most tumors, IATG mRNA expression is adversely associated with methylation. RT-PCR demonstrated that EGFR, ANXA5, CACNA1C, CD209, UVRAG were upregulated and CLEC4M was downregulated in KIRC cell lines, consistent with the TCGA and GTEx data. Conclusion Genomic changes and clinical characteristics of IATGs were identified, which may offer fresh perspectives linking the influenza A virus to cancer. -
Key words:
- Genomic changes /
- Immune microenvironment /
- Prognosis /
- Drug resistance /
- Experimental validation
The authors declare that there is no conflict of interests.
&These authors contributed equally to this work.
注释:1) AUTHOR CONTRIBUTIONS: 2) CONFLICT OF INTERESTS: -
Figure 2. Expression and survival analysis of IATGs. Only genes with significant differential expression are shown. (A) Different expression profiles of IATGs in tumor samples compared to normal samples. (B) Survival differences of IATGs. The size of the dots shows the importance of the influence of the gene on survival for every type of cancer; the P-value was calculated using the Kaplan–Meier method. Red dots indicate a low survival rate of gene expression in the stated cancer type, whereas blue dots indicate a high survival rate of gene expression in the indicated cancer type. FDR, false discovery rate; IATGs, influenza-A virus target genes.
Figure 3. Frequencies of SNVs and variant forms of IATGs. (A) IATG mutation frequencies. The numbers indicate the mutations of the identified gene in a specific type of cancer. “0” shows that there were no mutations in the coding region of the gene, and “null” shows that there were no mutations in any area of the gene. (B) A waterfall graph of tumor distribution illustrates the spectrum of mutations in IATGs as well as the categorization of SNV types. (C) The connection between SNV and the survival of IATGs. Risk ratios and Cox P values are indicated by the size and color of the bubbles. The hue of the bubbles, from blue to red, represents low to high hazard ratios, and bubble size is positively connected with the Cox P value. Black border outlines show Cox P values ≤ 0.05. SNV, single-nucleotide variants; IATGs, influenza-A virus target genes.
Figure 4. Significant role of CNVs in the aberrant expression of IATGs. (A) Distribution of CNVs in 33 kinds of cancer. CNV pie charts depicts the heterozygous or pure CNV of each gene within the specified cancer type. Pie charts depict the proportion of various CNV forms of a certain gene in a particular tumor, with different colors representing the various CNV kinds. (B) Correlations between the mRNA expression of IATGs and CNV levels are depicted using bubble plots. The darker the color, the greater the association. There is a positive correlation between bubble size and FDR significance. The black border edge signified FDR ≤ 0.05. (C) The relationship between CNV and IATG survival in cancer. Log-rank P values are represented by the size and color of the bubbles. The bubble size is positively connected with the importance of Log-rank P value. The bubble hue from blue to red shows the significance of Log-rank P value from low to high. The black outline suggests that the Log-rank P value is less than 0.05. CNV, copy number variants; FDR, false discovery rate; IATGs, influenza-A virus target genes.
Figure 5. Methylation status of IATGs. (A) Correlation between methylation and the expression of IATGs. Blue dots represent a negative connection, while red dots represent a positive correlation; the darker the hue, the greater the association. (B) Variations in survival between samples with hypermethylated and hypomethylated DNA of IATGs. Risk ratios and Cox P values are indicated by the size and color of the bubbles. The hue of the bubbles, from blue to red, signify low to high hazard ratios, and bubble size is strongly connected with the significance of the Cox P value. Cox P-value ≤ 0.05 are denoted by black outline borders. FDR, false discovery rate; IATGs, influenza-A virus target genes.
Figure 6. (A) Differences in IATGs’ methylation between tumor and normal samples for each type of cancer. Blue dots show decreased methylation, whereas red dots indicate increased methylation; the darker the color, the greater the methylation. (B) The numbers in each cell indicate the percentage of the gene which is associated with a specific pathway in all the cancer types stated in the study. (C) Bubble diagrams illustrate the relationship between IATGs expression and drug IC50 concentration. Blue bubbles represent unfavorable associations, while red bubbles represent favorable associations; the deeper the color, the stronger the association was. There is a positive correlation between bubble size and FDR significance. Black border outlines denote FDR ≤ 0.05. (D) Bar graphs illustrate the enrichment distribution of input IATGs in various types of cancer. The transformation of the bar from white to red signified FDR’s significance. FDR, false discovery rate; IATGs, influenza-A virus target genes.
Figure 7. Immunoassay of IATGs. (A) Relationship between immune cell infiltration and IATG Score. The significance of P values and FDR is summarized in a heat map based on the Pearman correlation analysis between input gene set GSA scores and immune cell infiltration. Blue dots represent unfavorable correlations, while red dots represent favorable correlations; the denser the color, the stronger the correlation. *P-value ≤ 0.05; #FDR ≤ 0.05. (B) IATGs and immunostimulant immune pathway correlation. R denotes the correlation, and R > 0 shows that the correlation is positive. (C) IATGs are correlated with the MHC immune pathway. R is the correlation coefficient, and R > 0 indicated that the correlation is positive. (D) IATGs are correlated with the chemokine immune pathway. R denoted the relationship between variables, and R > 0 showed that the correlation was positive. IATGs, influenza-A virus target genes.
Figure 8. IATG expression in kidney renal clear cell carcinoma (KIRC). (A) Gene transcript level in KIRC according to The Cancer Genome Atlas (TCGA) and GTEx databases; (B) qRT-PCR evaluation of IATGs in KIRC (n = 3). P < 0.05 is statistically significant. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, ns: not significant. IATGs, influenza-A virus target genes.
Figure 9. Gene-based two-dimensional map (heat map) of the correlation between related factors. Red signifies an uptrend adjustment, a positive correlation, or a positive presence. Blue represents negative correlation and down-regulation. White represents a correlation that is insignificant, nonexistent, or absent. (A) EGFR, (B) CD209, (C) CACNA1C, (D) ANXA5, (E) CLEC4M, (F) UVRAG.
Table 1. Primers
Gene Forward sequence 5'-3' Reverse sequence 5'-3' EGFR TTGCCGCAAAGTGTGTAACG GTCACCCCTAAATGCCACCG CACNA1C AATCGCCTATGGACTCCTCTT GCGCCTTCACATCAAATCCG CLEC4M GAGTAACCGCTTCTCCTGGATG CGCACAGTCTTCATTCCCGCTA ANXA5 AACCCTCTCGGCTTTATGATGC CGCTGGTAGTACCCTGAAGTG CD209 TCAAGCAGTATTGGAACAGAGGA CAGGAGGCTGCGGACTTTTT UVRAG CTTGGGTCAGCAGATTCATGC CATCGTAAGAATTGCGAACACAG S1. Cancer types and cases
Cancer type (Abbreviation) Cases Adrenocortical carcinoma (ACC) 92 Breast cancer (BRCA) 1,218 Bladder uroepithelial carcinoma (BLCA) 411 Cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) 310 Cholangiocarcinoma (CHOL) 45 Colon adenocarcinoma (COAD) 329 Lymphoid neoplasm diffuse large B-cell lymphoma (DLBC) 48 Head and neck squamous cell carcinoma (HNSC) 566 Esophageal carcinoma (ESCA) 196 Glioblastoma multiforme (GBM) 174 Kidney chromophobe (KICH) 91 Kidney renal clear cell carcinoma (KIRC) 606 Kidney renal papillary cell carcinoma (KIRP) 323 Acute myeloid leukemia (LAML) 173 Brain lower grade glioma (LGG) 534 Liver hepatocellular carcinoma (LIHC) 359 Lung adenocarcinoma (LUAD) 576 Thyroid cancer (THCA) 572 Thymoma (THYM) 122 Uterine corpus endometrial carcinoma (UCEC) 201 Uterine carcinosarcoma (UCS) 57 Uveal melanoma (UVM) 80 -
[1] Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2021; 71, 209−49. doi: 10.3322/caac.21660 [2] Smola S. Immunopathogenesis of HPV-associated cancers and prospects for immunotherapy. Viruses, 2017; 9, 254. doi: 10.3390/v9090254 [3] Müller-Coan BG, Caetano BFR, Pagano JS, et al. Cancer progression goes viral: the role of oncoviruses in aggressiveness of malignancies. Trends Cancer, 2018; 4, 485−98. doi: 10.1016/j.trecan.2018.04.006 [4] Krump NA, You JX. From merkel cell polyomavirus infection to merkel cell carcinoma oncogenesis. Front Microbiol, 2021; 12, 739695. doi: 10.3389/fmicb.2021.739695 [5] Mesri EA, Feitelson MA, Munger K. Human viral oncogenesis: a cancer hallmarks analysis. Cell Host Microbe, 2014; 15, 266−82. doi: 10.1016/j.chom.2014.02.011 [6] McLaughlin-Drubin ME, Munger K. Viruses associated with human cancer. Biochim Biophys Acta (BBA)-Mol Basis Dis, 2008; 1782, 127−50. doi: 10.1016/j.bbadis.2007.12.005 [7] Chen JD, Wang JH, Zhang JP, et al. Advances in development and application of influenza vaccines. Front Immunol, 2021; 12, 711997. doi: 10.3389/fimmu.2021.711997 [8] Taubenberger JK, Morens DM. The pathology of influenza virus infections. Annu Rev Pathol Mech Dis, 2008; 3, 499−522. doi: 10.1146/annurev.pathmechdis.3.121806.154316 [9] Taubenberger JK, Morens DM. Influenza: the once and future pandemic. Public Health Rep, 2010; 125, 16−26. doi: 10.1177/00333549101250S405 [10] Angrini M, Varthaman A, Garcia-Verdugo I, et al. To vaccinate or not: influenza virus and lung cancer progression. Trends Cancer, 2021; 7, 573−6. doi: 10.1016/j.trecan.2021.02.006 [11] Iheagwara UK, Beatty PL, Van PT, et al. Influenza virus infection elicits protective antibodies and T cells specific for host cell antigens also expressed as tumor-associated antigens: a new view of cancer immunosurveillance. Cancer Immunol Res, 2014; 2, 263−73. doi: 10.1158/2326-6066.CIR-13-0125 [12] Miri SM, Ebrahimzadeh MS, Abdolalipour E, et al. Synergy between hemagglutinin 2 (HA2) subunit of influenza fusogenic membrane glycoprotein and oncolytic Newcastle disease virus suppressed tumor growth and further enhanced by Immune checkpoint PD-1 blockade. Cancer Cell Int, 2020; 20, 380. doi: 10.1186/s12935-020-01476-5 [13] Takizawa T, Matsukawa S, Higuchi Y, et al. Induction of programmed cell death (apoptosis) by influenza virus infection in tissue culture cells. J Gen Virol, 1993; 74, 2347−55. doi: 10.1099/0022-1317-74-11-2347 [14] Chaganty BKR, Qiu SB, Lu Y, et al. Redirecting host preexisting influenza A virus immunity for cancer immunotherapy. Cancer Immunol Immunother, 2022; 71, 1611−23. doi: 10.1007/s00262-021-03099-9 [15] Eierhoff T, Hrincius ER, Rescher U, et al. The epidermal growth factor receptor (EGFR) promotes uptake of influenza A viruses (IAV) into host cells. PLoS Pathog, 2010; 6, e1001099. doi: 10.1371/journal.ppat.1001099 [16] Ueki IF, Min-Oo G, Kalinowski A, et al. Respiratory virus-induced EGFR activation suppresses IRF1-dependent interferon λ and antiviral defense in airway epithelium. J Exp Med, 2013; 210, 1929−36. doi: 10.1084/jem.20121401 [17] Wang QS, Pan WL, Wang S, et al. Protein tyrosine phosphatase SHP2 suppresses host innate immunity against influenza A virus by regulating EGFR-mediated signaling. J Virol, 2021; 95, e02001−20. [18] Ampomah PB, Kong WT, Zharkova O, et al. Annexins in influenza virus replication and pathogenesis. Front Pharmacol, 2018; 9, 1282. doi: 10.3389/fphar.2018.01282 [19] Londrigan SL, Turville SG, Tate MD, et al. N-linked glycosylation facilitates sialic acid-independent attachment and entry of influenza A viruses into cells expressing DC-SIGN or L-SIGN. J Virol, 2011; 85, 2990−3000. doi: 10.1128/JVI.01705-10 [20] Pirooz SD, He SS, Zhang T, et al. UVRAG is required for virus entry through combinatorial interaction with the class C-Vps complex and SNAREs. Proc Natl Acad Sci USA, 2014; 111, 2716−21. doi: 10.1073/pnas.1320629111 [21] Orr-Burks N, Murray J, Todd KV, et al. G-protein-coupled receptor and ion channel genes used by influenza virus for replication. J Virol, 2021; 95, e02410−20. [22] Olivier M, Asmis R, Hawkins GA, et al. The need for multi-omics biomarker signatures in precision medicine. Int J Mol Sci, 2019; 20, 4781. doi: 10.3390/ijms20194781 [23] Chen DS, Tan C, Ding PW, et al. VThunter: a database for single-cell screening of virus target cells in the animal kingdom. Nucleic Acids Res, 2022; 50, D934−42. doi: 10.1093/nar/gkab894 [24] Liao Y, Liu ZP, Ye WL, et al. Exploring the characteristics of monkeypox-related genes in pan-cancer. Cells, 2022; 11, 3909. doi: 10.3390/cells11233909 [25] Liao Y, Wang JJ, Zou JM, et al. Multi-omics analysis reveals genomic, clinical and immunological features of SARS-CoV-2 virus target genes in pan-cancer. Front Immunol, 2023; 14, 1112704. doi: 10.3389/fimmu.2023.1112704 [26] Hutter C, Zenklusen JC. The cancer genome atlas: creating lasting value beyond its data. Cell, 2018; 173, 283−5. doi: 10.1016/j.cell.2018.03.042 [27] Liu CJ, Hu FF, Xie GY, et al. GSCA: an integrated platform for gene set cancer analysis at genomic, pharmacogenomic and immunogenomic levels. Brief Bioinform, 2023; 24, bbac558. doi: 10.1093/bib/bbac558 [28] Li J, Lu YL, Akbani R, et al. TCPA: a resource for cancer functional proteomics data. Nat Methods, 2013; 10, 1046−7. [29] Yang WJ, Soares J, Greninger P, et al. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res, 2013; 41, D955−61. [30] Mermel CH, Schumacher SE, Hill B, et al. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol, 2011; 12, R41. doi: 10.1186/gb-2011-12-4-r41 [31] Schlattl A, Anders S, Waszak SM, et al. Relating CNVs to transcriptome data at fine resolution: assessment of the effect of variant size, type, and overlap with functional regions. Genome Res, 2011; 21, 2004−13. doi: 10.1101/gr.122614.111 [32] Chen MJM, Li J, Wang YM, et al. TCPA v3.0: an integrative platform to explore the pan-cancer analysis of functional proteomic data. Mol Cell Proteomics, 2019; 18, S15−25. [33] Akbani R, Ng PK, Werner HMJ, et al. A pan-cancer proteomic perspective on The Cancer Genome Atlas. Nat Commun, 2014; 5, 3887. doi: 10.1038/ncomms4887 [34] Subramanian A, Tamayo P, Mootha VK, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA, 2005; 102, 15545−50. doi: 10.1073/pnas.0506580102 [35] Miao YR, Zhang Q, Lei Q, et al. ImmuCellAI: a unique method for comprehensive T-cell subsets abundance prediction and its application in cancer immunotherapy. Adv Sci, 2020; 7, 1902880. doi: 10.1002/advs.201902880 [36] Ru BB, Wong CN, Tong Y, et al. TISIDB: an integrated repository portal for tumor-immune system interactions. Bioinformatics, 2019; 35, 4200−2. doi: 10.1093/bioinformatics/btz210 [37] Tang ZF, Kang BX, Li CW, et al. GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res, 2019; 47, W556−60. doi: 10.1093/nar/gkz430 [38] GTEx project maps wide range of normal human genetic variation: a unique catalog and follow-up effort associate variation with gene expression across dozens of body tissues. Am J Med Genet A, 2018; 176, 263-4. [39] Martincorena I, Campbell PJ. Somatic mutation in cancer and normal cells. Science, 2015; 349, 1483−9. doi: 10.1126/science.aab4082 [40] Pleasance ED, Cheetham RK, Stephens PJ, et al. A comprehensive catalogue of somatic mutations from a human cancer genome. Nature, 2010; 463, 191−6. doi: 10.1038/nature08658 [41] Hastings PJ, Lupski JR, Rosenberg SM, et al. Mechanisms of change in gene copy number. Nat Rev Genet, 2009; 10, 551−64. [42] Zhao SG, Chen WS, Li HL, et al. The DNA methylation landscape of advanced prostate cancer. Nat Genet, 2020; 52, 778−89. doi: 10.1038/s41588-020-0648-8 [43] Dor Y, Cedar H. Principles of DNA methylation and their implications for biology and medicine. Lancet, 2018; 392, 777−86. doi: 10.1016/S0140-6736(18)31268-6 [44] Baumeister SH, Freeman GJ, Dranoff G, et al. Coinhibitory pathways in immunotherapy for cancer. Annu Rev Immunol, 2016; 34, 539−73. doi: 10.1146/annurev-immunol-032414-112049 [45] Zhang YY, Zhang ZM. The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol, 2020; 17, 807−21. doi: 10.1038/s41423-020-0488-6 [46] Wang LH, Chard Dunmall LS, Cheng ZG, et al. Remodeling the tumor microenvironment by oncolytic viruses: beyond oncolysis of tumor cells for cancer treatment. J Immunother Cancer, 2022; 10, e004167. doi: 10.1136/jitc-2021-004167 [47] de Oliveira DE, Ballon G, Cesarman E. NF-κB signaling modulation by EBV and KSHV. Trends Microbiol, 2010; 18, 248−57. doi: 10.1016/j.tim.2010.04.001 [48] Biggi AFB, de Oliveira DE. The epstein-barr virus hacks immune checkpoints: evidence and consequences for lymphoproliferative disorders and cancers. Biomolecules, 2022; 12, 397. doi: 10.3390/biom12030397 -
23352+Supplementary Materials.pdf