Abstract
Osteosarcoma is a common malignancy seen mainly in children and adolescents. The disease is characterized by poor overall prognosis and lower survival due to a lack of predictive markers. Many gene signatures with diagnostic, prognostic, and predictive values were evaluated to achieve better clinical outcomes. Two public data series, GSE21257 and UCSC Xena, were used to identify the minimum number of robust genes needed for a predictive signature to guide prognosis of patients with osteosarcoma. The lasso regression algorithm was used to analyze sequencing data from TCGA-TARGET, and methods such as Cox regression analysis, risk factor scoring, receiving operating curve, KMplot prognosis analysis, and nomogram were used to characterize the prognostic predictive power of the identified genes. Their utility was assessed using the GEO osteosarcoma dataset. Finally, the functional enrichment analysis of the identified genes was performed. A total of twenty-gene signatures were found to have a good prognostic value for predicting patient survival. Gene ontology analysis showed that the key genes related to osteosarcoma were categorized as peptide–antigen binding, clathrin-coated endocytic vesicle membrane, peptide binding, and MHC class II protein complex. The osteosarcoma related genes in these modules were significantly enriched in the processes of antigen processing and presentation, phagocytosis, cell adhesion molecules, Staphylococcus aureus infection. Twenty gene signatures were identified related to osteosarcoma, which would be helpful for predicting prognosis of patients with OS. Further, these signatures can be used to determine the subtypes of osteosarcoma.
Keywords: Osteosarcoma, Gene signature, Prognosis, Survival rate, Coexpression analysis
Introduction
Osteosarcoma (OS) is a common primary malignancy seen mainly in children and adolescents (Yang et al., 2018). Its annual incidence ranges from two to three in 1 million people (De Azevedo et al., 2020), and it accounts for 40.51% of primary bone malignancies (Li et al., 2020). The incidence of OS is higher in males than in females (Damron, Ward & Stewart, 2007), and it accounts for 15% of all extracranial tumors in the 10–19-year-old age group) (Nie & Peng, 2018). It is considered the third most common cancer in adolescence (Zhang, Lan & Lin, 2018). OS treatment is aggressive and combines neoadjuvant chemotherapy, extensive surgical resection, and additional postoperative adjuvant chemotherapy. The five-year survival rate of non-metastatic patients is 65%–70% (Siegel, Miller & Jemal, 2018). Regarding patients with distant metastases, the 5-year survival rate is only 15%–30% (Whelan & Davis, 2018). Further, relapse rate remains high at approximately 35% (Yu et al., 2014). Metastasis remains the main cause of death in patients with OS. Many prognostic factors are reported to be related to OS: miR-195, miR-21, TGF-β, MMP-9, HIF-1, APE1, and COX2. However, use of these factors has not improved survival rate related to OS (Zamborsky et al., 2019). Therefore, clarifying the molecular mechanisms underlying the occurrence and progression of OS and exploring the potential diagnostic and therapeutic targets are of great importance for the diagnosis and treatment of OS. Many biomarkers have been evaluated (Table 1) (Wan-Ibrahim et al., 2015), but none have been approved by the Food and Drug Administration for clinical settings (Wan-Ibrahim et al., 2015). This lack may be due to research deficiencies, unacceptable heterogeneity, or absence of effective evaluation. Biomarkers for patients with OS, particularly those with metastases, are urgently needed for early diagnosis and establishment of treatment goals. Serum biomarkers are used for predicting prognosis of other cancers but are rarely characterized in OS (Zamborsky et al., 2019). An obvious need in OS is effective biomarkers for characterizing disease progression and associated prognosis.
Table 1. Potential biomarkers for osteosarcoma.
Study approach | Type of biomarker | Candidate biomarker | Regulation | Sample | Technique | Reference |
---|---|---|---|---|---|---|
Genomics | Diagnostic | C7orf24 | ↑ | Tissue; cell lines | RT-PCR; qRT-PCR; Western blot (WB); siRNA transfection | Uejima et al. (2011) |
Diagnostic And Predictive | miRNA signatures | − | Tissue; cell lines | qRT-PCR | Gougelet et al. (2011) | |
Predictive | Multigene classifier | − | Tissue | cDNA microarray; qRT-PCR | Man et al. (2005) | |
Predictive | Multigene classifier | − | Tissue | cDNA microarray; RT-PC | Mintz et al. (2005) | |
Predictive And Prognosis | miRNA signatures | − | Tissue | miRNA microarray; qRT-PCR; immunohistochemistry (IHC) | Jones et al. (2012) | |
Predictive And Prognosis | miR-21 | ↑ | Serum | qRT-PCR | Yuan et al. (2012) | |
Prognosis | Tenascin-C | ↓ | Cell lines | cDNA microarray; RT-PCR; WB; IHC | Xiong et al. (2009) | |
Prognostic | miRNA-214 | ↑ | Tissue | qRT-PCR | Wang et al. (2014) | |
Proteomics | Diagnostic | Serum amyloid A (SAA) | ↑ | Plasma | SELDI-TOF MS; WB | Li et al. (2010) |
Diagnostic | Ezrin (EZR); a crystallin β chain (CRYAB) | ↑ | Tissue | 2D-DIGE; LC-ESI-MS/MS; RT-PCR for mRNA; tissue microarray and IHC | Folio et al. (2009) | |
Diagnostic | Cytochrome C1 (CYC-1) | ↑ | Serum; cell lines | SELDI-TOF MS; Gene microarray (cell lines) | Li et al. (2009) | |
Diagnostic | Zinc finger protein 133 (ZNF 133); tubulin-a1c (TUBA1C) | ↑ | Tissue | 2-DE; MALDI-TOF MS; WB; IHC | Li et al. (2010) | |
Diagnostic | Protein NDRG 1 | ↑ | Plasma membrane from cell line and tissue | 2-DE; LC-ESI-MS/MS; IHC; WB | Hua et al. (2011) | |
Diagnostic | Gelsolin | ↓ | Serum | 2D-DIGE; MALDIHTOF; WB; ELISA | Jin et al. (2012) | |
Predictive And Prognostic | SAA | ↑ | Serum | 2D-DIGE; MALDI-TOF MS; WB; ELISA | Jin et al. (2007) | |
Predictive | Peroxiredoxin 2 (PRDX2) | ↑(poor prognosis) | Tissue | 2D-DIGE; LC-ESI-MS/MS; WB | Kikuta et al. (2010) | |
Predictive | SAA; transthyretin (TTR) | ↑(poor prognosis) | Plasma | SELDI-TOF MS; WB | Li et al. (2011) |
One study reported that CDC20 and its downstream substrates, secure, cyclin A2 and cyclin B2 are good prognostic factors for OS (Wu et al., 2019). Savage et al. (2013) suggested that two loci in the GRM4 gene at 6p21.3 and in the gene, desert, at 2p25.2, These two loci warrant further exploration to uncover the biological mechanisms underlying susceptibility to osteosarcoma.The study addressed a single gene and did not take into account interactions among molecules that regulate tumorigenesis. Three candidate genes (ALOX5AP, CD74 and FCGR2A) were found. Their expression levels in lung and lymph nodes were higher than levels in matched cancer tissues, and they may be expressed in microenvironments (Li et al., 2020). Some limitations exist in these studies. First, accuracy cannot be guaranteed with only one dataset because of an expected high false-positive rate. Further, using a single high-throughput analysis method (only sequencing or chip data), results obtained will be biased. Second, a patient’s sample data are too limited. Finally, clinical information is incomplete.
Identifying the minimum number of robust genes needed to produce a predictive signature for prognosis for patients with OS was the objective of this study. The lasso regression algorithm was used to analyze sequencing data from TCGA-TARGET, and Cox regression analysis, risk factor score, receiving operating curve (ROC), KMplot prognosis analysis, nomogram and other methods were used to assess genes for their predictive power. Next, the accuracy and predictive power of twenty-gene identified in this process were assessed using the GEO OS dataset. Finally, we performed functional enrichment analysis on these twenty-gene.
Material and Methods
Data collection and preprocessing
Training set: The TARGET-OS RNA-sequencing dataset (presented as fragments per kilobase million, FPKM), corresponding clinical characteristics and prognosis information were downloaded from UCSC Xena (Goldman et al., 2019) (https://xena.ucsc.edu/). Patients with expression profiles but no prognostic information and clinical characteristics were excluded. Finally, 84 patients with OS were included in a training set. FPKM data were converted to TPM data and annotated using gencode.v22.annotation.gene.probeMap.
Validation set: The gene expression data GSE21257 (Buddingh et al., 2011) (GPL570 (HG-U133_Plus_2) Affymetrix Human Genome U133 Plus 2.0 Array) for 53 patients with OS were downloaded from the GEO database, and corrected and annotated with R software.
Construction of gene signatures
A linear regression multiple regression model was developed for the underlying expression levels of genes for prognostic risk scores. The method chosen by lasso Cox was 10-fold cross-validation. According to the median cutoff value (the cutoff value refers to the content before the brackets of the HR value in each dataset) of the risk score, patients with OS were divided into high-risk and low-risk groups. Model prediction efficiency using the training set was evaluated by Kaplan–Meier log-rank test, time-dependent ROC curve analysis, Cox regression analysis, and risk factor score for validation and test sets. A nomogram was constructed using Iasso’s guidelines.
Weighted correlation network analysis of genes
Based on the variance of gene expression in TARGET-OS data, the top 5000 genes were selected for WGCNA (Langfelder & Horvath, 2008). This analysis proceeded as: check for outliers in all samples, construct sample tree with hclust, and remove outliers according to cut height. To explore the correlation between expression data and clinical phenotypes, the sample tree and characteristic heat map were visualized. Subsequently, the strength of associations between pairs of nodes of the adjacency matrix aij was calculated as: sij = |cor (xi, xj)| aij = Sijβ. xi and xj are genes i and j. The vector of expression value, sij, indicates the strength of Pearson’s correlation coefficient between genes i and j. The aij coding network connects genes i and j. β value is a soft threshold (power value). Further, the Scale-Free Topology Fit Index (scale-free R2) range from 0 to 1 is used to determine the scale-free topological model. Selecting a set of soft threshold powers (range: 1 to 20) assists in calculating scale-free topological model fitting. The soft threshold of β = 7 was used to define the adjacency matrix. The corresponding scale-free R2 value is 0.87, suggesting a satisfactory scale-free topology model.
In coexpression networks, the highest absolute association genes were clustered into the same module to generate a clustering dendrogram. Relationships between clinical traits and risk scores were analyzed by Pearson correlation and results were visualized by heat map analysis. Genes in the module were analyzed for gene ontology (GO) and KEGG pathway enrichment. Moreover, Cytoscape (Smoot et al., 2010) (version 3.7.2) was used to visualize the weighted coexpression network. gene ontology (GO) analysis, which includes annotation of biological processes (BPs), molecular functions (MFs), and cellular components (CCs), serves as a major bioinformatics tool to annotate genes and analyze the biological processes of these genes. Huimei Wang et al. analyzed the biological classification of DEGs, showing that changes in BPs of DEGs were significantly enriched in positive regulation of associated cell response, by GO analysis (Wang et al., 2018).
Functional enrichment analysis
Clusterprofiler (Yu et al., 2012) was used to study modules related to biological function for determining functional and pathway enrichment. A multiple testing correction was performed using hypergeometric test functions and the Benjamini–Hochberg method. The GOplot (Walter, Sánchez-Cabo & Ricote, 2015) package was used to visualize the enrichment analysis.
Statistical analysis
Statistical analysis was conducted in R software (version 3.6.1) with the following packages: “glmnet” (Friedman, Hastie & Tibshirani, 2009), “survivalROC” (Heagerty, Lumley & Pepe, 2000), “WGCNA,” and “clusterProfiler.” All the statistical tests were two sided, and P-values of < 0.05 were considered statistically significant.
Results
Construction of genes classifier for OS
Prognostically significant genes for OS in TARGET-OS data were analyzed and a total of 1151 genes were significantly associated with poor prognosis. These 1151 candidate genes were included in the lasso prognostic classifier for further screening and model construction. A twenty-gene classifier for the OS was developed (Figs. 1A and 1B). The gene information in the model is shown in Table 1. Patients were divided into a high-risk group (n = 42) and low-risk group (n = 42) based on risk scores. The median risk score was set as the cutoff (the cutoff value refers to the content before the brackets of the HR value in each dataset). Table 2 shows clinical characteristics of patients with OS in the training set according to their high risk and low risk scores. The Kaplan–Meier log-rank test suggested a significant difference between high-risk and low-risk groups in the training set (P < 0.001; Fig. 1C). In the time-dependent analysis of the ROC curve, AUCs for OS in the first, third, and fifth years were 0.94, 0.98, and 0.97, respectively (Fig. 1D).
Table 2. Twenty genes significantly related to the overall survival in the training set underlying the LASSO model.
Ensemble ID | Gene | Chromosome location | Coefficient of lasso model | HR | p value |
---|---|---|---|---|---|
ENSG00000103274.9 | NUBP1 | chr16:10743786-10769351:(+) | −0.2216 | 0.905 (0.86, 0.953) | 0.000 |
ENSG00000163219.10 | ARHGAP25 | chr2:68679601-68826833:(+) | −0.0503 | 0.795 (0.685, 0.924) | 0.003 |
ENSG00000077420.14 | APBB1IP | chr10:26438203-26567803:(+) | −0.0429 | 0.924 (0.88, 0.97) | 0.001 |
ENSG00000162517.11 | PEF1 | chr1:31629862-31644896:(-) | −0.0230 | 0.979 (0.966, 0.992) | 0.002 |
ENSG00000102226.8 | USP11 | chrX:47232690-47248328:(+) | −0.0123 | 0.982 (0.97, 0.994) | 0.004 |
ENSG00000179163.11 | FUCA1 | chr1:23845077-23868294:(-) | −0.0104 | 0.969 (0.949, 0.99) | 0.003 |
ENSG00000189171.12 | S100A13 | chr1:153618787-153634092:(-) | 0.0019 | 1.007 (1.004, 1.011) | 0.000 |
ENSG00000132535.17 | DLG4 | chr17:7189890-7219702:(-) | 0.0088 | 1.064 (1.021, 1.109) | 0.003 |
ENSG00000158315.9 | RHBDL2 | chr1:38885807-38941799:(-) | 0.0144 | 1.01 (1.006, 1.013) | 0.000 |
ENSG00000176171.10 | BNIP3 | chr10:131966455-131981931:(-) | 0.0170 | 1.009 (1.004, 1.014) | 0.000 |
ENSG00000167549.17 | CORO6 | chr17:29614756-29622907:(-) | 0.0425 | 1.054 (1.021, 1.088) | 0.001 |
ENSG00000125337.15 | KIF25 | chr6:167996241-168045089:(+) | 0.0570 | 1.148 (1.076, 1.224) | 0.000 |
ENSG00000179262.8 | RAD23A | chr19:12945855-12953642:(+) | 0.0741 | 1.005 (1.002, 1.008) | 0.000 |
ENSG00000147378.10 | FATE1 | chrX:151716035-151723194:(+) | 0.0862 | 2.217 (1.518, 3.238) | 0.000 |
ENSG00000113739.9 | STC2 | chr5:173314713-173329503:(-) | 0.0895 | 1.016 (1.005, 1.027) | 0.004 |
ENSG00000197467.12 | COL13A1 | chr10:69801931-69964275:(+) | 0.0910 | 1.021 (1.013, 1.029) | 0.000 |
ENSG00000017483.13 | SLC38A5 | chrX:48458537-48470256:(-) | 0.1003 | 1.008 (1.003, 1.014) | 0.002 |
ENSG00000136997.13 | MYC | chr8:127735434-127741434:(+) | 0.1635 | 1.003 (1.002, 1.005) | 0.000 |
ENSG00000138028.13 | CGREF1 | chr2:27098889-27119115:(-) | 0.2105 | 1.015 (1.009, 1.021) | 0.000 |
ENSG00000241563.3 | CORT | chr1:10449719-10451902:(+) | 0.2504 | 1.026 (1.016, 1.036) | 0.000 |
Notes.
- HR
- Hazard Ratio
- CI
- Confidence Interval
Identification of a prognostic risk score model based on the training set
Based on univariate regression analysis (Cox’s proportional hazard model), forest plots of selected genes with p-value and hazard ratios are shown in Fig. 2A. HR, Hazard Rate in Fig. 2A, shows that the p-value of selected genes was less than 0.05, showing the statistical difference, which cannot be ignored. Only when 95% CI conclude 1, it could be proved that these selected genes signatures were not significantly associated with the prognosis of patients with OS. Although the extremum of HR is very close to 1, they show that these selected genes might be associated with the prognosis of patients with OS. Risk scores and survival status of patients with OS are shown in Figs. 2B and 2C, respectively. These results indicate that patients with high risk scores have poor outcomes compared with patients with low risk scores (Fig. 2D). Using these data, a nomogram combining the classifier with clinicopathological features to predict the survival probability of patients with different risk scores was prepared (Fig. 3A). The calibration chart demonstrated that predicted three-year and five-year survival rates were very close to observed ratios (Fig. 3B).
Validation of twenty-gene signature for survival prediction in the validation set
Subsequently, the validation set was used to assess the power of the twenty-gene signature in predicting prognosis. OS of the low-risk group was superior to the high-risk group (P < 0.05; Fig. 4A). Also, time-dependent ROC analysis is used to assess the effectiveness of risk models in predicting outcomes. Areas under ROC was 0.81, 0.78, and 0.76 at one, three and five years, respectively, suggesting that the twenty-gene signature displays good predictive power (Fig. 4B). The C-index value is 0.944. The risk score (Fig. 4C) and survival status (Fig. 4D) of patients with OS, and distribution of risk scores of twenty-gene expression profiles are shown in a heat map of 53 patients in the validation set (Fig. 4E). These results suggest that the twenty-gene signature shows good prognostic value for patient survival. This finding is further validated since patients with high risk scores were associated with poorer prognoses compared with patients with low risk scores. Previous studies have investigated gene factors in an attempt to identify new prognostic OS markers (Goh et al., 2019; Guan, Guan & Song, 2020). Hence, I have compared the prognostic gene set for osteosarcoma, Cox univariate and multivariate analysis showed that BACE2, ING2 ALOX5AP, HLA-DMB, HLA-DRA, and SPINT2 were not the independent prognostic factors for osteosarcoma (Table 3). Table 3 shows the determinants of BACE2, ING2 ALOX5AP, HLA-DMB, HLA-DRA, and SPINT2 for all the patients. In the univariate analysis, These gene signatures exhibited a negative correlation with age, gender, specific and primary tumor site, but illustrate a positive correlation with recurrence and risk score. After multiple stepwise analysis, these biomarkers correlated independently with Recurrence (hazard ratio (HR) 5.374, 95% CI [1.183–24.404], p = 0.029) and risk score (HR 9.869, 95% CI [4.663–20.887], p < 0.001).
Table 3. Univariate and multivariate Cox regression analyses genes signiture for patients with osteosarcoma in study cohort.
Univariate | Multivariate | |||
---|---|---|---|---|
Characteristics | HR | p.value | HR | p.value |
Gender (Female vs Male) | 0.666 (0.317, 1.399) | 0.283 | ||
Age (<18 vs ≥18) | 0.909 (0.345, 2.393) | 0.846 | ||
Recurrence (No vs Yes) | 19.348 (4.589, 81.563) | <0.001 | 5.374 (1.183, 24.404) | 0.029 |
Primary tumor site (Arm/Hand OR Pelvis vs Leg/Foot) | 0.487 (0.168, 1.413) | 0.185 | ||
Specific tumor site (Femur vs Tibia or others) | 0.642 (0.309, 1.337) | 0.236 | ||
riskscore | 13.915 (7.067, 27.398) | <0.001 | 9.869 (4.663, 20.887) | <0.001 |
Gene co-expression network analysis
The relationship between risk scores and gene expression profiles was evaluated using the top 5000 genes in the variance filter for WGCNA analysis. The red line (cut height = 8000) was used to remove the abnormal samples in the sample tree. TARGET-40-PAUXOZ-01A was excluded after removing outliers (Fig. 5). The sample dendrogram and trait heat map placed selected samples into different sample clusters that provided clinical trait information (Fig. 6A). Independence and average connectivity of coexpression modules were determined by power (β) and scale R2 value. A series of soft thresholds and corresponding performance power were plotted. The threshold for scale R2 value was set at 0.85. The power value of seven is the threshold that first reaches the scale R2 value, and was chosen as the soft threshold to construct and identify coexpression modules (Figs. 6B and 6C). The expression matrix was converted to an adjacency matrix, and subsequently into a topology matrix. Based on TOM, genes are clustered based on criteria of mixed dynamic shear trees using average connection–level distance. The minimum number of genes in each module was set at 7. Finally, a total of 11 modules were identified with the WGCNA package (Fig. 6D). Statistics of gene numbers in each module are shown in Table 4. Associations of these modules with clinical characteristics (including sex, age, relapse, OS, metastasis, major cancer sites, specific cancer sites, and risk scores) are shown in Fig. 6E. T module showed the highest correlation with the risk score. Further, the magenta module also was negatively correlated with sex and OS and positively correlated with major cancer sites. GSEA analysis between high-risk and low-risk groups can better illustrate the risk-score related to the biological process. But GSEA analysis between high-risk and low-risk groups only considers the expression level of gene sets. Analysis of WGCNA is based on the clinical relevant factors, which makes it sense, although its risk-score is low.
Table 4. Relationship between risk score of the 20-marker-based prognostic classifier with os and clinical characteristics in the training set.
Characters | Level | Low risk | High risk | P value |
---|---|---|---|---|
n | 42 | 42 | ||
Gender | Female (%) | 16 ( 38.1) | 21 (50.0) | 0.379 |
Male (%) | 26 ( 61.9) | 21 (50.0) | ||
Age | <18 (%) | 32 ( 76.2) | 34 (81.0) | 0.791 |
≥18 (%) | 10 ( 23.8) | 8 (19.0) | ||
Overall survival | Alived (%) | 42 (100.0) | 13 (31.0) | <0.001 |
Dead (%) | 0 ( 0.0) | 29 (69.0) | ||
Recurrence | No (%) | 32 ( 76.2) | 13 (31.0) | <0.001 |
Yes (%) | 10 ( 23.8) | 29 (69.0) | ||
Metastasis | No (%) | 37 ( 88.1) | 26 (61.9) | 0.011 |
Yes (%) | 5 ( 11.9) | 16 (38.1) | ||
Primary tumor site | Arm/Hand OR Pelvis (%) | 3 ( 7.1) | 5 (11.9) | 0.713 |
Leg/Foot (%) | 39 ( 92.9) | 37 (88.1) | ||
Specific tumor site | Femur (%) | 13 ( 31.0) | 25 (59.5) | 0.015 |
Tibia or others (%) | 29 ( 69.0) | 17 (40.5) |
Functional enrichment analysis in the gene coexpression network of the magenta module
GO and KEGG enrichment analysis was used to characterize biological functions of genes in the magenta module as they related to risk scores. GO analysis showed that key genes related to OS were mainly enriched in peptide–antigen binding, clathrin-coated endocytic vesicle membrane, peptide binding, and MHC class II protein complex (Fig. 7A). KEGG analysis showed that the key genes related to OS were mainly enriched in antigen processing and presentation, phagosome, cell adhesion molecules, Staphylococcus aureus infection (Fig. 7B). Subsequently, the interaction network for enrichment pathways in the magenta module was visualized (Fig. 7C). Moreover, an interaction network was constructed to visualize genes in the coexpression magenta module (Fig. 7D).
Discussion
OS is a disease involving complex interactions among many factors. Overall, OS disrupts cell signaling pathways, causing loss of bone tissue homeostasis (Otoukesh et al., 2018). The urgent need to obtain better clinical results highlights the related need for better diagnostic, prognostic, and predictive biomarkers (Ludwig & Weinstein, 2005). Presently, no specific markers are available for OS diagnosis. To reduce mortality and increase limb salvage, biomarkers are needed for early identification of disease (Smida et al., 2017). Possible genetic biomarkers to address this need were identified in the present study. Lasso regression screened in twenty-gene in a training set. These genes have certain prognostic value in time ROC, risk factor, and Kaplan–Meier plots. Results were verified with a validation set.
One current study showed that inhibition of BNIP3 expression by baicalein treatment could inhibit cell apoptosis (Ye et al., 2015). Moreover, the MYC gene is also reported to be amplified in a subset of OS (Ladanyi et al., 1993). MYC promotes the proliferation of OS cells through the autophagy pathway (Mo et al., 2019). Cross-species genomics identified DLG2 as a tumor suppressor in OS (Shao et al., 2019). Further, depletion of KIF25 leads to the formation of actin stress fibers, which may be due to the changes of Rho signaling observed before microtubule destabilization (Wittmann & Waterman-Storer, 2001). In addition to the above studies of genes related to OS, relationships between other genes screened in this study and malignancies have been reported in the literature but have not been reported in OS. A lack of related research reports to help assess the potential of these genes as targets for the treatment and diagnosis of OS currently exits.
The current study also explored the relationship between risk scores and gene expression profiles using WGCNA analysis. Important genes related to OS were enriched in 11 different modules. Results showed that pathways related to inflammation and immunity were primarily enriched in the turquoise module. The gray and turquoise modules share the most pathways among all pairwise comparisons. Genes in these modules may play similar roles in OS.
LASSO is a method of shrinking and variable selection linear regression model. The purpose of LASSO regression is to obtain a subset of the predictors to minimize the prediction errors of the quantitative response variables. The lasso does this by constraining the model parameters, making the regression coefficients of some variables approach 0. “Coxnet” fits a lasso penalty, and its adaptive forms, such as adaptive lasso. Moreover, it treats the number of non-zero coefficients as another tuning parameter and simultaneously selects with the regularization parameter “lambda”. And Xiong et al. (2020) established a 13 gene-based survival score for prognostic prediction of Lung adenocarcinoma. They filtrated the relevant gene by LASSO and identified the wide applicability of these genes.
Previous studies report that CXCR3 may be an independent prognostic risk factor, suggesting a possible benefit of immunotherapy for OS (Tang et al., 2019). The susceptibility and severity of OS may also be related to functional polymorphism of inflammatory genes (Oliveira et al., 2007). Another study has shown that increased expression of MIF indicates an increased risk of metastasis, and MIF is related to angiogenesis and cell infiltration of OS. MIF can be used as a prognostic marker of OS and a potential therapeutic target (Kim et al., 2008). Therefore, clarifying molecular mechanisms of OS may facilitate the identification of novel therapeutic and prognostic targets. Limitations to the study exist. First, although these twenty-gene were identified to have certain prognostic value for OS, all data analyzed in the study were retrieved from the online databases. Thus, further experimental evidence, such as real-time PCR, western blot, immunohistochemistry assays, is required to fully elucidate the role of 20-gene signatures. Second, to determine the diagnostic accuracy of gene associations, a larger sample size would be useful for additional internal validation. Third, assessment of all clinically relevant influencing factors for OS was not included, and more clinical information and PFS-related data are needed. Finally, differentially expressed genes were not evaluated and nor was the need to add a normal control group and joint verification of multiple tumor sites.In conclusion, the current study proposes a 20-gene signature for diagnostic and prognostic purposes for OS. The twenty-gene signature is independently related to prognostic parameters of OS classification. Also, the signature is a good classifier for different subtypes of patients with OS. This signature may provide a new perspective on the prognosis of OS. The biological functions and pathways enriched in specific modules will be beneficial to the development of new therapeutic methods for the treatment of OS.
Supplemental Information
Funding Statement
The authors received no funding for this work.
Additional Information and Declarations
Competing Interests
The authors declare there are no competing interests.
Author Contributions
Zhongpeng Qiu and Gang Li conceived and designed the experiments, performed the experiments, analyzed the data, authored or reviewed drafts of the paper, and approved the final draft.
Xinhui Du and Kai Chen conceived and designed the experiments, performed the experiments, analyzed the data, prepared figures and/or tables, and approved the final draft.
Yi Dai and Sibo Wang analyzed the data, prepared figures and/or tables, and approved the final draft.
Jun Xiao conceived and designed the experiments, authored or reviewed drafts of the paper, and approved the final draft.
Data Availability
The following information was supplied regarding data availability:
The raw measurements are available in the Supplemental Files.
References
- Buddingh et al. (2011).Buddingh EP, Kuijjer ML, Duim RAJ, Bürger H, Agelopoulos K, Myklebost O, Serra M, Mertens F, Hogendoorn PCW, Lankester AC, Cleton-Jansen A. Tumor-infiltrating macrophages are associated with metastasis suppression in high-grade osteosarcoma: a rationale for treatment with macrophage activating agents. Clinical Cancer Research. 2011;17:2011–2019. doi: 10.1158/1078-0432.CCR-10-2047. [DOI] [PubMed] [Google Scholar]
- Damron, Ward & Stewart (2007).Damron TA, Ward WG, Stewart A. Osteosarcoma, Chondrosarcoma, and Ewing’s sarcoma: national cancer data base report. Clinical Orthopaedics and Related Research. 2007;459:40–47. doi: 10.1097/BLO.0b013e318059b8c9. [DOI] [PubMed] [Google Scholar]
- De Azevedo et al. (2020).De Azevedo JWV, Fernandes TAAD, Fernandes JV, De Azevedo JCV, Lanza DCF, Bezerra CM, Andrade VS, De Araújo JMG, Fernandes JV. Biology and pathogenesis of human osteosarcoma (Review) Oncology Letters. 2020;19:1099–1116. doi: 10.3892/ol.2019.11229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Friedman, Hastie & Tibshirani (2010).Friedman J, Hastie T, Tibshirani R. Regularization Paths for Generalized Linear Models via Coordinate Descent. http://www.jstatsoft.org/v33/i01/ Journal of Statistical Software. 2010;33(1):1–22. [PMC free article] [PubMed] [Google Scholar]
- Folio et al. (2009).Folio C, Mora MI, Zalacain M, Corrales FJ, Segura V, Sierrasesúmaga L, Toledo G, San-Julián M, Patiño-García A. Proteomic analysis of chemonaive pediatric osteosarcomas and corresponding normal bone reveals multiple altered molecular targets. Journal of Proteome Research. 2009;8:3882–3888. doi: 10.1021/pr900113w. [DOI] [PubMed] [Google Scholar]
- Goh et al. (2019).Goh TS, Lee JS, IlKim J, Park YG, Pak K, Jeong DC, Oh S-O, Kim YH. Prognostic scoring system for osteosarcoma using network-regularized high-dimensional Cox-regression analysis and potential therapeutic targets. Journal of Cellular Physiology. 2019;234:13851–13857. doi: 10.1002/jcp.28065. [DOI] [PubMed] [Google Scholar]
- Goldman et al. (2019).Goldman M, Craft B, Hastie M, Repečka K, Mc Dade F, Kamath A, Banerjee A, Luo Y, Rogers D, Brooks AN, Zhu J, Haussler D. The UCSC Xena platform for public and private cancer genomics data visualization and interpretation. bioRxiv. 2019:326470. doi: 10.1101/326470. [DOI]
- Gougelet et al. (2011).Gougelet A, Pissaloux D, Besse A, Perez J, Duc A, Dutour A, Blay JY, Alberti L. Micro-RNA profiles in osteosarcoma as a predictive tool for ifosfamide response. International Journal of Cancer. 2011;129:680–690. doi: 10.1002/ijc.25715. [DOI] [PubMed] [Google Scholar]
- Guan, Guan & Song (2020).Guan X, Guan Z, Song C. Expression profile analysis identifies key genes as prognostic markers for metastasis of osteosarcoma. Cancer Cell International. 2020;20:104. doi: 10.1186/s12935-020-01179-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heagerty, Lumley & Pepe (2000).Heagerty PJ, Lumley T, Pepe MS. Time-dependent ROC curves for censored survival data and a diagnostic marker. Biometrics. 2000;56:337–344. doi: 10.1111/j.0006-341X.2000.00337.x. [DOI] [PubMed] [Google Scholar]
- Hua et al. (2011).Hua Y, Jia X, Sun M, Zheng L, Yin L, Zhang L, Cai Z. Plasma membrane proteomic analysis of human osteosarcoma and osteoblastic cells: revealing NDRG1 as a marker for osteosarcoma. Tumor Biology. 2011;32:1013–1021. doi: 10.1007/s13277-011-0203-4. [DOI] [PubMed] [Google Scholar]
- Jin et al. (2007).Jin S, Shen J, Guo Q, Zhou J, Wang J, Huang G, Zou C, Yin J, Liu S, Liu W, Li M, Wang L. 2-D DIGE and MALDI-TOF-MS analysis of the serum proteome in human osteosarcoma. Proteomics –Clinical Applications. 2007;1:272–285. doi: 10.1002/prca.200600869. [DOI] [PubMed] [Google Scholar]
- Jin et al. (2012).Jin S, Shen JN, Peng JQ, Wang J, Huang G, Li MT. Increased expression of serum gelsolin in patients with osteosarcoma. Chinese Medical Journal. 2012;125:262–269. [PubMed] [Google Scholar]
- Jones et al. (2012).Jones KB, Salah Z, Del Mare S, Galasso M, Gaudio E, Nuovo GJ, Lovat F, LeBlanc K, Palatini J, Randall RL, Volinia S, Stein GS, Croce CM, Lian JB, Aqeilan RI. miRNA signatures associate with pathogenesis and progression of osteosarcoma. Cancer Research. 2012;72:1865–1877. doi: 10.1002/prca.200900172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kikuta et al. (2010).Kikuta K, Tochigi N, Saito S, Shimoda T, Morioka H, Toyama Y, Hosono A, Suehara Y, Beppu Y, Kawai A, Hirohashi S, Kondo T. Peroxiredoxin 2 as a chemotherapy responsiveness biomarker candidate in osteosarcoma revealed by proteomics. Proteomics Clinical Applications. 2010;4:560–567. doi: 10.1002/prca.200900172. [DOI] [PubMed] [Google Scholar]
- Kim et al. (2008).Kim H, Han I, Lee M, Oh JH, Kim EH. Expression of macrophage migration inhibitory factor (MIF) is related to prognosis in osteosarcoma. Cancer Research. 2008;68:2107–2113. doi: 10.1007/s11999-008-0333-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ladanyi et al. (1993).Ladanyi M, Park CK, Lewis R, Jhanwar SC, Healey JH, Huvos AG. Sporadic amplification of the MYC gene in human osteosarcomas. Diagnostic Molecular Pathology: the American Journal of Surgical Pathology, Part B. 1993;2:163–167. doi: 10.1097/00019606-199309000-00004. [DOI] [PubMed] [Google Scholar]
- Langfelder & Horvath (2008).Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics. 2008;9:559. doi: 10.1186/1471-2105-9-559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li et al. (2011).Li Y, Dang TA, Shen J, Hicks J, Chintagumpala M, Lau CC, Man TK. Plasma proteome predicts chemotherapy response in osteosarcoma patients. Oncology Reports. 2011;25:303–314. doi: 10.3892/or.2010.1111. [DOI] [PubMed] [Google Scholar]
- Li et al. (2020).Li M, Jin X, Li H, Wu G, Wang S, Yang C, Deng S. Key genes with prognostic values in suppression of osteosarcoma metastasis using comprehensive analysis. BMC Cancer. 2020;20:65. doi: 10.1186/s12885-020-6542-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li et al. (2010).Li Y, Liang Q, Wen YQ, Chen LL, Wang LT, Liu YL, Luo CQ, Liang HZ, Li MT, Li Z. Comparative proteomics analysis of human osteosarcomas and benign tumor of bone. Cancer Genet Cytogenet. 2010;198:97–106. doi: 10.1016/j.cancergencyto.2010.01.003. [DOI] [PubMed] [Google Scholar]
- Li et al. (2009).Li G, Zhang W, Zeng H, Chen L, Wang W, Liu J, Zhang Z, Cai Z Z. An integrative multi-platform analysis for discovering biomarkers of osteosarcoma. BMC Cancer. 2009;9:150. doi: 10.1186/1471-2407-9-150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ludwig & Weinstein (2005).Ludwig JA, Weinstein JN. Biomarkers in cancer staging, prognosis and treatment selection. Nature Reviews Cancer. 2005;5:845–856. doi: 10.1038/nrc1739. [DOI] [PubMed] [Google Scholar]
- Man et al. (2005).Man TK, Chintagumpala M, Visvanathan J, Shen J, Perlaky L, Hicks J, Johnson M, Davino N, Murray J, Helman L, Meyer W, Triche T, Wong KK, Lau CC. Expression profiles of osteosarcoma that can predict response to chemotherapy. Cancer Research. 2005;65:8142–8150. doi: 10.1158/0008-5472.can-05-0985. [DOI] [PubMed] [Google Scholar]
- Mintz et al. (2005).Mintz MB, Sowers R, Brown KM, Hilmer SC, Mazza B, Huvos AG, Meyers PA, Lafleur B, McDonough WS, Henry MM, Ramsey KE, Antonescu CR, Chen W, Healey JH, Daluski A, Berens ME, Macdonald TJ, Gorlick R, Stephan DA. An expression signature classifies chemotherapy-resistant pediatric osteosarcoma. Cancer Research. 2005;65:1748–1754. doi: 10.1158/0008-5472.can-04-2463. [DOI] [PubMed] [Google Scholar]
- Mo et al. (2019).Mo H, He J, Yuan Z, Wu Z, Liu B, Lin X, Guan J. PLK1 contributes to autophagy by regulating MYC stabilization in osteosarcoma cells. OncoTargets and Therapy. 2019;12:7527–7536. doi: 10.2147/OTT.S210575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nie & Peng (2018).Nie Z, Peng H. Osteosarcoma in patients below 25 years of age: an observational study of incidence, metastasis, treatment and outcomes. Oncology Letters. 2018;16:6502–6514. doi: 10.3892/ol.2018.9453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oliveira et al. (2007).Oliveira ID, Petrilli AS, Tavela MH, Zago MA, De Toledo SRC. TNF-alpha, TNF-beta, IL-6, IL-10, PECAM-1 and the MPO inflammatory gene polymorphisms in osteosarcoma. Journal of Pediatric Hematology/Oncology. 2007;29(5):293–297. doi: 10.1097/MPH.0b013e3180587e69. [DOI] [PubMed] [Google Scholar]
- Otoukesh et al. (2018).Otoukesh B, Boddouhi B, Moghtadaei M, Kaghazian P, Kaghazian M. Novel molecular insights and new therapeutic strategies in osteosarcoma. Cancer Cell International. 2018;18:158. doi: 10.1186/s12935-018-0654-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Savage et al. (2013).Savage SA, Mirabello L, Wang Z, Gastier-Foster JM, Gorlick R, Khanna C, Flanagan AM, Tirabosco R, Andrulis IL, Wunder JS, Gokgoz N, Patino-Garcia A, Sierrasesumaga L, Lecanda F, Kurucu N, Ilhan IE, Sari N, Serra M, Hattinger C, Picci P, Spector LG, Barkauskas DA, Marina N, de Toledo SR, Petrilli AS, Amary MF, Halai D, Thomas DM, Douglass C, Meltzer PS, Jacobs K, Chung CC, Berndt SI, Purdue MP, Caporaso NE, Tucker M, Rothman N, Landi MT, Silverman DT, Kraft P, Hunter DJ, Malats N, Kogevinas M, Wacholder S, Troisi R, Helman L, Fraumeni JF, Jr., Yeager M, Hoover RN, Chanock SJ. Genome-wide association study identifies two susceptibility loci for osteosarcoma. Nature Genetics. 2013;45:799–803. doi: 10.1038/ng.2645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shao et al. (2019).Shao YW, Wood GA, Lu J, Tang Q, Liu J, Molyneux S, Chen Y, Fang H, Adissu H, McKee T, Waterhouse P, Khokha R. Cross-species genomics identifies DLG2 as a tumor suppressor in osteosarcoma. Oncogene. 2019;38:291–298. doi: 10.1038/s41388-018-0444-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siegel, Miller & Jemal (2018).Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA: A Cancer Journal for Clinicians. 2018;68:7–30. doi: 10.3322/caac.21442. [DOI] [PubMed] [Google Scholar]
- Smida et al. (2017).Smida J, Xu H, Zhang Y, Baumhoer D, Ribi S, Kovac M, vonLuettichau I, Bielack S, O’Leary VB, Leib-Mösch C, Frishman D, Nathrath M. Genome-wide analysis of somatic copy number alterations and chromosomal breakages in osteosarcoma. International Journal of Cancer. 2017;141:816–828. doi: 10.1002/ijc.30778. [DOI] [PubMed] [Google Scholar]
- Smoot et al. (2010).Smoot ME, Ono K, Ruscheinski J, Wang P, Ideker T. Cytoscape 2.8: new features for data integration and network visualization. Bioinformatics. 2010;27:431–432. doi: 10.1093/bioinformatics/btq675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang et al. (2019).Tang Y, Gu Z, Fu Y, Wang J. CXCR3 from chemokine receptor family correlates with immune infiltration and predicts poor survival in osteosarcoma. Bioscience Reports. 2019;39(11):BSR20192134. doi: 10.1042/BSR20192134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uejima et al. (2011).Uejima D, Nishijo K, Kajita Y, Ishibe T, Aoyama T, Kageyama S, Iwaki H, Nakamura T, Iida H, Yoshiki T, Toguchida J. Involvement of cancer biomarker C7orf24 in the growth of human osteosarcoma. Anticancer Research. 2011;31:1297–1305. [PubMed] [Google Scholar]
- Walter, Sánchez-Cabo & Ricote (2015).Walter W, Sánchez-Cabo F, Ricote M. GOplot: an R package for visually combining expression data with functional analysis. Bioinformatics. 2015;31:2912–2914. doi: 10.1093/bioinformatics/btv300. [DOI] [PubMed] [Google Scholar]
- Wan-Ibrahim et al. (2015).Wan-Ibrahim WI, Singh VA, Hashim OH, Abdul-Rahman PS. Biomarkers for bone tumors: discovery from genomics and proteomics studies and their challenges. Molecular Medicine. 2015;21:861–872. doi: 10.2119/molmed.2015.00183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang et al. (2014).Wang Z, Cai H, Lin L, Tang M, Cai H. Upregulated expression of microRNA-214 is linked to tumor progression and adverse prognosis in pediatric osteosarcoma. Pediatric Blood & Cancer. 2014;61:206–210. doi: 10.1002/pbc.24763. [DOI] [PubMed] [Google Scholar]
- Wang et al. (2018).Wang H, Xie Q, Ou-Yang W, Zhang M. Integrative analyses of genes associated with idiopathic pulmonary fibrosis. Journal of Cellular Biochemistry. 2018 doi: 10.1002/jcb.28153. [DOI] [PubMed] [Google Scholar]
- Whelan & Davis (2018).Whelan JS, Davis LE. Osteosarcoma, chondrosarcoma, and chordoma. Journal of Clinical Oncology. 2018;36:188–193. doi: 10.1200/JCO.2017.75.1743. [DOI] [PubMed] [Google Scholar]
- Wittmann & Waterman-Storer (2001).Wittmann T, Waterman-Storer CM. Cell motility: can Rho GTPases and microtubules point the way? Journal of Cell Science. 2001;114:3795–3803. doi: 10.1242/jcs.114.21.3795. [DOI] [PubMed] [Google Scholar]
- Wu et al. (2019).Wu M, Ma Q, Liu D, Li X, Deng L, Li N, Shen J, Zhao Z, Chen J. CDC20 and its downstream genes: potential prognosis factors of osteosarcoma. International Journal of Clinical Oncology. 2019;24:1479–1489. doi: 10.1007/s10147-019-01500-3. [DOI] [PubMed] [Google Scholar]
- Xiong et al. (2020).Xiong Y, Lei J, Zhao J, Lu Q, Feng Y, Qiao T, Xin S, Han Y, Jiang T. A gene-based survival score for lung adenocarcinoma by multiple transcriptional datasets analysis. BMC Cancer. 2020;20:1046. doi: 10.1186/s12885-020-07473-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiong et al. (2009).Xiong W, Niu PY, Zhu WT, Chen J. Tenascin-C as a prognostic biomarker in osteosarcoma? Chinese Medical Journal. 2009;122:2737–2743. [PubMed] [Google Scholar]
- Yang et al. (2018).Yang Y, Han L, He Z, Li X, Yang S, Yang J, Zhang Y, Li D, Yang Y, Yang Z. Advances in limb salvage treatment of osteosarcoma. Journal of Bone Oncology. 2018;10:36–40. doi: 10.1016/j.jbo.2017.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye et al. (2015).Ye F, Wang H, Zhang L, Zou Y, Han H, Huang J. Baicalein induces human osteosarcoma cell line MG-63 apoptosis via ROS-induced BNIP3 expression. Tumor Biology. 2015;36:4731–4740. doi: 10.1007/s13277-015-3122-y. [DOI] [PubMed] [Google Scholar]
- Yu et al. (2014).Yu W, Tang L, Lin F, Li D, Wang J, Yang Y, Shen Z. Stereotactic radiosurgery, a potential alternative treatment for pulmonary metastases from osteosarcoma. International Journal of Oncology. 2014;44:1091–1098. doi: 10.3892/ijo.2014.2295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu et al. (2012).Yu G, Wang L, Han Y, He Q. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics: A Journal of Integrative Biology. 2012;16:284–287. doi: 10.1089/omi.2011.0118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuan et al. (2012).Yuan J, Chen L, Chen X, Sun W, Zhou X. Identification of serum microRNA-21 as a biomarker for chemosensitivity and prognosis in human osteosarcoma. Journal of International Medical Research. 20‘2;40:2090–2097. doi: 10.1177/030006051204000606. [DOI] [PubMed] [Google Scholar]
- Zamborsky et al. (2019).Zamborsky R, Kokavec M, Harsanyi S, Danisovic L. Identification of prognostic and predictive osteosarcoma biomarkers. Medical Sciences. 2019;7(2):28. doi: 10.3390/medsci7020028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang, Lan & Lin (2018).Zhang J, Lan Q, Lin J. Identification of key gene modules for human osteosarcoma by co-expression analysis. World Journal of Surgical Oncology. 2018;16:89. doi: 10.1186/s12957-018-1381-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The following information was supplied regarding data availability:
The raw measurements are available in the Supplemental Files.