Skip to main content

ORIGINAL RESEARCH article

Front. Genet., 22 July 2022
Sec. Computational Genomics
This article is part of the Research Topic Integrative Analysis of Single-Cell and/or Bulk Multi-omics Sequencing Data View all 14 articles

8-Gene signature related to CD8+ T cell infiltration by integrating single-cell and bulk RNA-sequencing in head and neck squamous cell carcinoma

  • 1Department of Oral and Maxillofacial Surgery, Tianjin Medical University School and Hospital of Stomatology, Tianjin, China
  • 2Department of Prosthodontics, Tianjin Medical University School and Hospital of Stomatology, Tianjin, China

Background: CD8+ T cells, a critical component of the tumor immune microenvironment, have become a key target of cancer immunotherapy. Considering the deficiency of robust biomarkers for head and neck squamous cell carcinoma (HNSCC), this study aimed at establishing a molecular signature associated with CD8+T cells infiltration.

Methods: Single-cell RNA sequencing data retrieved from the Gene Expression Omnibus (GEO) database was analyzed to obtain the different cell types. Next, the cell proportions were investigated through deconvolution of RNA sequencing in the Cancer Genome Atlas (TCGA) database, and then the immune-related genes (IRGs) were identified by weighted gene co-expression network analysis (WGCNA). LASSO-Cox analysis was employed to establish a gene signature, followed by validation using a GEO dataset. Finally, the molecular and immunological properties, and drug responses between two subgroups were explored by applying “CIBERSORT”, “ESTIMATE”, and single sample gene set enrichment analysis (ssGSEA) methods.

Results: A total of 215 differentially expressed IRGs were identified, of which 45 were associated with the overall survival of HNSCC. A risk model was then established based on eight genes, including DEFB1, AICDA, TYK2, CCR7, SCARB1, ULBP2, STC2, and LGR5. The low-risk group presented higher infiltration of memory activated CD4+ T cells, CD8+ T cells, and plasma cells, as well as a higher immune score, suggesting that they could benefit more from immunotherapy. On the other hand, the high-risk group showed higher abundance of activated mast cells and M2 macrophages, as well as a lower immune score.

Conclusion: It was evident that the 8-gene signature could accurately predict HNSCC prognosis and thus it may serve as an index for clinical treatment.

Introduction

Head and neck squamous cell carcinoma (HNSCC) is the seventh most common malignancy worldwide (Siegel et al., 2020). Despite the effective and aggressive treatment strategies involving surgery combined with radio- and chemotherapy, patients with advanced stage HNSCC only have a 50% five-year survival rate (Vigneswaran and Williams, 2014). In recent years, immunotherapy involving checkpoint inhibitors blocking programmed cell death protein 1 (PD-1) or programmed death ligand-1 (PD-L1) has been approved for clinical use, with preliminary results showing that the strategy significantly improves the overall survival of recurrent or metastatic HNSCC patients. However, several clinical trials have demonstrated that anti-PD-1/PD-L1 therapy is only beneficial to a few patients (Ferris et al., 2016; Siu et al., 2019). Studies have suggested that CD8+ T lymphocytes substantially express PD-1 and may play an important role in the efficacy of immunotherapy (Jia et al., 2020). It is worth noting that high dense infiltration of CD8+ T cells in HNSCC patients is generally associated with a good prognosis (Fridman et al., 2017). Moreover, PD-1+ CD8+ T cells showed excellent anti-tumor effect in an anti-PD1-resistant murine HNSCC model (Xu et al., 2020). Therefore, there is an urgent need to explore the molecular mechanisms associated with CD8+ T cells infiltration.

Single-cell RNA sequencing (scRNA-seq) has been the subject of rapid technological developments in the last decade, thereby resulting in significant improvements in describing and defining the tumor heterogeneity at a single-cell level (Qi et al., 2019). Besides, application of scRNA-seq to characterize the tumor microenvironment (TME) may provide valuable insights into immune landscapes and even effective immunotherapy strategies (Kurten et al., 2021). Similarly, the gene signature identified based on immune molecular characteristics might be a strong predictor of clinical outcome and immunotherapy response (Song et al., 2022). However, the predictive potential of the molecular mechanisms describing immunophenotypic features in HNSCC have not yet been elucidated.

This study explored the mechanism associated with infiltration of CD8+ T cells through integrating bulk and scRNA sequencing. Specifically, a LASSO-Cox regression risk model was built and verified based on the hub immune-related genes (IRGs) identified by weighted gene co-expression network analysis (WGCNA) (Langfelder and Horvath, 2008). Next, we comprehensively represented the various immune features of an 8-gene signature using “ESTIMATE” (Yoshihara et al., 2013), “CIBERSORT” (Newman et al., 2015), single sample gene set enrichment analysis (ssGSEA) approaches, and immunophenoscore (IPS) data. It is expected that the identified risk score will not only be used as an efficient indicator for HNSCC prognosis, but also as a potential therapeutic target.

Materials and methods

The study design is illustrated using a flow diagram (Figure 1).

FIGURE 1
www.frontiersin.org

FIGURE 1. Flow chart schematic of this study.

Data acquisition

The single cell RNA-sequencing profile of GSE103322 dataset (Puram et al., 2017), comprising 5,902 single cells of 18 patients, was downloaded from the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/) (accessed date 13 October 2021). HNSCC RNA-sequencing, clinical and mutation data were downloaded from The Cancer Genome Atlas (TCGA) database using the GDC Data Portal (https://portal.gdc.cancer.gov/ (accessed date 13 October 2021). The Fragments per Kilobase per Million (FPKM) values were first converted to transcripts per million kilobase (TPM) values. To validate the prognostic power of the model, the transcriptome and clinical files of the GSE65858 dataset, containing 270 HNSCC samples, were obtained from the GEO database (Wichmann et al., 2015). Notably, a total of 2,720 IRGs were obtained from the ImmPort (https://www.immport.org/home) and InnateDB (https://www.innatedb.com/) databases (accessed date 13 October 2021).

Processing of single-cell and bulk RNA-seq files

The “Seurat” (version 4.1.1) package in R (version 4.1.2) was applied to group 5,902 cells into appropriate clusters, with the resolution set to 0.8. Results were presented by employing the T-distributed stochastic neighbor embedding (t-SNE) for dimension reduction. Next, diverse cell types, B/plasma cells, endothelial cells, regulatory T cells (Treg cells), mast cells, CD8+ T cells, epithelial cells, dendritic cells, macrophages, fibroblasts, and CD4+ T cells were identified based on their specific markers. The “Cellchat” (version 1.1.3) package was used to analyze the cell–cell communication, and then deconvolution was performed using the “BisqueRNA” (version 1.0.5) method (Jew et al., 2020) to calculate the cells fractions of TCGA bulk profiles. Based on the TCGA RNA-seq profiles, differentially expressed genes (DEGs) were identified with FDR < 0.05 and |log2FC| > 1 set as the cutoff values.

Determination of immune-related candidate genes

The differential IRGs were determined by overlapping DEGs and IRGs, and then used to screen the hub genes by WGCNA (version 1.7.0). First, Pearson correlation coefficient was determined for every gene, and a suitable soft threshold β was automatically selected through the pick Soft Threshold function. Next, gene expression similarity matrix was transformed into an adjacency matrix using a network type of signed and soft powers β = 3, followed by employing TOM (topological overlap measure) to cluster genes into network modules. The 1-TOM (dissimilarity TOM) was then applied as the input for hierarchical clustering and the “DynamicTreeCut” algorithm was employed to detect modules (clusters of highly interconnected genes) as branches of the dendrogram. Finally, we identified and selected a module (215 genes) that significantly correlated with CD8+ T cells content. Kaplan–Meier (KM) survival and univariate Cox analysis were utilized to determine the hub genes associated with survival at a threshold of p < 0.05.

Development of a prognostic signature in TCGA (n = 498)

LASSO-Cox analysis was performed using “glmnet” package to determine the optimal prognostic gene set. The risk score of each HNSCC patient was determined as the sum of normalized gene expression values weighted by their LASSO-Cox coefficients in accordance with the following formula:

risk score=i=1nCoefiExpi

Where Coefi indicates the calculated regression coefficient of each gene in the LASSO-Cox model and  Expi represents the mRNA expression value. Kaplan-Meier (KM) analysis, receiver operating characteristic (ROC) curves, and univariate and multivariate Cox regression analyses were employed to validate the independent prognostic factors in TCGA-HNSC and GSE65858 datasets. For better clinical prediction of HNSCC patient survival probabilities, a nomogram was constructed using the “rms” R package based on multivariate Cox analysis results. The concordance index (C-index) of the nomogram was calculated to assess the discriminative ability.

Immune features and therapy prediction in distinct risk groups

“CIBERSORT” (version 1.03) and “ESTIMATE” (version 1.0.13) analyses were applied to determine the abundance of 22 immune cells and immune infiltration scores. The ssGSEA approach was employed via the “GSVA” (version 1.42.0) package to compute the enrichment scores of 29 immune features (Hänzelmann et al., 2013). To predict the susceptibility of eight common chemotherapeutic drugs (5-Fluorouracil, bleomycin, cetuximab, cisplatin, docetaxel, methotrexate, rapamycin, and sunitinib) for HNSCC, the “pRRophetic” (version 0.5) method was performed to evaluate the half-maximal inhibitory concentration (IC50) of patients in distinct groups (Geeleher et al., 2014). The immunophenoscore (IPS) of HNSCC patients, which is a scoring scheme that characterizes the determinants of tumor immunogenicity (Charoentong et al., 2017), were downloaded from The Cancer Imaging Archive (TCIA) database (https://tcia.at/home, accessed date 15 November 2021). To predict the anti-CTLA4 and anti-PD1 responses, patients with different IPS were further compared between the two risk groups. Finally, the “Maftools” (version 2.10.05) (Mayakonda et al., 2018) package was used to determine the tumor mutational burden (TMB) and identify the driver genes.

Enrichment analysis

The reference gene sets of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway (c2. cp.kegg.v7.5.1. symbols.gmt) were obtained from the MSigDB database (https://www.gsea-msigdb.org/gsea/msigdb, accessed date 15 November 2021). GSEA software (version 4.2.3) and Gene Set Variation Analysis (GSVA) were conducted to determine the KEGG pathways with FDR < 0.05.

Results

Cell typing in head and neck squamous cell carcinoma scRNA-seq and deconvolution in the Cancer Genome Atlas-HNSC

We first collected the Smart-seq2 profile data of 5,902 cells in the GSE103322 dataset. Principal component analysis (PCA) and t-SNE analysis identified 27 cell clusters (Figure 2A). According to expressions of marker genes, 10 distinct cell clusters were identified, including CD8+ T cells, macrophages, CD4+ T cells, fibroblasts, endothelial cells, B/plasma cells, mast cells, Treg cells, epithelial cells, and dendritic cells (Figures 2B, 3A). GSVA results showed that “MYC_TARGETS_V2” and “MYC_TARGETS_V1” were activated in epithelial cells, whereas “HYPOXIA” was abundant in fibroblasts (Figure 3B). Results obtained after applying the “CellChat” method showed that there was a strong connectivity between different cell types (Figures 3C,D). Next, the BisqueRNA approach was performed to calculate proportions of the 10 cell types by deconvoluting the TCGA bulk profiles. Supplementary Table S1 shows proportion of the 10 cell types in 497 samples. Survival analysis demonstrated that mast cells (p = 0.001), CD8+ T cells (p = 0.011), and Treg cells (p = 0.002) were significantly associated with HNSCC outcome (Figures 2C–E). Moreover, univariate Cox analysis indicated that Treg cells were associated with good outcome (p = 0.018), whereas mast cells were intimately linked to poor prognosis (p = 0.019) (Figure 2F).

FIGURE 2
www.frontiersin.org

FIGURE 2. Identification of the HNSCC-associated cell subtypes. (A) t-SNE plot classified cell clusters based on scRNA sequencing data. (B) t-SNE plot identified the various cell subtypes. (C–E) Kaplan-Meier survival analysis of three cell subtypes using the deconvolved TCGA data. (C) CD8+ T cells: p = 0.011, (D) Mast cells: p = 0.001, (E) Treg cells: p = 0.002. (F) Univariate analysis of ten cell subtypes.

FIGURE 3
www.frontiersin.org

FIGURE 3. (A) The heatmap depicting marker genes associated with ten cell subtypes. (B) GSVA enrichment analysis of the cell subtypes. (C,D) Cell-cell communication network of ten cell subtypes.

Construction and validation of a gene risk signature associated with CD8+ T cells

First, 9,244 DEGs were obtained from the TCGA-HNSC dataset comprising 501 tumor and 44 normal samples (Figure 4A). Subsequently, 2,720 IRGs from ImmPort and InnateDB databases were matched with DEGs, from which 840 differentially expressed IRGs were obtained for further analysis (Figure 4B). Based on the 840 IRGs and proportions of the 10 cell types in TCGA, the weighted gene co-expression network was generated using the soft-thresholding power β = 3, which resulted in identification of 10 modules (Figures 5A,B). To further explore the features of CD8+ T cells infiltration, we selected the turquoise module (215 genes) which had the strongest correlation with CD8+ T cells (r = 0.86, p = 1e-17). Univariate Cox analysis demonstrated that 45 of the 215 hub genes were closely associated with HNSCC survival (Figure 5C). Therefore, the 45 genes were subjected to LASSO regression analysis to identify the optimal penalty coefficient (Figures 5D,E). The survival analysis identified eight genes, including DEFB1, AICDA, TYK2, CCR7, SCARB1, ULBP2, STC2, and LGR5, which were significantly associated with HNSCC prognosis (Figures 6A–H). The eight risk regression coefficients were then employed to compute individual risk score of HNSCC patients according to the following formula:

Risk score=(0.097)DEFB1+(0.444)AICDA+(0.175)TYK2+(0.071)CCR7+0.020SCARB1+0.079ULBP2+0.161STC2+(0.128)LGR5

FIGURE 4
www.frontiersin.org

FIGURE 4. The heatmap (A) and Venn diagram (B) identified the differentially expressed genes (DEGs) and immune-related DEGs between tumor and normal samples in TCGA.

FIGURE 5
www.frontiersin.org

FIGURE 5. Development of an 8-gene signature. (A) The Cluster dendrogram of co-expression network modules obtained by WGCNA. (B) Correlation heatmap among ten co-expression modules and the levels of cell subtypes. The turquoise module had the greatest correlation with CD8+ T cells (r = 0.86, p = 1e-17). (C) Univariate analysis of 45 immune-related hub genes. (D) LASSO coefficient profiles of 45 immune-related genes. (E) Tuning parameter selection in the LASSO model using ten-time cross-validation.

FIGURE 6
www.frontiersin.org

FIGURE 6. Survival analysis of eight genes in risk signature. (A) ULBP2: p < 0.001. (B) SCARB1: p = 0.003. (C) STC2: p < 0.001. (D) DEFB1: p < 0.001. (E) AICDA: p = 0.003. (F) CCR7: p < 0.001. (G) TYK2: p < 0.001. (H) LGR5: p < 0.001. (I,J) Survival analysis of the 8-gene signature in TCGA (p < 0.001) and GSE65858 cohorts (p = 0.005).

Next, the 498 HNSCC patients were stratified into high- and low-risk groups based on the median risk score. KM survival analysis results indicated that the high-risk group patients showed poorer outcomes compared to the low-risk group (p < 0.001, Figure 6I). Consistently, similar results were observed in the GSE65858 dataset (p = 0.005, Figure 6J).

Validation in the Cancer Genome Atlas-HNSC and GSE65858 cohorts, and scRNA-seq data

The risk score, survival status distributions of HNSCC patients, and correlation analysis are displayed in Figure 7. Results demonstrated that survival reduced with rising risk score, and there was a significant correlation between risk score and survival in TCGA cohort (r = -0.2, p = 6.4e-06). Time-dependent ROC and calibration curves at one-, three-, and five-years were then constructed (Figures 8A,B). In the TCGA cohort, the areas under the ROC curves (AUCs) were 0.679, 0.703, and 0.644 for 1-, 3-, and 5-years survival, respectively. In both the TCGA and GSE65858 cohorts, univariate and multivariate Cox analyses demonstrated that the risk score was an independent predictor for prognosis (Figures 8C–F). To determine the cells that these eight genes were enriched, the distribution plots for expressions of the eight genes in the 10 cell types identified in the GSE103322 dataset were generated and are shown in Figures 9A–I. Results showed that the expression levels of DEFB1 and ULBP2 were higher in epithelial cells, whereas TYK2 and CCR7 levels were abundant in dendritic cells. In addition, the endothelial cells had higher expressions of SCARB1 and STC2, and LGR5 was highly expressed in both dendritic cells and fibroblasts. Based on proportions of the 10 cell types obtained after deconvolution, correlation analysis was performed to evaluate the association among proportion of CD8+ T cells and risk score. Obtained results revealed that fractions of CD8+ T cells declined as the risk score increased (r = −0.41, p < 2.2 e-16, Figures 9J,K).

FIGURE 7
www.frontiersin.org

FIGURE 7. The relationship between risk score and HNSCC survival. (A–D) Distribution of risk score and survival status of 8-gene signature in TCGA (A,C) and GSE65858 (B,D) cohorts. (E,F) The correlation analysis between overall survival (OS) and risk score in TCGA (E) and GSE65858 (F) cohorts.

FIGURE 8
www.frontiersin.org

FIGURE 8. Validation of the 8-gene signature in TCGA and GSE65858 cohorts. (A,B) The ROC and calibration curves for determining the accuracy of model. (C–F) Univariate and multivariate analysis of clinical features and risk score.

FIGURE 9
www.frontiersin.org

FIGURE 9. Verification using single-cell sequencing data. (A–H) Colors indicating the localization of the expression of eight genes: AICDA, CCR7, DEFB1, LGR5, SCARB1, STC2, TYK2, and ULBP2. (I) Heatmap depicting expressions of the eight genes in the cell subtypes. (J) The levels of CD8+ cells in TCGA deconvoluted data between low- and high-risk groups (p = 1.4 e-13). (K) Correlation analysis between CD8+ T cells levels and risk score (r = −0.41, p < 2.2 e-16).

Construction of a nomogram for clinical practice

A heatmap was generated to depict the changes in expression of the eight genes between different clinical subgroups (Figure 10A). The performance of the risk score was then explored in different clinicopathological subgroups, including clinical stage (stage I-III and stage IV), age (<=60 and >60), grade (G1-2 and G3-4), T stage (T0-2 and T3-4), N stage (N0-1 and N2-3), and gender (female and male). According to the survival analysis results, HNSCC patients with high-risk scores consistently had a poorer outcome in all subgroups (Figures 10B–G). Next, the three remarkable variables in the multivariate analysis, including age, N stage, and risk score, were selected and used to build a nomogram (C-index: 0.676) for estimating the 1-, 3-, and 5-year survival rate (Figure 11A). By drawing a vertical line to the axis points, we could estimate patient survival based on total points. Overall, the calibration curves and the AUC’s (1-, 3-, and 5-year: 0.733, 0.749, and 0.691, respectively) suggested that the risk model could accurately predict the HNSCC survival rate (Figures 11B,C).

FIGURE 10
www.frontiersin.org

FIGURE 10. The relationship between risk signature and the clinical characteristics. (A) The heatmap depicting eight gene expressions among distinct clinical patterns. (B–G) Kaplan-Meier survival analysis according to the 8-gene signature stratified by clinicopathological factors. (B) age<=60: p = 0.011, age>60: p < 0.001. (C) Female: p = 0.001, Male: p < 0.001. (D) G1-2: p = 0.002, G3-4: p < 0.001. (E) N0-1: p < 0.001, N2-3: p = 0.009. (F) Stage I-III: p = 0.004, Stage IV: p = 0.001. (G) T0-2: p = 0.006, T3-4: p < 0.001.

FIGURE 11
www.frontiersin.org

FIGURE 11. Construction of a nomogram for predicting survival of HNSCC patients. (A) Nomogram using two clinical traits (N stage and age) and the risk score. (B,C) The calibration and ROC curves for determining the reliability of the nomogram to predict one-, three-, and five-year survival rates.

The immune landscape of the two risk groups

To elucidate the biological characteristics activated in distinct risk groups, KEGG pathway enrichment analysis was performed using GSVA and GSEA methods. By setting the adjusted p value (FDR) < 0.05, a total of 51 and 16 pathways were obtained in GSVA and GSEA, respectively (Figures 12A,B). Several overlapping immunoregulatory processes were enhanced in the low-risk group, including “hematopoietic cell lineage”, “T cell receptor signaling pathway”, “antigen processing and presentation” and “natural killer cell-mediated cytotoxicity”. To describe the patterns of immune infiltrations, CIBERSORT and ESTIMATE methods were implemented for calculating the cell fractions and immune-related scores of HNSCC samples (Figures 13A,B). The low-risk group showed more significant infiltrations of CD8+ T cells, M1 macrophages, follicular helper T cells, plasma cells, regulatory T cells, and memory activated CD4+ T cells, as well as a higher immune score. With regard to the high-risk group, abundant infiltrations of activated mast cells, M2 macrophages, resting NK cells, and low immune score were observed. The ssGSEA approach was then applied to estimate the scores of specific immune functions and cells. Results revealed significant differences of most immune cells and functions between high- and low-risk groups (Figure 13C). Besides, 15 immune checkpoint molecules (IFNG, GZMB, HAVCR2, CD274, CD8A, PDCD1, TBX2, IDO1, GZMA, LAG3, CXCL10, CTLA4, PRF1, CXCL9, and TNF) were selected and their expressions were compared between the two risk groups (Figure 13D). Based on the correlation analysis results, it was evident that the expressions of CD274 and CTLA4 in the two groups were significantly different (CD274: p = 0.0006; CTLA4: p = 2.5e-14), and decreased as the risk score rose (CD274: r = −0.18, p = 3.6 e-05; CTLA4: r = −0.43, p < 2.2 e-16) (Figures 13E–H). Next, the pRRophetic algorithm was applied to predict the IC50 of eight common chemotherapeutic drugs between the two groups. Patients with a high-risk score showed an increased susceptibility to bleomycin (p = 0.00014), cisplatin (p = 3.2e-05), and methotrexate (p = 0.039). On the other hand, low-risk group patients showed increased sensitivity to rapamycin (p = 5.6e-06) (Figures 14A–H). To forecast the response to anti-PD1 and anti-CTLA4 immunotherapy, the IPS scores of HNSCC patients were used to compare the two risk groups (Figure 14I–L). Results indicated that patients in the low-risk group exhibited higher IPS scores and showed greater response to anti-PD1 therapy and anti-PD1 plus anti-CTLA4 therapy (ips_ctla4_neg_pd1_pos: p = 0.0054, ips_ctla4_pos_pd1_pos: p = 1.6e -05) relative to patients in the high-risk group. Given the important role of TMB in prognosis, the intrinsic connection between TMB and risk score was explored to assess genetic signature. It was found that the high-risk group exhibited higher TMB (Figure 15A). A significant correlation was observed between TMB and risk score (r = 0.22, p = 1.3e-06, Figure 15B). Survival curve suggested that a low TMB/low risk group showed a great outcome compared with the other groups (p < 0.001, Figure 15C). The top 20 driver genes with the highest alteration frequency were analyzed (Figures 15D,E) and four genes (TP53, PKHD1L1, DNAH9, FAT1) were significantly different between high- and low-risk groups (Supplementary Table S2).

FIGURE 12
www.frontiersin.org

FIGURE 12. Functional enrichment characteristics of the risk signature. (A) Different activities of KEGG pathway scored by GSVA between high- and low-risk groups. (B) GSEA analysis showing the sixteen KEGG functional pathways enriched in low-risk group.

FIGURE 13
www.frontiersin.org

FIGURE 13. Patterns of immune cells infiltration in two risk groups. (A) The box plot showing the fractions of 22 infiltrating immune cells and immune-related scores based on CIBERSORT and ESTIMATE algorithms. (B) A heatmap presenting the 22 immune cells in the two risk score subgroups with different immune-related scores. (C) The ssGSEA scores for 29 immune gene sets. (D) Differential expressions of the 15 immune checkpoint-related genes. (E) CD274 expression difference among the high- and low-risk groups (p = 0.0006). (F) The spearman correlation plot between CD274 expression and risk score (r = −0.18, p = 3.6 e-05). (G) CTLA4 expression difference among the high- and low-risk groups (p = 2.5 e-14). (H) The spearman correlation plot between CTLA4 expression and risk score (r = −0.43, p < 2.2 e-16). ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ns: no significance.

FIGURE 14
www.frontiersin.org

FIGURE 14. Drug response prediction between the two risk groups. (A–H) The IC50 of eight common chemotherapeutic agents (5-Fluorouracil, Bleomycin, Cetuximab, Cisplatin, Docetaxel, Methotrexate, Rapamycin, and Sunitinib) and correlation analysis with risk score. (I–L) The difference of immunophenoscore (IPS) scores among high- and low-risk groups.

FIGURE 15
www.frontiersin.org

FIGURE 15. (A) TMB difference among the high and low risk groups. p = 0.00028. (B) The Spearman correlation analysis between risk score and TMB. r = 0.22, p = 1.3e-06. (C) K-M survival analysis stratified by both TMB and risk scores. p < 0.001. (D,E) Distribution of the top 20 variant mutated genes among high (D) and low (E) risk groups. The waterfall plot showing the genetic alterations types.

Discussion

Immunotherapy has been successful used to treat cancer patients in the advanced tumor stage. Nevertheless, clinical application of the strategy is hampered by several limitations, including low response rates, development of serious side effects, and drug resistance (Sacco et al., 2021). One of the key reasons for these limitations is the paucity of potential predictive markers. In the present study, we calculated the proportion of CD8+ T cells, and selected IRGs-related to CD8+ T cells infiltration by integrating scRNA and bulk sequencing profiles. As a result, 215 differential IRGs were identified by WGCNA, of which 45 genes were significantly associated with HNSCC survival. Subsequently, we developed and validated an 8-gene risk model which may be useful for predicting prognosis and immunotherapeutic effect.

The eight critical genes, including DEFB1, AICDA, TYK2, CCR7, SCARB1, ULBP2, STC2, and LGR5, play essential roles in tumor progression and immune-modulatory effects. For example, DEFB1, the human antimicrobial peptide defensin β 1, is considered as a potential tumor suppressor gene and has been shown to mediate PI3K/mTOR signaling, thereby leading to death of tumor cells (Sun et al., 2006; Lee et al., 2015). DEFB1 was also found to be theoretically useful as a prognostic biomarker for HNSCC (Han et al., 2014). Moreover, DEFB1 was commonly detected in epithelial cells, which is consistent with our results. UL16-binding protein 2 (ULBP2), a ligand of the activating NK cell receptor NKG2D, was found to be engaged in target recognition by NK cells (Textor et al., 2011). A previous study confirmed that the soluble ULBP2 secreted by cancer cells contributed to the immune escape (Waldhauer and Steinle, 2006). Herein, we observed that ULBP2 was upregulated in epithelial cells. Meanwhile, ULBP2 has been shown to be a prognosis indicator for several cancers, such as lung cancer and pancreatic cancer (Chang et al., 2011; Yamaguchi et al., 2012). The activation-induced cytidine deaminase (AICDA) is an essential enzyme of the adaptive immune system. A recent study found that elevated expression of AICDA regulates the function of B cells in regional lymph nodes and significantly improves prognosis of HNSCC patients (Pylaeva et al., 2021). Tyrosine kinase 2 (TYK2), a member of the Janus kinase (JAK) family, has emerged as both a promising biomarker and a target for anti-cancer therapies (Borcherding et al., 2021). It has been reported that high expression of TYK2 is associated with better prognosis of HNSCC (Fang et al., 2021). A recent review concluded that CC motif chemokine receptor (CCR7) is correlated with good outcomes of HNSCC patients (Korbecki et al., 2020). However, if located on cancer cells, CCR7 and its ligands (CCL19/CCL21) is a vital axis for carcinogenic properties, such as epithelial-mesenchymal transition (EMT) tumor invasion and migration (Chen et al., 2020; Korbecki et al., 2020). Notably, the present study found that CCR7 was predominantly expressed in dendritic cells. SCARB1 has been demonstrated to be involved in cholesterol metabolism, thereby facilitating cancer progression (Gutierrez-Pajares et al., 2016). In addition, stanniocalcin-2 (STC2) exerted a significant role in a wide variety of signaling pathways in HNSCC apoptosis and autophagy (Li et al., 2020). Studies have revealed that downregulated expression of STC2 can suppress growth of HNSCC cells (Li et al., 2019; Li et al., 2020). Moreover, the leucine-rich repeat-containing G protein-coupled receptor LGR5 participated in Wnt signaling and was intimately linked to the severity of HNSCC (Dalley et al., 2015).

Given the important role of immune cell infiltrations in the diagnosis and treatment of diseases, we further explored the immune landscape in different HNSCC groups. Based on the degree of immune cell infiltrations, particularly CD8+ T cells, the tumor phenotypes can be defined as two major patterns, “hot” and “cold”, which are associated with good and poor antitumor immune responses, respectively (Galon and Bruni, 2019). This study explored the abundance of immune cells and functions using CIBERSORT, ESTIMATE, and ssGSEA methods. According to the obtained results, the low-risk group exhibited more infiltration of CD8+ T cells, memory activated CD4+ T cells, and plasma cells, as well as higher immune score, and thus can be categorized as “hot” tumor phenotype. On the other hand, the high-risk group showed greater abundance of activated mast cells, resting NK cells, and M2 macrophages, and lower immune score, suggesting the “cold” tumor phenotype. Furthermore, the immune checkpoint-related genes exhibited relatively high expressions in the low-risk group, including IFNG, PRF1, GZMA, GZMB, CXCL10, CXCL9, CD8A, CD274 (PD-L1), HAVCR2, IDO1, LAG3, CTLA4, and PDCD1. Studies have confirmed that infiltration of M2 macrophages is associated with tumorigenic chronic inflammation with secretion of protumorigenic factors, such as IL-6, VEGF, and TGFβ (Ruffell and Coussens, 2015). Accumulating evidence suggests that preexisting CD8+ T cells and PD-L1 expression are generally correlated with improved efficacy of immunotherapy (Farhood et al., 2019; Gavrielatou et al., 2020). Consistently, our results suggested that patients with low-risk score, as a consequence of higher IPS scores, had more vigorous immune responses to anti-PD1 therapy and anti-PD1 plus anti-CTLA4 therapy. Moreover, patients in the two groups exhibited varying sensitivity to four common chemotherapeutic drugs, including bleomycin, cisplatin, methotrexate, and rapamycin (Cramer et al., 2019). Notably, previous studies have verified the therapeutic safety and effectiveness of chemotherapy in combination with PD-L1 blockade (Burtness et al., 2019; Cohen et al., 2019). Nevertheless, different sensitivities to 5-Fluorouracil, cetuximab, docetaxel, and sunitinib were not observed in this study. TMB level was considered to be an indicator of immunotherapy response (Rizvi et al., 2015). We then examined the relationship between TMB and the risk score. The alteration frequency of TP53, PKHD1L1, DNAH9 and FAT1 was significantly different between high- and low-risk groups. TP53 is one of the most frequently mutated genes in HNSCC and TP53 mutations play a critical role in tumorigenesis and progression (Nathan et al., 2022). Understanding the DNAH9 and FAT1 mutations may contribute to cancer surveillance and treatment (Huang et al., 2021; Yang et al., 2022). Investigation of the mutational signatures may allow for an improved selection of immunotherapies in individual patients.

However, this study was limited by the fact that it lacked experimental and clinical pathology studies to validate the function of the eight genes. Therefore, further clinical trials are needed to confirm the predictive potential of the risk signature.

Conclusion

In conclusion, by comprehensively analyzing the single-cell and bulk RNA sequencing of HNSCC, this study developed and externally validated a novel and robust model based on eight CD8+ T cells-related genes. It is expected that the 8-gene signature will facilitate understanding of HNSCC immune characteristics, predict prognosis of HNSCC patients, and guide the clinical use of immunotherapy.

Data availability statement

The datasets presented in this study can be found in online repositories. The names of the repository/repositories and accession number(s) can be found in the article/Supplementary Material.

Author contributions

JZ designed the study and collected the data. SZ and WZ carried out the data analyses, prepared all figures and tables and wrote the manuscript. All authors participated in improving the writing of the manuscript and approved the submitted version.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fgene.2022.938611/full#supplementary-material

References

Borcherding, D. C., He, K., Amin, N. V., and Hirbe, A. C. (2021). TYK2 in cancer metastases: Genomic and proteomic discovery. Cancers (Basel) 13 (16), 4171. doi:10.3390/cancers13164171

PubMed Abstract | CrossRef Full Text | Google Scholar

Burtness, B., Harrington, K. J., Greil, R., Soulières, D., Tahara, M., de Castro, G., et al. (2019). Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): A randomised, open-label, phase 3 study. Lancet 394 (10212), 1915–1928. doi:10.1016/s0140-6736(19)32591-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Chang, Y. T., Wu, C. C., Shyr, Y. M., Chen, T. C., Hwang, T. L., Yeh, T. S., et al. (2011). Secretome-based identification of ULBP2 as a novel serum marker for pancreatic cancer detection. PLoS One 6 (5), e20029. doi:10.1371/journal.pone.0020029

PubMed Abstract | CrossRef Full Text | Google Scholar

Charoentong, P., Finotello, F., Angelova, M., Mayer, C., Efremova, M., Rieder, D., et al. (2017). Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep. 18 (1), 248–262. doi:10.1016/j.celrep.2016.12.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Shao, Z., Jiang, E., Zhou, X., Wang, L., Wang, H., et al. (2020). CCL21/CCR7 interaction promotes EMT and enhances the stemness of OSCC via a JAK2/STAT3 signaling pathway. J. Cell. Physiol. 235 (9), 5995–6009. doi:10.1002/jcp.29525

PubMed Abstract | CrossRef Full Text | Google Scholar

Cohen, E. E. W., Soulières, D., Le Tourneau, C., Dinis, J., Licitra, L., Ahn, M.-J., et al. (2019). Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): A randomised, open-label, phase 3 study. Lancet 393 (10167), 156–167. doi:10.1016/s0140-6736(18)31999-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Cramer, J. D., Burtness, B., Le, Q. T., and Ferris, R. L. (2019). The changing therapeutic landscape of head and neck cancer. Nat. Rev. Clin. Oncol. 16 (11), 669–683. doi:10.1038/s41571-019-0227-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Dalley, A. J., Abdul Majeed, A. A., Pitty, L. P., Major, A. G., and Farah, C. S. (2015). LGR5 expression in oral epithelial dysplasia and oral squamous cell carcinoma. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 119 (4), 436–440. e431. doi:10.1016/j.oooo.2014.11.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Fang, L., Wang, W., Shi, L., Chen, Q., and Rao, X. (2021). Prognostic values and clinical relationship of TYK2 in laryngeal squamous cell cancer. Med. Baltim. 100 (34), e27062. doi:10.1097/MD.0000000000027062

CrossRef Full Text | Google Scholar

Farhood, B., Najafi, M., and Mortezaee, K. (2019). CD8(+) cytotoxic T lymphocytes in cancer immunotherapy: A review. J. Cell. Physiol. 234 (6), 8509–8521. doi:10.1002/jcp.27782

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferris, R. L., Blumenschein, G., Fayette, J., Guigay, J., Colevas, A. D., Licitra, L., et al. (2016). Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N. Engl. J. Med. 375 (19), 1856–1867. doi:10.1056/NEJMoa1602252

PubMed Abstract | CrossRef Full Text | Google Scholar

Fridman, W. H., Zitvogel, L., Sautes-Fridman, C., and Kroemer, G. (2017). The immune contexture in cancer prognosis and treatment. Nat. Rev. Clin. Oncol. 14 (12), 717–734. doi:10.1038/nrclinonc.2017.101

PubMed Abstract | CrossRef Full Text | Google Scholar

Galon, J., and Bruni, D. (2019). Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 18 (3), 197–218. doi:10.1038/s41573-018-0007-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Gavrielatou, N., Doumas, S., Economopoulou, P., Foukas, P. G., and Psyrri, A. (2020). Biomarkers for immunotherapy response in head and neck cancer. Cancer Treat. Rev. 84, 101977. doi:10.1016/j.ctrv.2020.101977

PubMed Abstract | CrossRef Full Text | Google Scholar

Geeleher, P., Cox, N., and Huang, R. S. (2014). pRRophetic: an R package for prediction of clinical chemotherapeutic response from tumor gene expression levels. PLoS One 9 (9), e107468. doi:10.1371/journal.pone.0107468

PubMed Abstract | CrossRef Full Text | Google Scholar

Gutierrez-Pajares, J. L., Ben Hassen, C., Chevalier, S., and Frank, P. G. (2016). SR-BI: Linking cholesterol and lipoprotein metabolism with breast and prostate cancer. Front. Pharmacol. 7, 338. doi:10.3389/fphar.2016.00338

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, Q., Wang, R., Sun, C., Jin, X., Liu, D., Zhao, X., et al. (2014). Human beta-defensin-1 suppresses tumor migration and invasion and is an independent predictor for survival of oral squamous cell carcinoma patients. PLoS One 9 (3), e91867. doi:10.1371/journal.pone.0091867

PubMed Abstract | CrossRef Full Text | Google Scholar

Hänzelmann, S., Castelo, R., and Guinney, J. (2013). Gsva: Gene set variation analysis for microarray and RNA-seq data. BMC Bioinforma. 14, 7. doi:10.1186/1471-2105-14-7

CrossRef Full Text | Google Scholar

Huang, C., Chen, L., Savage, S. R., Eguez, R. V., Dou, Y., Li, Y., et al. (2021). Proteogenomic insights into the biology and treatment of HPV-negative head and neck squamous cell carcinoma. Cancer Cell 39 (3), 361–379. e316. doi:10.1016/j.ccell.2020.12.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Jew, B., Alvarez, M., Rahmani, E., Miao, Z., Ko, A., Garske, K. M., et al. (2020). Accurate estimation of cell composition in bulk expression through robust integration of single-cell information. Nat. Commun. 11 (1), 1971. doi:10.1038/s41467-020-15816-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Jia, L., Zhang, W., and Wang, C. Y. (2020). BMI1 inhibition eliminates residual cancer stem cells after PD1 blockade and activates antitumor immunity to prevent metastasis and relapse. Cell Stem Cell 27 (2), 238–253. e236. doi:10.1016/j.stem.2020.06.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Korbecki, J., Grochans, S., Gutowska, I., Barczak, K., and Baranowska-Bosiacka, I. (2020). CC chemokines in a tumor: A review of pro-cancer and anti-cancer properties of receptors CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10 ligands. Int. J. Mol. Sci. 21 (20), E7619. doi:10.3390/ijms21207619

PubMed Abstract | CrossRef Full Text | Google Scholar

Kurten, C. H. L., Kulkarni, A., Cillo, A. R., Santos, P. M., Roble, A. K., Onkar, S., et al. (2021). Investigating immune and non-immune cell interactions in head and neck tumors by single-cell RNA sequencing. Nat. Commun. 12 (1), 7338. doi:10.1038/s41467-021-27619-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Langfelder, P., and Horvath, S. (2008). Wgcna: an R package for weighted correlation network analysis. BMC Bioinforma. 9, 559. doi:10.1186/1471-2105-9-559

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, M., Wiedemann, T., Gross, C., Leinhauser, I., Roncaroli, F., Braren, R., et al. (2015). Targeting PI3K/mTOR signaling displays potent antitumor efficacy against nonfunctioning pituitary adenomas. Clin. Cancer Res. 21 (14), 3204–3215. doi:10.1158/1078-0432.CCR-15-0288

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, T., Feng, Z., Wang, Y., Zhang, H., Li, Q., Schiferle, E., et al. (2020). Antioncogenic effect of MicroRNA-206 on neck squamous cell carcinoma through inhibition of proliferation and promotion of apoptosis and autophagy. Hum. Gene Ther. 31 (23-24), 1260–1273. doi:10.1089/hum.2020.090

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, T., Qin, Y., Zhen, Z., Shen, H., Cong, T., Schiferle, E., et al. (2019). Long non-coding RNA HOTAIR/microRNA-206 sponge regulates STC2 and further influences cell biological functions in head and neck squamous cell carcinoma. Cell Prolif. 52 (5), e12651. doi:10.1111/cpr.12651

PubMed Abstract | CrossRef Full Text | Google Scholar

Mayakonda, A., Lin, D.-C., Assenov, Y., Plass, C., and Koeffler, H. P. (2018). Maftools: Efficient and comprehensive analysis of somatic variants in cancer. Genome Res. 28 (11), 1747–1756. doi:10.1101/gr.239244.118

PubMed Abstract | CrossRef Full Text | Google Scholar

Nathan, C. A., Khandelwal, A. R., Wolf, G. T., Rodrigo, J. P., Mäkitie, A. A., Saba, N. F., et al. (2022). TP53 mutations in head and neck cancer. Mol. Carcinog. 61 (4), 385–391. doi:10.1002/mc.23385

PubMed Abstract | CrossRef Full Text | Google Scholar

Newman, A. M., Liu, C. L., Green, M. R., Gentles, A. J., Feng, W., Xu, Y., et al. (2015). Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 12 (5), 453–457. doi:10.1038/nmeth.3337

PubMed Abstract | CrossRef Full Text | Google Scholar

Puram, S. V., Tirosh, I., Parikh, A. S., Patel, A. P., Yizhak, K., Gillespie, S., et al. (2017). Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell 171 (7), 1611–1624. doi:10.1016/j.cell.2017.10.044

PubMed Abstract | CrossRef Full Text | Google Scholar

Pylaeva, E., Ozel, I., Squire, A., Spyra, I., Wallner, C., Korek, M., et al. (2021). B-helper neutrophils in regional lymph nodes correlate with improved prognosis in patients with head and neck cancer. Cancers (Basel) 13 (12), 3092. doi:10.3390/cancers13123092

PubMed Abstract | CrossRef Full Text | Google Scholar

Qi, Z., Barrett, T., Parikh, A. S., Tirosh, I., and Puram, S. V. (2019). Single-cell sequencing and its applications in head and neck cancer. Oral Oncol. 99, 104441. doi:10.1016/j.oraloncology.2019.104441

PubMed Abstract | CrossRef Full Text | Google Scholar

Rizvi, N. A., Hellmann, M. D., Snyder, A., Kvistborg, P., Makarov, V., Havel, J. J., et al. (2015). Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Sci. (New York, N.Y.) 348 (6230), 124–128. doi:10.1126/science.aaa1348

PubMed Abstract | CrossRef Full Text | Google Scholar

Ruffell, B., and Coussens, L. M. (2015). Macrophages and therapeutic resistance in cancer. Cancer Cell 27 (4), 462–472. doi:10.1016/j.ccell.2015.02.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Sacco, A. G., Chen, R., Worden, F. P., Wong, D. J. L., Adkins, D., Swiecicki, P., et al. (2021). Pembrolizumab plus cetuximab in patients with recurrent or metastatic head and neck squamous cell carcinoma: An open-label, multi-arm, non-randomised, multicentre, phase 2 trial. Lancet. Oncol. 22 (6), 883–892. doi:10.1016/s1470-2045(21)00136-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Siegel, R. L., Miller, K. D., and Jemal, A. (2020). Cancer statistics, 2020. Ca. Cancer J. Clin. 70 (1), 7–30. doi:10.3322/caac.21590

PubMed Abstract | CrossRef Full Text | Google Scholar

Siu, L. L., Even, C., Mesia, R., Remenar, E., Daste, A., Delord, J. P., et al. (2019). Safety and efficacy of durvalumab with or without tremelimumab in patients with PD-L1-low/negative recurrent or metastatic HNSCC: The phase 2 CONDOR randomized clinical trial. JAMA Oncol. 5 (2), 195–203. doi:10.1001/jamaoncol.2018.4628

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, P., Li, W., Wu, X., Qian, Z., Ying, J., Gao, S., et al. (2022). Integrated analysis of single-cell and bulk RNA-sequencing identifies a signature based on B cell marker genes to predict prognosis and immunotherapy response in lung adenocarcinoma. Cancer Immunol. Immunother. 1, 1. doi:10.1007/s00262-022-03143-2

CrossRef Full Text | Google Scholar

Sun, C. Q., Arnold, R., Fernandez-Golarz, C., Parrish, A. B., Almekinder, T., He, J., et al. (2006). Human beta-defensin-1, a potential chromosome 8p tumor suppressor: Control of transcription and induction of apoptosis in renal cell carcinoma. Cancer Res. 66 (17), 8542–8549. doi:10.1158/0008-5472.CAN-06-0294

PubMed Abstract | CrossRef Full Text | Google Scholar

Textor, S., Fiegler, N., Arnold, A., Porgador, A., Hofmann, T. G., Cerwenka, A., et al. (2011). Human NK cells are alerted to induction of p53 in cancer cells by upregulation of the NKG2D ligands ULBP1 and ULBP2. Cancer Res. 71 (18), 5998–6009. doi:10.1158/0008-5472.CAN-10-3211

PubMed Abstract | CrossRef Full Text | Google Scholar

Vigneswaran, N., and Williams, M. D. (2014). Epidemiologic trends in head and neck cancer and aids in diagnosis. Oral Maxillofac. Surg. Clin. North Am. 26 (2), 123–141. doi:10.1016/j.coms.2014.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Waldhauer, I., and Steinle, A. (2006). Proteolytic release of soluble UL16-binding protein 2 from tumor cells. Cancer Res. 66 (5), 2520–2526. doi:10.1158/0008-5472.CAN-05-2520

PubMed Abstract | CrossRef Full Text | Google Scholar

Wichmann, G., Rosolowski, M., Krohn, K., Kreuz, M., Boehm, A., Reiche, A., et al. (2015). The role of HPV RNA transcription, immune response-related gene expression and disruptive TP53 mutations in diagnostic and prognostic profiling of head and neck cancer. Int. J. Cancer 137 (12), 2846–2857. doi:10.1002/ijc.29649

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, K., Fu, Y., Han, Y., Xia, R., Xu, S., Duan, S., et al. (2020). Fewer tumour-specific PD-1(+)CD8(+) TILs in high-risk "Infiltrating" HPV(-) HNSCC. Br. J. Cancer 123 (6), 932–941. doi:10.1038/s41416-020-0966-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamaguchi, K., Chikumi, H., Shimizu, A., Takata, M., Kinoshita, N., Hashimoto, K., et al. (2012). Diagnostic and prognostic impact of serum-soluble UL16-binding protein 2 in lung cancer patients. Cancer Sci. 103 (8), 1405–1413. doi:10.1111/j.1349-7006.2012.02330.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, F., Long, N., Anekpuritanang, T., Bottomly, D., Savage, J. C., Lee, T., et al. (2022). Identification and prioritization of myeloid malignancy germline variants in a large cohort of adult patients with AML. Blood 139 (8), 1208–1221. doi:10.1182/blood.2021011354

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoshihara, K., Shahmoradgoli, M., Martinez, E., Vegesna, R., Kim, H., Torres-Garcia, W., et al. (2013). Inferring tumour purity and stromal and immune cell admixture from expression data. Nat. Commun. 4, 2612. doi:10.1038/ncomms3612

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: CD8+ T cells, head and neck squamous cell carcinoma, immunotherapy, prognosis, weighted gene co-expression network analysis

Citation: Zhang S, Zhang W and Zhang J (2022) 8-Gene signature related to CD8+ T cell infiltration by integrating single-cell and bulk RNA-sequencing in head and neck squamous cell carcinoma. Front. Genet. 13:938611. doi: 10.3389/fgene.2022.938611

Received: 07 May 2022; Accepted: 04 July 2022;
Published: 22 July 2022.

Edited by:

Geng Chen, GeneCast Biotechnology Co., Ltd., China

Reviewed by:

Qingjia Chi, Wuhan University of Technology, China
Ting Li, National Center for Toxicological Research (FDA), United States

Copyright © 2022 Zhang, Zhang and Zhang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jian Zhang, zj301doctor@126.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.