Skip to main content

Survival-related indicators ALOX12B and SPRR1A are associated with DNA damage repair and tumor microenvironment status in HPV 16-negative head and neck squamous cell carcinoma patients

A Correction to this article was published on 11 July 2022

This article has been updated

Abstract

Objectives

To investigate prognostic-related gene signature based on DNA damage repair and tumor microenvironment statue in human papillomavirus 16 negative (HPV16-) head and neck squamous cell carcinoma (HNSCC).

Methods

For the RNA-sequence matrix in HPV16- HNSCC in the Cancer Genome Atlas (TCGA) cohort, the DNA damage response (DDR) and tumor microenvironment (TM) status of each patient sample was estimated by using the ssGSEA algorithm. Through bioinformatics analysis in DDR_high/TM_high (n = 311) and DDR_high/TM_low (n = 53) groups, a survival-related gene signature was selected in the TCGA cohort. Two independent external validation cohorts (GSE65858 (n = 210) and GSE41613 (n = 97)) with HPV16- HNSCC patients validated the gene signature. Correlations among the clinical-related hub differentially expressed genes (DEGs) and infiltrated immunocytes were explored with the TIMER2.0 server. Drug screening based on hub DEGs was performed using the CellMiner and GSCALite databases. The loss-of-function studies were used to evaluate the effect of screened survival-related gene on the motility of HPV- HNSCC cells in vitro.

Results

A high DDR level (P = 0.025) and low TM score (P = 0.012) were independent risk factors for HPV16- HNSCC. Downregulated expression of ALOX12B or SPRR1A was associated with poor survival rate and advanced cancer stages. The pathway enrichment analysis showed the DDR_high/TM_low samples were enriched in glycosphingolipid biosynthesis-lacto and neolacto series, glutathione metabolism, platinum drug resistance, and ferroptosis pathways, while the DDR_high/TM_low samples were enriched in Th17 cell differentiation, Neutrophil extracellular trap formation, PD − L1 expression and PD − 1 checkpoint pathway in cancer. Notably, the expression of ALOX12B and SPRR1A were negatively correlated with cancer-associated fibroblasts (CAFs) infiltration and CAFs downstream effectors. Sensitivity to specific chemotherapy regimens can be derived from gene expressions. In addition, ALOX12B and SPRR1A expression was associated with the mRNA expression of insulin like growth factor 1 receptor (IGF1R), AKT serine/threonine kinase 1 (AKT1), mammalian target of rapamycin (MTOR), and eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1) in HPV negative HNSCC. Down-regulation of ALOX12B promoted HPV- HNSCC cells migration and invasion in vitro.

Conclusions

ALOX12B and SPRR1A served as a gene signature for overall survival in HPV16- HNSCC patients, and correlated with the amount of infiltrated CAFs. The specific drug pattern was determined by the gene signature.

Peer Review reports

Introduction

Head and neck squamous cell carcinoma (HNSCC) are heterogeneous epithelial tumor that arise from the oropharynx, oral cavity, lip, larynx, nasopharynx, and hypopharynx. The 5-year overall survival rate for patients with HNSCC is approximately 50% [1]. The main causative risk factors include excessive tobacco usage, heavy alcohol usage, and human papillomavirus 16 (HPV16) infection [2]. HPV16 infection-related E6/E7 is generally considered the oncogenic protein associated with HNSCC [3]. HPV-negative (HPV-) HNSCC patients are characterized by distinct molecular landscapes, worse overall survival outcomes and a poor response rate to induction chemotherapy when compared with HPV-positive (HPV+) HNSCC patients [4,5,6]. Hence, there is an urgent need to investigate the molecular spectra of HPV16 negative (HPV16-) HNSCC and identify survival-related biomarkers.

Tumor cells respond to endogenous or exogenous DNA damage through the DNA damage response (DDR) pathways. Homologous recombination repair (HRR), nonhomologous end joining (NHEJ), mismatch repair (MMR), nucleotide excision repair (NER), and base excision repair (BER) are key pathways participating in the DNA damage response (DDR) [7]. A previous study developed a homologous recombination deficiency (HRD) score at transcriptome level to predict the prognosis of HNSCC patients, and the HRD score was not an independent indicator for prognosis in univariate survival analysis [8]. However, another study has shown that HNSCC patients exhibited double-strand breaks repair (DBR)- and MMR- related genes upregulated as compared with healthy subjects, and patients with low expression of NER-related genes showed prolonged progression-free survival under concurrent chemoradiotherapy treatment [9]. Abnormal DDR status is also an important biological factor leading to radiotherapy resistance [10]. Meanwhile, DDR status could affect expressions of Immune checkpoint protein, interferon receptors, and neoantigens, and thereby affect therapeutic effect of immunotherapy [11, 12]. Therefore, DDR level represents the tumorigenesis and development process, as well as reflecting the efficacy of anti-tumor therapy to a certain extent. Considering the higher DNA repair gene/protein expression levels in HPV+ HNSCC tissue samples than those in HPV- HNSCC samples [13], the prognostic value and therapeutic efficacy value of DDR levels in HPV16- HNSCC patients need to be further clarified. In addition to the DDR level reflecting the malignant evolution of tumor cells, the tumor microenvironment (TM) around tumor cells also affects or reflects the malignant behavior of tumor cells. The TM comprises a network of interactions among blood vessels, immune cells, fibroblasts, adipocyte, endothelial cells, tumor cells, and the surrounding extracellular components [14]. According to previous studies, TM is crucial for modulating tumor progress and evaluating anti-tumor treatment responses [15, 16]. The immune profiles of HPV+ HNSCC is distinct from that in HPV- HNSCC [17, 18]. Interesting, HPV+ patients exhibited enhanced immune cell infiltration compared with HPV- patients [18], which suggests that in the study of analysing the prognostic and efficacy prediction value of tumor microenvironment for HNSCC patients, the HPV infection status of the patients also needs to be considered.

Recently, DNA-targeted therapy combined with radiotherapy, chemotherapy or immunotherapy has shown synergistic anti-tumor effects [19,20,21,22]. An earlier study identified prognostic related immune genes based on differences in immune status between tumor tissue and normal tissue in HNSCC patients, which could help develop therapeutic regimens toward specific targets [23]. However, the DDR status differed between tumors and adjacent normal tissue, and immune status (eg, PD-L1 expression) in tumor tissue varies among patients. In the present study, we used the TCGA and GEO databases to explore and verify prognostic-related genes reflecting both the DDR and the TM status as well as exploring the drug sensitivity pattern in HPV16- HNSCC patients.

Materials and methods

Patient cohort and data source

The RNA sequencing data (fragments per kilobase per million (FPKM) and read counts) for HNSCC were downloaded from the Cancer Genome Atlas (TCGA) database (https://portal.gdc.cancer.gov). Patient clinical data were obtained from the Broad Genome Data Analysis Centers Firehose server (https://gdac.broadinstitute.org). Related survival data were downloaded from the UCSC Xena browser (https://xenabrowser.net). Based on the HPV status recorded in the Firehose server and available survival data in the Xena browser, 433 HPV16- HNSCC patients were included in the TCGA discovery cohort. The validation cohort included 210 HPV16- patients in the Gene Expression Omnibus (GEO, https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/geo) database (GSE65858 data set) and 97 HPV- patients in the GSE41613 data set. The data in this study were all obtained from open available databases, and data download process comply with the data use certification agreement of TCGA, Firehose, UCSC Xena and GEO databases. The requirements for institutional review board approval and informed consent were waived.

Collection of DDR-related gene sets and cluster TCGA samples at the DNA damage repair level

Five DDR-associated gene sets were collected from online Molecular Signatures Database (MSigDB) (https://gsea-msigdb.org). There were 55 genes in the HRR gene set (hsa03440, R-HSA-5693579), 13 in the NHEJ gene set (hsa03450), 23 in the MMR gene set (R-HSA-5358508, hsa03430), 111 in the NER gene set (hsa03420, R-HSA-5696398), and 98 in the BER gene set (hsa03410, R-HSA-73884). The single-sample Gene Set Enrichment Analysis (ssGSEA) algorithm (R package “GSVA”) was used to obtain a score for the DDR level in each HPV16- tumor sample (FPKM) of the TCGA discovery cohort. The whole cohort was then clustered into DDR_high and DDR_low groups via the “sparcl” package in R [24, 25]. In univariate survival analysis, the log-rank test was used to compare the difference in overall survival rate between the above two groups.

Tumor microenvironment status identification in the TCGA cohort

In the TCGA discovery cohort, the ESTIMATE algorithm (R package “ESTIMATE”) was used to calculate scores for immune and stromal cell infiltration in the transcriptome profiles (FPKM) [26]. The maximally selected rank statistics method (“maxstat” package in R) was applied to classify the cohort into tumor microenvironment high (TM_high) and tumor microenvironment low (TM_low) groups [27]. Among the two groups, the TM_high group contains more immune and stromal cell infiltration than that of the TM_low group. The prognostic value of the TM classification method was calculated using the Kaplan-Meier curve and log-rank tests. Combining DDR and TM status in TCGA discovery cohort can obtain four subgroups, namely, The DDR_high/TM_low, DDR_high/TM_high, DDR_low/TM_high, and DDR_low/TM_low groups. The DDR_high/TM_high and DDR_high/TM_low groups were extracted and integrated into the TCGA final discovery cohort. A multivariate survival analysis was conducted to evaluate the influence of the grouping factor on the overall survival rate for patients in the TCGA final discovery cohort.

Somatic alteration analysis of the DDR_high/TM_high and DDR_high/TM_low groups

Gene mutation data were obtained from the TCGA database (https://portal.gdc.cancer.gov). The “maftool” package in R was applied to visualize the top 20 mutant genes in DDR_high/TM_low and DDR_high/TM_high groups, respectively [28]. A forest plot showed the mutant genes that significantly differed between the above two groups (P < 0.05).

Extracted DDR- and TM-related hub genes and enrichment analysis of the hub genes in the TCGA final discovery cohort

For the TCGA final discovery cohort, the raw data (read counts) for tumor samples were standardized using the cpm function and genes with high expression remained (mean read counts per million was larger than one). Differentially expressed genes (DEGs) with FDR < 0.05 and log |fold change| > 0.5 between DDR_high/TM_high and DDR_high/TM_low groups were calculated with the “limma” package in R. A weighted gene co-expression network analysis (WGCNA) algorithm (“WGCNA” package in R) was used to select the DEGs modules correlated with the DDR_high/TM_high and DDR_high/TM_low groups [29]. In the WGCNA analyzing process, DEGs with variance > 50% among samples were selected. Using pearson’s correlation coefficient, paired genes were used to build a co-expression network. The co-expression network was transformed into an adjacency matrix by selecting the soft threshold (R2 > 0.8). Then, a topology overlap matrix (TOM) was established using the tomsimilarity function to calculate the degree of association of genes in the adjacency matrix. The distance matrix 1-TOM was used to construct a hierarchical cluster tree and identify the various modules via dynamic tree cut. Then, modules with optimal eigenvalue similarity values were extracted for further analysis. Finally, a plot was constructed to show the correlation between the extracted modules and the subgroups. Eigenvalue gene modules that were significantly correlated with subgrouping (P < 0.05) were extracted for further analysis. Hub genes were obtained by applying a protein–protein interaction (PPI) analysis to the eigenvalue gene modules (STRING (http://string-db.org)). For the nodule degree rank, the top 50 or 100 genes were defined as hub genes. Enrichment analysis of the hub genes in each eigenvalue gene module was performed with the “clusterProfiler” package in R [30], and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis in the “clusterProfiler” package is based on KEGG website (https://www.kegg.jp/) [31,32,33]. P < 0.05 was considered statistically significant.

Prognostic-related hub gene identification and validation

A univariate survival analysis (P < 0.2) was conducted to select the significant hub genes related to overall survival rate of patients in the TCGA final discovery cohort. Independent prognostic-related hub genes were identified by multivariate survival analysis. A correlation analysis between the prognostic-related hub genes and the clinical traits was performed. The prognostic-related hub genes were validated in the GSE65858 and GSE41613 data sets.

Therapeutic response prediction

Immunophenoscores (IPS) were calculated according to the four major immunogenicity categories, namely, effector cells, immunosuppressive cells, MHC molecules, and immunomodulators [34]. The Cancer Immunome Atlas (TCIA; https://tcia.at/home) webtool provided four indexes for each TCGA patient: 1, The IPS index, and a high IPS value showed increased immunogenicity; 2, The IPS-PD1/PD-L1/PD-L2 blocker index, and a high value means more sensitivity to PD1/PD-L1/PD-L2 antibodies; 3, The IPS-CTLA4 blocker index, and a high value means more sensitivity to CTLA4 antibodies; 4, The IPS-PD1/PD-L1/PD-L2 + CTLA4 blocker index, and a high value means more sensitivity to PD1/PD-L1/PD-L2 and CTLA4 antibodies. According to the expression of hub genes, samples were divided into hub gene high- and low- group by the maximally selected rank statistics method. IPS and derived indexes were downloaded, and the differences in those indexes between high- and low- groups were analysed. Except for surgery and radiotherapy, other important treatment options for HNSCC patients are chemotherapy and small-molecule targeted drugs. At the same time, we are gradually increasing our understanding of the rationality of concurrent chemotherapy and immunotherapy. The CellMiner (https://discover.nci.nih.gov/cellminer) and GSCALite (http://bioinfo.life.hust.edu.cn/web/GSCALite/) databases can provide correlations between specific genes and drug sensitivity in the NCI-60 cell line set and in the CTRP or GDSC databases, respectively. Accordingly, we analysed the associations between the hub genes and the drug response in the CellMiner, CTRP, and GDSC databases.

Correlations among prognostic-related hub genes, immune infiltration levels, and downstream immune cell effectors

The online webserver TIMER2.0 database (http://timer.comp-genomics.org) is a comprehensive resource providing gene-associated immune infiltration data across 32 cancer types. TIMER2.0 database can provide multiple algorithms such as xCell, CIBERSORT, EPIC, MCP-counter and TIMER algorithms. The relationships between prognostic-related hub genes and infiltrated immune cells were explored. A Spearman’s rank analysis was performed to analyse the correlations between the hub genes and the downstream immune cell effectors. The associate between the mRNA expression of the identified hub genes and the mRNA expression of insulin like growth factor 1 receptor (IGF1R), AKT serine/threonine kinase 1 (AKT1), mammalian target of rapamycin (MTOR), and eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1) for HPV- HNSCC were also explored in TIMER2.0 database.

Cell culture and transfection of RNA oligonucleotides

HPV- HNSCC cell lines (HN6, CAL27) were cultured in DMEM with 5% FBS (Gibco, USA) and maintained in a humidified 5% CO2 environment at 37 °C. Total RNA was extracted using RNAiso Plus (Takara 9109, Japan). Quantitative Real-time PCR (qRT-PCR) was performed with ABI Real-Time PCR System (ABI 7500, Thermofisher CA). Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used to normalize the mRNA expression of test gene, and the ∆∆ Ct method was conducted to calculate the relative expression levels of test gene. Test gene primers used in qRT-PCR were listed as follows: ALOX12B, forward, 5′-TCTCACTGACCATTGTGGGGA-3′; ALOX12B, reverse, 5′-TTGTGCAGGCGGATGATGATG-3′. The small interfering RNA (siRNA) was chemically synthesized by Geneseed (Guangzhou, China). HPV- HNSCC cell lines was transfected with siRNA using lipofectamine™ 2000 (Invitrogen, USA). The siRNA-mediated knockdown of ALOX12B was achieved by targeting the sequence 5′-CGCTATGCGGAGTTCTACA-3′.

Cell proliferation, invasion assays and metastasis assays

The CCK8 assays (YEASEN, Shanghai) was used to assess cell proliferation. HN6 cells were seed into 96-well plates with 1 × 103/well and Cell Counting Kit-8 solution (YEASEN, Shanghai) was added to each well. Subsequently, the cells counting was performed daily for 4 days. The matrigel-coated transwell assay and transwell migration assay were used to test invasion and migration ability, respectively. In the matrigel-coated transwell assay, HN6 cells (1 × 105/well) were seed into 6-well transwell plates (COSTAR, USA) that precoated Matrigel solution (BD Biosciences, USA), and the 6-well plates was incubated in a humidified incubator with 5% CO2 at 37 °C for 24 h. In the transwell migration assay, HN6 cells (1 × 105/well) were seed into 6-well transwell plates (COSTAR, USA), and tested after 24 h. The migrated or invaded cells were fixed and stained with crystal violet and counted using ImageJ software.

Statistical analysis

All data were processed in R v. 3.6.1 (http://www.R-project.org) and GraphPad Prism 8 software (GraphPad Software, Inc., USA). P < 0.05 was considered statistically significant in both the multivariate survival analysis and correlation analysis.

Results

DDR-related patients clustering in the TCGA discovery cohort

A total of 433 HPV16- HNSCC patients were included in the TCGA discovery cohort (Table S1). Based on the DDR-related gene sets (Table S2), the ssGSEA method categorized patients into the DDR_high group (Cluster 1; 364 patients) and the DDR_low group (Cluster 2; 69 patients) (Fig. 1A). The log-rank test showed a difference in survival between the two groups (P = 0.025) (Fig. 1B).

Fig. 1
figure 1

Patient sample stratification based on DNA damage response (DDR) level in human papillomavirus 16 negative (HPV16-) head and neck squamous cell carcinoma (HNSCC) TCGA discovery cohort (n = 433). A ssGSEA matrix plot of two subtypes identified in HPV16- HNSCC TCGA discovery cohort according to five DDR-associated genesets. B Kaplan-Meier (K-M) plot of overall survival probability for patients in the above two subtypes. One patient belonging to DDR_high subtype lacked survival data as shown in risk table

TM- and DDR- status classification in the TCGA discovery cohort

In the TCGA discovery cohort, we divided patients into two cluster through ESTIMATE scoring and the maximally selected rank statistics method. The cut-off value for the maximally selected rank statistics algorithm was − 951.85 (Fig. 2A). A univariate survival analysis showed a significant difference in survival between TM_high (n = 380) and TM_low (n = 53) groups (P = 0.012; Fig. 2B). After integrating the DDR-related clustering, there were 53, 311, 69, and 0 patients in the DDR_high/TM_low, DDR_high/TM_high, DDR_low/TM_high and DDR_low/TM_low groups, respectively. We performed a survival log-rank test among DDR_high/TM_low, DDR_high/TM_high, and DDR_low/TM_high groups and found that the DDR_low/TM_high group showed the best overall survival rate (P = 0.0072; Fig. 2C). We compared the survival rates of the DDR_high/TM_high and DDR_high/TM_low groups and observed that the former exhibited higher survival than the latter (P = 0.032; Fig. 2D). Then, we integrated the data for the DDR_high/TM_low (n = 53) and DDR_high/TM_high (n = 311) groups to create the TCGA final discovery cohort (n = 364).

Fig. 2
figure 2

The Kaplan–Meier (K-M) overall survival (OS) curve of patients in the TCGA final discovery cohort (n = 364). A Two clusters were obtained from TCGA discovery cohort (n = 433) by dichotomizing tumor microenvironment (TM) score (“ESTIMATE” package in R). B K-M plot of OS probability in high- and low- TM score group. C K-M plot of OS probability in DDR_high/TM_low, DDR_high/TM_high, and DDR_low/TM_high groups. D K-M plot of OS probability for patients in the TCGA final discovery cohort including only DDR_high/TM_high and DDR_high/TM_low groups. One patient in the TCGA final discovery cohort lacked survival data as mentioned in Fig. 1

The clinical characteristics of the DDR_high/TM_low and DDR_high/TM_high groups were shown in Table S3. The multivariate survival analysis disclosed that the DDR_high/TM_low status was a risk factor for overall survival (OS) in HPV16- HNSCC (Table 1). As shown in Table S3, N stage, T stage, TNM stage, alcohol history, smoking history, lymphovascular invasion status, margin status, perineural invasion status, pathological nodal extracapsular spread status, anatomic neoplasm subdivision, neoadjuvant treatment, radiation therapy, additional pharmaceutical therapy, and additional radiation therapy were all balanced between the DDR_high/TM_low and DDR_high/TM_high groups. However, two groups differed in terms of sex and age.

Table 1 Multivariate survival analysis in the TCGA final discovery cohort

Somatic mutant gene distinction between DDR_high/TM_low and DDR_high/TM_high groups in the TCGA final discovery cohort

We analysed the top 20 genes with the highest mutation frequency in the TCGA final discovery cohort. In the DDR_high/TM_low group, they were TP53, TTN, NSD1, CDKN2A, and PKHD1L1 (Fig. 3A). In the DDR_high/TM_high group, they were TP53, TTN, FAT1, CSMD3, and MUC16 (Fig. 3B). NSD1, CSMD2, ERBB4, ITGA4, CUL3, and TP53 showed relatively a higher mutation rate in the DDR_high/TM_low group, whereas CASP8 showed a relatively higher mutation frequency in the DDR_high/TM_high group (Fig. 3C).

Fig. 3
figure 3

Somatic mutant genes in DDR_high/TM_low and DDR_high/TM_high groups of the TCGA final discovery cohort. A Mutant genes (top 20) in DDR_high/TM_low group. B Mutant genes (top 20) in DDR_high/TM_high group. C Differential mutant genes between DDR_high/TM_low and DDR_high/TM_high groups

TM- and DDR- related hub DEGs identification and functional enrichment analysis in the TCGA final discovery cohort

We obtained 2140 DEGs in the TCGA final discovery cohort. Of these, 1589 were downregulated and 551 were upregulated in the DDR_high/TM_low group, when compared with the DDR_high/TM_high group. The heatmap shows clustering of the top 50 upregulated and top 50 downregulated genes in the DDR_high/TM_low group (Fig. 4A). During the WGCNA calculation, the corresponding soft threshold was three and R2 > 0.8 (Fig. 4B). Modules with eigenvalue similarity > 0.75 were merged for further analysis (Fig. 4C). Then, the WGCNA algorithm identified seven DEGs modules designated blue, green, red, pink, black, brown, and gray (Fig. 4D). For the PPI analysis, we calculated gene nodes in the black, brown, blue, green, and red gene modules, respectively. After ranking the connecting node numbers between genes in each module, we screened out the top 50 hub genes in the black and brown modules (Fig. 4E, F). Similarly, we screened out the top 100 hub genes in the blue, green, and red modules, respectively (Fig. S1).

Fig. 4
figure 4

Identification of DDR- and TM- related hub genes in the TCGA final discovery cohort. A Heatmap of clustered top 100 differentially expressed genes (DEGs) between DDR_high/TM_high and DDR_high/TM_low groups. B-D WGCNA algorithm screened out seven eigengenes module (blue, green, red, pink, black, brown, and grey) based on DDR and TM status. E-F The top 50 hub genes selected in black and brown modules by protein-to-protein network method; respectively

For the black gene module, the top 50 hub genes were enriched in ten biological process (BP) items, ten molecular function (MF) items, ten cellular component (CC) items, and one KEGG pathway (Fig. 5A, B). For the brown module, the top 50 hub genes were enriched in ten biological process (BP) items, ten molecular function (MF) items, one cellular component (CC) item, and 24 KEGG pathways (Fig. 5C, D). In the BP analysis of the black module, the hub genes were enriched in keratinocyte differentiation, epidermal cell differentiation, and keratinization. In the BP analysis of the brown module, the hub genes were enriched in xenobiotic stimulus, glutathione metabolic process, and cellular detoxification. The involved KEGG pathways in the black module were glycosphingolipid biosynthesis-lacto and neolacto series. The KEGG pathways involved in the brown module were metabolism of glutathione metabolism, drug metabolism−cytochrome P450, platinum drug resistance, and ferroptosis. For the blue gene module, the top 100 hub genes were enriched in several KEGG pathway (Fig. 6A), such as, Natural killer cell mediated cytotoxicity, Th17 cell differentiation, Neutrophil extracellular trap formation, PD − L1 expression and PD − 1 checkpoint pathway in cancer, and Leukocyte trans-endothelial migration pathways. For the green module, the top 100 hub genes were enriched in 30 KEGG pathways (Fig. 6B), such as, PI3K − Akt signaling pathway, Focal adhesion, MAPK signaling pathway, and TGF − beta signaling pathways. For the green module, the top 100 hub genes were enriched in 31 KEGG pathways (Fig. 6C), such as, Herpes simplex virus 1 infection, Epstein−Barr virus infection, Human papillomavirus infection, and Viral carcinogenesis pathways.

Fig. 5
figure 5

Biological characteristics of hub genes in black and brown modules. A Biological process, molecular function, and cellular component terms enriched by hub genes in black module. B KEGG pathway enriched by hub genes in black module. C Biological process, molecular function, and cellular component terms enriched by hub genes in brown module. D KEGG pathways enriched by hub genes in brown module

Fig. 6
figure 6

KEGG pathways analysis of hub genes in blue, green, and red modules. A KEGG pathways enriched by top 100 hub genes in blue module. B KEGG pathways enriched by top 100 hub genes in green module. C KEGG pathways enriched by top 100 hub genes in red module

Prognostic value of the hub genes in the TCGA final discovery and GEO validation data sets

Among the 100 hub genes in the black and brown modules, 28 genes were correlated with OS in HPV16- HNSCC (Table 2). The multivariate survival analysis identified ALOX12B and SPRR1A as protective survival-related genes (Table 3). Geographical validations were performed on the GSE65858 and GSE41613 data sets. Multivariate survival analysis revealed that SPRR1A was significantly correlated with better OS in both GEO cohorts, whereas ALOX12B was an independent predictor of survival in patients at GSE65858 data set.

Table 2 Univariate survival analysis for the prognostic value of hub genes in the TCGA final discovery cohort
Table 3 Multivariate survival analysis for the prognostic value of SPRR1A and ALOX12B in the TCGA final discovery cohort and GEO validation data sets

Correlation analysis demonstrated that ALOX12B was upregulated in N0–1 stage as compared with N2–3 stage in the TCGA final discovery cohort (P = 0.028). Elevated ALOX12B expression was also observed in the earlier cancer stage in the GSE65858 and GSE41613 data sets (P = 0.024 and P = 0.028, respectively). SPRR1A downregulation was correlated with advanced N stage and cancer stage in the TCGA final discovery cohort (P = 0.0016) and GSE41613 (P = 0.038), respectively (Fig. 7).

Fig. 7
figure 7

Wilcoxon rank test of prognostic-related hub gene expression between early and advanced TNM stage groups. A-C Correlation between the expression of ALOX12B and N stage in the TCGA final discovery cohort and GSE65858 data set, respectively. Association between the expression of ALOX12B and TNM stage in GSE41613 data set. D-F Correlation between the expression of SPRR1A and N stage in the TCGA final discovery cohort and GSE65858 data set, respectively. Association between the expression of SPRR1A and TNM stage in GSE41613 data set

Drug sensitivity analysis based on prognostic gene expression levels

Increased IPS and IPS-CTLA4 blocker indexes were observed in the groups with high expression of ALOX12B (P = 0.0018, P = 0.018) or SPRR1A (P = 0.0014, P = 0.063) (Fig. 8A–H). The IPS-PD1/PD-L1/PD-L2 blocker and IPS-PD1/PD-L1/PD-L2 + CTLA4 blocker indices did not markedly differ between the two groups. Subsequently, we explored anti-tumor drug sensitivity based on hub genes. Poor survival patients with downregulated SPRR1A and ALOX12B were sensitive to cisplatin, rapamycin, Idelalisib, everolimus (Fig. 8I), and were resistant to lapatinib, and afatinib, fluorouracil, and PHA-793887(Fig. S2, Fig. S3). We also observed that CSTA downregulation was an indicative of docetaxel resistance (Fig. S2, Fig. S3).

Fig. 8
figure 8

Immunogenicity analysis based on the expression of prognostic-related hub genes. Differences of IPS, IPS-PD1/PD-L1/PD-L2 blocker, IPS-CTLA4 blocker, and IPS-PD1/PD-L1/PD-L2 + CTLA4 blocker between the groups with high and low expression of ALOX12B (A-D) and SPRR1A (E-H), respectively. I Drug patterns based on ALOX12B and SPRR1A in CellMiner database, and panels were arranged according to correlation coefficient

Correlation between survival-related hub DEGs and immune/stromal cell infiltration determined in TIMER2.0

In the HPV- HNSCC TCGA database (TIMER2.0 online database), the xCell, EPIC, MCP-counter, and TIDE algorithms showed that downregulation of both hub genes (ALOX12B and SPRR1A) were correlated with increased cancer-associated fibroblasts (CAFs) (Table 4). The downstream effector of CAFs contained FAP, IGF1/2, PDGFs, IL6, TGFβ, LIF, NT5E, ADORA2B, CCL2/5, CXCL12, and CXCR4 [35, 36]. Table 5 shows that nearly all the foregoing effectors were negatively associated with the expression of ALOX12B and SPRR1A. After adjusting tumor purity, the expression of ALOX12B was negatively associated with IGFR1, AKT1, MTOR, and EIF4EBP1, and the same correlation could also be observed in SPRR1A (Fig. 9).

Table 4 The correlation analysis between the expression of SPRR1A and ALOX12B and the amount of infiltrated cancer associated fibroblasts
Table 5 The correlation analysis between the expression of SPRR1A and ALOX12B and the expression of cancer associated fibroblasts effector
Fig. 9
figure 9

Association between the mRNA expression of ALOX12B (A-D) or SPRR1A (E-H) and the expression of insulin like growth factor 1 receptor (IGF1R), AKT serine/threonine kinase 1 (AKT1), mammalian target of rapamycin (MTOR), and eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1) in HPV- HNSCC samples at TIMER2.0 online database

ALOX12B suppressed invasion and migration of HPV- HNSCC cell

The mRNA expression of ALOX12B was tested in two HPV- HNSCC cell lines, and the basal expression level of ALOX12B was high in HN6 cell line (Fig. 10A). Therefore, we knock down ALOX12B in HN6 cell line by siRNA, and the expression of ALOX12B was found to be suppressed using qRT-PCR (P < 0.05) (Fig. 10B). Knockdown of ALOX12B promoted HN6 cell proliferation in the CCK8 cell viability assay (P < 0.05) (Fig. 10C). Besides, transwell assays demonstrated that the knockdown of ALOX12B increased the migratory (P < 0.01) and invasive (P < 0.05) ability of HN6 cell (Fig. 10D-E).

Fig. 10
figure 10

ALOX12B suppresses the migration and metastasis of HN6 cells in vitro. A Quantification mRNA expression of ALOX12B in HN6 and CAL27 cell lines by qRT-PCR. B HN6 cells were transfected with siNC and siALOX12B, and the expression of ALOX12B was examined using qRT-PCR. C CCK8 assay was conducted to assess cell viability in HN6-siNC and HN6-siALOX12B cells. D-E Transwell assays were performed to examine the invasion and migration of HN6-siNC and HN6-siALOX12B cells. Column, mean; Error bars, S.D.; *p < 0.05; **p < 0.01; ALOX12B, Arachidonate 12-Lipoxygenase, 12R Type; qRT-PCR, quantitative real-time polymerase chain reaction

Discussion

Based on the transcriptional expression profiles of HPV16-HNSCC patients, we found that high DNA damage levels or low tumor microenvironment scores were associated with poor prognosis in HPV16- HNSCC patients. In addition, there were significant differences in the enriched core signaling pathways between the DDR_high/TM_high group and the DDR_high/TM_low group, and the DDR_high/TM_high group was enriched in immune cell development, polarization and activity-related signaling pathway, while the DDR_high/TM_low group was enriched in glutathione metabolism, drug metabolizing enzymes, platinum resistance, and ferroptosis. In multivariate survival analysis, we identified ALOX12B and SPRR1A as two protective survival-related genes in HPV16- HNSCC and found that the expression of the above two genes were negatively correlated with CAFs infiltration. We also showed the mRNA expression of ALOX12B and SPRR1A was negatively correlated with the mRNA expression of IGF1R, AKT1, MTOR, and EIF4EBP1 in HPV- HNSCC. In addition, downregulation the expression of ALOX12B promoted the invasion and migration ability of HPV- HNSCC cell.

WGCNA clusters genes with highly similar biological functions into a single module [37]. In the DDR_high/TM_low group, the positively correlated black and brown modules were enriched in several pathways. In the black module, the glycosphingolipid biosynthesis-lacto and neolacto series pathway was upregulated. A recent study revealed that an increased neolacto-series glycosphingolipid on the membrane of tumor cells hinders the interaction between HLA-I and CD8+ T cells, and then impedes CD8 + T cell activation [38]. In the brown module, glutathione metabolism and drug metabolism-cytochrome P450 pathways were enriched. Previously studies reported that cancer stem cells can scavenge intracellular reactive oxygen species (ROS) through glutathione or resistant to treatment through abnormal drug metabolizing enzyme pathways [39,40,41]. We also noticed the cancer stem cell gene signature SOX2 and EPCAM was clustered in the brown module, moreover, cancer stem cell has been reported exhibiting resistance to immunotherapy [42,43,44]. Besides, ferroptosis pathway that enriched in brown module was disturbed during epithelial-to-mesenchymal transition (EMT) process [45]. In the DDR_high/TM_high group, the positively correlated blue module was enriched the neutrophil extracellular traps (NETs) pathway and PD − L1 expression and PD − 1 checkpoint pathway in cancer. We note that a study has shown that the barrier formed by NETs impedes the cell-to-cell contact between tumor cells and CD8+ T cells, and then hinders the antitumor activity of cytotoxic T cells, and simultaneously targeting PD-1 and NETs can increase tumor regression in vivo [46]. The mutation of a single gene can reflect the immune status to a certain degree. An example is the MUC16 mutation status for gastric cancer [47]. Our somatic mutation analysis showed an increased NSD1 mutation frequency in the DDR_high/TM_low group and a high CASP8 mutation rate in the DDR_high/TM_high group. The results were consistent with previous reports that NSD1 mutation is an intrinsic feature of cold immune phenotype, and the frequency of CASP8 mutation is increased in HNSCC with hot immune phenotype [48, 49].

SPRR1A and ALOX12B downregulation was observed in HPV16- HNSCC patients at advanced cancer stage. SPRR1A expression was positively associated with favorable survival and lower lymph node metastasis in HNSCC patients [50]. ALOXE3 as a paralog of ALOX12B, inhibits glioblastoma tumor migration [51]. Our results revealed that ALOX12B is an independent protective prognostic indicator in HPV16- HNSCC patients. However, another study revealed that ALOX12B mediates cervical cancer cell proliferation and migration via the PI3K-ERK1 pathway [52]. We also noticed that both hub genes (SPRR1A and ALOX12B) participate in epidermal cell differentiation and the skin barrier. ALOX12B participates in constructing the mature corneocyte lipid envelope [53]. SPRR1A is a structural component of the epidermis [54]. Mutation of ALOX12B and/or SPRR1A may result in skin barrier-related diseases, such as psoriasis and autosomal-recessive exfoliative ichthyosis [55]. ZNF750 and GLIS1 that regulating ALOX12B and SPRR1A expression belong to Zinc-finger proteins (ZNFs) [55]. ZNF750 is recognized as a tumor suppressor gene in HNSCC, and ALOX12B downregulation may indirectly reflect a loss of ZNFs expression in advanced HNSCC [56, 57]. A single-cell transcriptomic research including HPV16- HNSCC samples assessed the gene signatures of epithelial differentiation. SPRR1A and ZNF750 were listed in the top 50 genes of epithelial differentiation characteristic genes [58]. A low epithelial differentiation score was negatively correlated with a high partial epithelial-to-mesenchymal transition (p-EMT) score in the mesenchymal and basal subtype. The proportion of mesenchymal and basal subtypes increased with the elevating of p-EMT score. Patients classified as mesenchymal and basal subtypes showed poor survival in the whole TCGA cohort. Hence, a low epithelial score could serve as a surrogate for worse survival in HPV16- HNSCC patients.

The xCell and MCP-counter algorithms used the gene marker-based method while EPIC was based on the deconvolution approach [59]. All four algorithms showed that the down-regulation of ALOX12B and SPRR1A genes was correlated with increased CAFs infiltration in HPV16- HNSCC. A study demonstrated SPRR1A downregulation in MCF-10A breast cancer cells when coculture with CAFs [60]. CAFs promote tumor progression via promoting EMT and metabolic reprogramming [61]. These vicious behaviors are achieved via the expression of specific membrane proteins and paracrine cytokines or chemokines, such as FAP, IL6, TGFβ, LIF, NT5E, ADORA2B, CCL2/5, CXCL12, CXCR4, IL7, IGF1/2 [35, 36]. Our research indicated that the ALOX12B and SPRR1A expression levels were nearly always negatively correlated with the above effectors. Thus, both genes potentially reflect infiltrated CAFs quantity and quality. However, another study identified ALOX12B as an immunosuppressive factor based on a cytolytic activity analysis [49]. Therefore, the integrated roles of ALOX12B in the tumor microenvironment (CD8+ T cell, CAFs, regulatory T cells, myeloid-derived suppressor cells and so on) merit further investigation.

A docetaxel plus cisplatin and fluorouracil (TPF) chemotherapy regimen is recommended as the induction therapy for patients with stage III-IV in head and neck cancer [62]. In the present study, we observed that the downregulation of SPRR1A was associated with high sensitivity of cisplatin, and upregulation of ALOX12B and CSTA was associated with high sensitivity of fluorouracil, and docetaxel, respectively. LUX-Head & Neck 1 trial showed the efficacy of afatinib as a second-line treatment for recurrence or metastatic (RM) HNSCC patients [63]. We found that SPRR1A and CSTA expression were correlated with afatinib sensitivity. A phase II trial reported the efficiency of lapatinib and capecitabine therapy against RM HNSCC [64], and our results showed an association between SPRR1A and CSTA expression with lapatinib response. We also found that high ALOX12B expression was sensitive to CTLA4 inhibitors by calculating IPS-CTLA4 blocker scores. Notably, our research showed the expression of ALOX12B was negatively correlated with CAFs infiltration, and decreased ALOX12B expression indicated a better response to rapamycin or everolimus (drug target for mTOR). We try to explain the result in this way: The proliferation and development of CAFs are regulated by PI3K/AKT/mTOR signaling pathway, and CAFs might promote tumor progress via IGF1R/AKT1/mTOR pathway in tumor microenvironment [65, 66]. Moreover, for patients with head and neck squamous cell carcinoma, the level of phosphorylated mTOR in the junction zone between tumor and normal tissue or in tumor area was higher than that in the normal mucosal tissue, and the level of phosphorylated mTOR in the junction zone was higher than that in tumor area [67]. Meanwhile, upregulated mTOR expression predicted poor overall survival in HPV16- HNSCC patients [68]. The combination of everolimus plus docetaxel represented greater tumor regression than the use of docetaxel alone in a nude mouse xenograft model [69]. Therefore, poor prognosis patients with low ALOX12B expression had high infiltration of CAFs surrounding tumor cells, and rapamycin or everolimus may provide survival benefits by inhibiting mTOR signaling.

Our study has several limitations: 1, The clinical value of ALOX12B and SPRR1A was only validated in one GEO data set. Considering the different anatomical sites in HNSCC have different transcriptome profiles [70], TCGA and GSE65858 data set contained mixed HNSCC samples, while the GSE41613 data set only included oral squamous cell carcinoma samples. Thus, the difference in the anatomy of the two external verification data sets may be the reason for the different verification results, and further prospective research is required to verify the prognostic and drug sensitivity value of ALOX12B and SPRR1A in HPV16- HNSCC; 2, The specific mechanism of the abnormal expression of ALOX12B affecting the invasion and metastasis of HPV-HNSCC cell needs to be revealed by further experiments in vitro and in vivo; 3, The association between hub genes and drug sensitivity in CellMiner platform was based on NCI-60 tumor cells, while the cell assay lacks HNSCC cells. Besides, a zero-inflation data was used to analyse the correlation of the expression of SPRR1A and cisplatin sensitivity, and the reliability of this association needs to be further verified. Thus, the identified drug pattern based on hub genes needs to be validated in a patient-derived xenograft model.

Conclusion

Our bioinformatics analysis indicated that the intrinsic DNA repair level and tumor microenvironment status were associated with prognosis in HPV16- HNSCC patients. We identified two hub genes ALOX12B and SPRR1A, and showed that they can predict the clinical outcomes of HPV16- HNSCC. In addition, the two genes may be indicators of the amount of infiltrated CAFs. Nevertheless, further clinical research is required to validate drug sensitivity based on the expression of the those genes.

Availability of data and materials

The data presented in this study are included in the article/Supplementary Material, further inquiries are available from the corresponding author on reasonable request.

Change history

Abbreviations

AKT1:

AKT serine/threonine kinase 1

BER:

Base excision repair

CAFs:

Cancer-associated fibroblasts

CLE:

Corneocyte lipid envelope

CTRP:

Cancer Therapeutics Response Portal

DDR:

DNA damage response

DEG:

Differentially expressed gene

EGFR:

Epidermal growth factor receptor

EIF4EBP1:

Eukaryotic translation initiation factor 4E binding protein 1

EMT:

Epithelial-to-mesenchymal transition

FDR:

False discovery rate

FPKM:

Fragments per kilobase per million reads

GDSC:

Genomics of Drug Sensitivity in Cancer

GEO:

Gene Expression Omnibus

HNSCC:

Head and neck squamous cell carcinoma

HPV:

Human papillomavirus

HRR:

Homologous recombination repair

IGF:

Insulin-like growth factor

IGF1R:

Insulin like growth factor 1 receptor

IPS:

Immunophenoscore

KEGG:

Kyoto Encyclopedia of Genes and Genomes

KM:

Kaplan-Meier

MHC:

Major histocompatibility complex

MMR:

Mismatch repair

MSigDB:

Molecular Signatures database

MTOR:

Mammalian target of rapamycin

NER:

Nucleotide excision repair

NETs:

Neutrophil extracellular traps

NHEJ:

Nonhomologous end joining

OS:

Overall survival

p-EMT:

Partial epithelial-to-mesenchymal transition

PPI:

Protein-protein interaction

RM:

Recurrence/metastasis

TCGA:

The Cancer Genome Atlas

TCIA:

The Cancer Immunome Atlas

TM:

Tumor microenvironment

TNM:

Tumor, node and metastasis

TOM:

Topology overlap matrix

WGCNA:

Weighted gene co-expression network analysis

ZNF:

Zinc finger protein

References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.

    Article  PubMed  Google Scholar 

  2. Du E, Mazul AL, Farquhar D, Brennan P, Anantharaman D, Abedi-Ardekani B, et al. Long-term survival in head and neck Cancer: impact of site, stage, smoking, and human papillomavirus status. Laryngoscope. 2019;129(11):2506–13.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Rampias T, Sasaki C, Weinberger P, Psyrri A. E6 and e7 gene silencing and transformed phenotype of human papillomavirus 16-positive oropharyngeal cancer cells. J Natl Cancer Inst. 2009;101(6):412–23.

    Article  CAS  PubMed  Google Scholar 

  4. Network TCGA. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 2015;517(7536):576–82.

    Article  CAS  Google Scholar 

  5. Ang KK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen-Tân PF, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010;363(1):24–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Fakhry C, Westra WH, Li S, Cmelak A, Ridge JA, Pinto H, et al. Improved survival of patients with human papillomavirus-positive head and neck squamous cell carcinoma in a prospective clinical trial. J Natl Cancer Inst. 2008;100(4):261–9.

    Article  CAS  PubMed  Google Scholar 

  7. Gourley C, Balmaña J, Ledermann JA, Serra V, Dent R, Loibl S, et al. Moving from poly (ADP-ribose) polymerase inhibition to targeting DNA repair and DNA damage response in cancer therapy. J Clin Oncol. 2019;37(25):2257–69.

    Article  CAS  PubMed  Google Scholar 

  8. Knijnenburg TA, Wang L, Zimmermann MT, Chambwe N, Gao GF, Cherniack AD, et al. Genomic and molecular landscape of DNA damage repair deficiency across the cancer genome atlas. Cell reports. 2018;23(1):239–254.e236.

    Article  CAS  PubMed  Google Scholar 

  9. Psyrri A, Gkotzamanidou M, Papaxoinis G, Krikoni L, Economopoulou P, Kotsantis I, et al. The DNA damage response network in the treatment of head and neck squamous cell carcinoma. ESMO Open. 2021;6(2):100075.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Huang RX, Zhou PK. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct Target Ther. 2020;5(1):60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chabanon RM, Rouanne M, Lord CJ, Soria JC, Pasero P, Postel-Vinay S. Targeting the DNA damage response in immuno-oncology: developments and opportunities. Nat Rev Cancer. 2021;21(11):701–17.

    Article  CAS  PubMed  Google Scholar 

  12. Zhang T, Zheng S, Liu Y, Li X, Wu J, Sun Y, et al. DNA damage response and PD-1/PD-L1 pathway in ovarian cancer. DNA Repair. 2021;102:103112.

    Article  CAS  PubMed  Google Scholar 

  13. Holcomb AJ, Brown L, Tawfik O, Madan R, Shnayder Y, Thomas SM, et al. DNA repair gene expression is increased in HPV positive head and neck squamous cell carcinomas. Virology. 2020;548:174–81.

    Article  CAS  PubMed  Google Scholar 

  14. Hirata E, Sahai E. Tumor microenvironment and differential responses to therapy. Cold Spring Harbor Perspect Med. 2017;7(7):a026781.

  15. Hinshaw DC, Shevde LA. The tumor microenvironment innately modulates Cancer progression. Cancer Res. 2019;79(18):4557–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wu T, Dai Y. Tumor microenvironment and therapeutic response. Cancer Lett. 2017;387:61–8.

    Article  CAS  PubMed  Google Scholar 

  17. Mandal R, Şenbabaoğlu Y, Desrichard A, Havel JJ, Dalin MG, Riaz N, et al. The head and neck cancer immune landscape and its immunotherapeutic implications. JCI Insight. 2016;1(17):e89829.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Canning M, Guo G, Yu M, Myint C, Groves MW, Byrd JK, et al. Heterogeneity of the head and neck squamous cell carcinoma immune landscape and its impact on immunotherapy. Front Cell Dev Biol. 2019;7:52.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Sato H, Jeggo PA, Shibata A. Regulation of programmed death-ligand 1 expression in response to DNA damage in cancer cells: implications for precision medicine. Cancer Sci. 2019;110(11):3415–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bever KM, Le DT. DNA repair defects and implications for immunotherapy. J Clin Invest. 2018;128(10):4236–42.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Molkentine JM, Molkentine DP, Bridges KA, Xie T, Yang L, Sheth A, et al. Targeting DNA damage response in head and neck cancers through abrogation of cell cycle checkpoints. Int J Radiat Biol. 2021;97(8):1121–8.

    Article  CAS  PubMed  Google Scholar 

  22. Vitti ET, Kacperek A, Parsons JL. Targeting DNA double-strand break repair enhances radiosensitivity of HPV-positive and HPV-negative head and neck squamous cell carcinoma to photons and protons. Cancers. 2020;12(6):1490.

  23. Chen Y, Li ZY, Zhou GQ, Sun Y. An immune-related gene prognostic index for head and neck squamous cell carcinoma. Clin Cancer Res. 2021;27(1):330–41.

    Article  PubMed  Google Scholar 

  24. Witten DM, Tibshirani R. A framework for feature selection in clustering. J Am Stat Assoc. 2010;105(490):713–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hänzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 2013;14:7.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Yoshihara K, Shahmoradgoli M, Martínez E, Vegesna R, Kim H, Torres-Garcia W, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 2013;4:2612.

    Article  PubMed  CAS  Google Scholar 

  27. Hothorn T, Zeileis A. Generalized maximally selected statistics. Biometrics. 2008;64(4):1263–9.

    Article  PubMed  Google Scholar 

  28. Mayakonda A, Lin DC, Assenov Y, Plass C, Koeffler HP. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res. 2018;28(11):1747–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhang B, Horvath S. A general framework for weighted gene co-expression network analysis. Stat Appl GenetMol Biol. 2005;4:Article17.

    Google Scholar 

  30. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics. 2012;16(5):284–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M, Tanabe M. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res. 2021;49(D1):D545–51.

    Article  CAS  PubMed  Google Scholar 

  32. Kanehisa M. Toward understanding the origin and evolution of cellular organisms. Protein Sci. 2019;28(11):1947–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kanehisa M, Goto S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Charoentong P, Finotello F, Angelova M, Mayer C, Efremova M, Rieder D, et al. Pan-cancer Immunogenomic analyses reveal genotype-Immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep. 2017;18(1):248–62.

    Article  CAS  PubMed  Google Scholar 

  35. Desbois M, Wang Y. Cancer-associated fibroblasts: key players in shaping the tumor immune microenvironment. Immunol Rev. 2021;302(1):241–58.

  36. Park JE, Lenter MC, Zimmermann RN, Garin-Chesa P, Old LJ, Rettig WJ. Fibroblast activation protein, a dual specificity serine protease expressed in reactive human tumor stromal fibroblasts. J Biol Chem. 1999;274(51):36505–12.

    Article  CAS  PubMed  Google Scholar 

  37. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC bioinformatics. 2008;9:559.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Jongsma MLM, de Waard AA, Raaben M, Zhang T, Cabukusta B, Platzer R, et al. The SPPL3-Defined Glycosphingolipid Repertoire Orchestrates HLA Class I-Mediated Immune Responses. Immunity. 2021;54(1):132–150.e139.

    Article  CAS  PubMed  Google Scholar 

  39. Ahmed Laskar A, Younus H. Aldehyde toxicity and metabolism: the role of aldehyde dehydrogenases in detoxification, drug resistance and carcinogenesis. Drug Metab Rev. 2019;51(1):42–64.

    Article  CAS  PubMed  Google Scholar 

  40. Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 2009;458(7239):780–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kaur G, Gupta SK, Singh P, Ali V, Kumar V, Verma M. Drug-metabolizing enzymes: role in drug resistance in cancer. Clin Transl Oncol. 2020;22(10):1667–80.

    Article  CAS  PubMed  Google Scholar 

  42. Maccalli C, Parmiani G, Ferrone S. Immunomodulating and Immunoresistance properties of Cancer-initiating cells: implications for the clinical success of immunotherapy. Immunol Investig. 2017;46(3):221–38.

    Article  CAS  Google Scholar 

  43. Malta TM, Sokolov A, Gentles AJ, Burzykowski T, Poisson L, Weinstein JN, et al. Machine learning identifies stemness features associated with oncogenic dedifferentiation. Cell. 2018;173(2):338–354.e315.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lytle NK, Barber AG, Reya T. Stem cell fate in cancer growth, progression and therapy resistance. Nat Rev Cancer. 2018;18(11):669–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chen X, Kang R, Kroemer G, Tang D. Broadening horizons: the role of ferroptosis in cancer. Nat Rev Clin Oncol. 2021;18(5):280–96.

    Article  CAS  PubMed  Google Scholar 

  46. Teijeira Á, Garasa S, Gato M, Alfaro C, Migueliz I, Cirella A, et al. CXCR1 and CXCR2 chemokine receptor agonists produced by tumors Induce neutrophil extracellular traps that interfere with immune cytotoxicity. Immunity. 2020;52(5):856–871.e858.

    Article  CAS  PubMed  Google Scholar 

  47. Li X, Pasche B, Zhang W, Chen K. Association of MUC16 mutation with tumor mutation load and outcomes in patients with gastric cancer. JAMA Oncol. 2018;4(12):1691–8.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Brennan K, Shin JH, Tay JK, Prunello M, Gentles AJ, Sunwoo JB, et al. NSD1 inactivation defines an immune cold, DNA hypomethylated subtype in squamous cell carcinoma. Sci Rep. 2017;7(1):17064.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160(1–2):48–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Pavón MA, Arroyo-Solera I, León X, Téllez-Gabriel M, Virós D, Gallardo A, et al. The combined use of EFS, GPX2, and SPRR1A expression could distinguish favorable from poor clinical outcome among epithelial-like head and neck carcinoma subtypes. Head Neck. 2019;41(6):1830–45.

    Article  PubMed  Google Scholar 

  51. Yang X, Liu J, Wang C, Cheng KK, Xu H, Li Q, et al. miR-18a promotes glioblastoma development by down-regulating ALOXE3-mediated ferroptotic and anti-migration activities. Oncogenesis. 2021;10(2):15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Jiang T, Zhou B, Li YM, Yang QY, Tu KJ, Li LY. ALOX12B promotes carcinogenesis in cervical cancer by regulating the PI3K/ERK1 signaling pathway. Oncol Lett. 2020;20(2):1360–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zheng Y, Yin H, Boeglin WE, Elias PM, Crumrine D, Beier DR, et al. Lipoxygenases mediate the effect of essential fatty acid in skin barrier formation: a proposed role in releasing omega-hydroxyceramide for construction of the corneocyte lipid envelope. J Biol Chem. 2011;286(27):24046–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Goodwin ZA, de Guzman Strong C. Recent positive selection in genes of the mammalian epidermal differentiation complex locus. Front Genet. 2016;7:227.

    PubMed  Google Scholar 

  55. Cassandri M, Smirnov A, Novelli F, Pitolli C, Agostini M, Malewicz M, et al. Zinc-finger proteins in health and disease. Cell Death Discov. 2017;3:17071.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Zhang P, He Q, Lei Y, Li Y, Wen X, Hong M, et al. m (6) A-mediated ZNF750 repression facilitates nasopharyngeal carcinoma progression. Cell Death Dis. 2018;9(12):1169.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Yang H, Pan L, Xu C, Zhang Y, Li K, Chen S, et al. Overexpression of tumor suppressor gene ZNF750 inhibits oral squamous cell carcinoma metastasis. Oncol Lett. 2017;14(5):5591–6.

    PubMed  PubMed Central  Google Scholar 

  58. Puram SV, Tirosh I, Parikh AS, Patel AP, Yizhak K, Gillespie S, et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell. 2017;171(7):1611–1624.e1624.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Sturm G, Finotello F, Petitprez F, Zhang JD, Baumbach J, Fridman WH, et al. Comprehensive evaluation of transcriptome-based cell-type quantification methods for immuno-oncology. Bioinformatics (Oxford, England). 2019;35(14):i436–45.

    Article  CAS  Google Scholar 

  60. Lin HJ, Zuo T, Lin CH, Kuo CT, Liyanarachchi S, Sun S, et al. Breast cancer-associated fibroblasts confer AKT1-mediated epigenetic silencing of cystatin M in epithelial cells. Cancer Res. 2008;68(24):10257–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Fiori ME, Di Franco S, Villanova L, Bianca P, Stassi G, De Maria R. Cancer-associated fibroblasts as abettors of tumor progression at the crossroads of EMT and therapy resistance. Mol Cancer. 2019;18(1):70.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Posner MR, Hershock DM, Blajman CR, Mickiewicz E, Winquist E, Gorbounova V, et al. Cisplatin and fluorouracil alone or with docetaxel in head and neck cancer. N Engl J Med. 2007;357(17):1705–15.

    Article  CAS  PubMed  Google Scholar 

  63. Machiels JP, Haddad RI, Fayette J, Licitra LF, Tahara M, Vermorken JB, et al. Afatinib versus methotrexate as second-line treatment in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck progressing on or after platinum-based therapy (LUX-Head & Neck 1): an open-label, randomised phase 3 trial. Lancet Oncol. 2015;16(5):583–94.

    Article  CAS  PubMed  Google Scholar 

  64. Weiss JM, Bagley S, Hwang WT, Bauml J, Olson JG, Cohen RB, et al. Capecitabine and lapatinib for the first-line treatment of metastatic/recurrent head and neck squamous cell carcinoma. Cancer. 2016;122(15):2350–5.

    Article  CAS  PubMed  Google Scholar 

  65. Tommelein J, De Vlieghere E, Verset L, Melsens E, Leenders J, Descamps B, et al. Radiotherapy-activated Cancer-associated fibroblasts promote tumor progression through paracrine IGF1R activation. Cancer Res. 2018;78(3):659–70.

    Article  CAS  PubMed  Google Scholar 

  66. Wu F, Yang J, Liu J, Wang Y, Mu J, Zeng Q, et al. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct Target Ther. 2021;6(1):218.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Clark C, Shah S, Herman-Ferdinandez L, Ekshyyan O, Abreo F, Rong X, et al. Teasing out the best molecular marker in the AKT/mTOR pathway in head and neck squamous cell cancer patients. Laryngoscope. 2010;120(6):1159–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Wilson TG, Hanna A, Recknagel J, Pruetz BL, Baschnagel AM, Wilson GD. Prognostic significance of MTOR expression in HPV positive and negative head and neck cancers treated by chemoradiation. Head Neck. 2020;42(2):153–62.

    Article  PubMed  Google Scholar 

  69. Li SH, Lin WC, Huang TL, Chen CH, Chiu TJ, Fang FM, et al. Significance of mammalian target of rapamycin in patients with locally advanced stage IV head and neck squamous cell carcinoma receiving induction chemotherapy with docetaxel, cisplatin, and fluorouracil. Head Neck. 2016;38(Suppl 1):E844–52.

    Article  PubMed  Google Scholar 

  70. Vossen DM, Verhagen CVM, Verheij M, Wessels LFA, Vens C, van den Brekel MWM. Comparative genomic analysis of oral versus laryngeal and pharyngeal cancer. Oral Oncol. 2018;81:35–44.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

None.

Funding

The project was supported by no funding.

Author information

Authors and Affiliations

Authors

Contributions

Jing Li contributed to the conceptualization; data collection and analysis; writing–original draft; writing–review&editing; Jun Ma, Ling-Long Tang supervision; writing– review&editing. The author(s) read and approved the final manuscript.

Corresponding author

Correspondence to Jun Ma.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, J., Tang, LL. & Ma, J. Survival-related indicators ALOX12B and SPRR1A are associated with DNA damage repair and tumor microenvironment status in HPV 16-negative head and neck squamous cell carcinoma patients. BMC Cancer 22, 714 (2022). https://0-doi-org.brum.beds.ac.uk/10.1186/s12885-022-09722-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12885-022-09722-x

Keywords