| Home | E-Submission | Sitemap | Contact Us |  
Clinical and Experimental Otorhinolaryngology > Volume 18(1); 2025 > Article
Zheng, Piao, Jung, Oh, Lim, Nguyen, Shen, Park, Cui, Piao, Kim, Kim, Won, Chang, Shan, Liu, and Koo: Gene Expression Alteration by Non-thermal Plasma-Activated Media Treatment in Radioresistant Head and Neck Squamous Cell Carcinoma

Abstract

Objectives.

Head and neck squamous cell carcinoma (HNSCC) exhibits high recurrence rates, particularly in cases of radioresistant HNSCC (RR-HNSCC). Non-thermal plasma (NTP) therapy effectively suppresses the progression of HNSCC. However, the therapeutic mechanisms of NTP therapy in treating RR-HNSCC are not well understood. In this study, we explored the regulatory role of NTP in the RR-HNSCC signaling pathway and identified its signature genes.

Methods.

After constructing two RR-HNSCC cell lines, we prepared cell lysates from cells treated or not treated with NTP-activated media (NTPAM) and performed RNA sequencing to determine their mRNA expression profiles. Based on the RNA sequencing results, we identified differentially expressed genes (DEGs), followed by a bioinformatics analysis to identify candidate molecules potentially associated with NTPAM therapy for RR-HNSCC.

Results.

NTPAM reduced RR-HNSCC cell viability in vitro. RNA sequencing results indicated that NTPAM treatment activated the reactive oxygen species (ROS) pathway and induced ferroptosis in RR-HNSCC cell lines. Among the 1,924 genes correlated with radiation treatment, eight showed statistical significance in both the cell lines and The Cancer Genome Atlas (TCGA) cohort. Only five genes—ABCC3, DUSP16, PDGFB, RAF1, and THBS1—showed consistent results between the NTPAM data sequencing and TCGA data. LASSO regression analysis revealed that five genes were associated with cancer prognosis, with a hazard ratio of 2.26. In RR-HNSCC cells, NTPAM affected DUSP16, PDGFB, and THBS1 as activated markers within 6 hours, and this effect persisted for 12 hours. Furthermore, enrichment analysis indicated that these three DEGs were associated with the extracellular matrix, transforming growth factor-beta, phosphoinositide 3-kinase/protein kinase B, and mesenchymal-epithelial transition factor pathways.

Conclusion.

NTPAM therapy exerts cytotoxic effects in RR-HNSCC cell lines by inducing specific ROS-mediated ferroptosis. DUSP16, PDGFB, and THBS1 were identified as crucial targets for reversing the radiation resistance induced by NTPAM therapy, providing insights into the mechanisms and clinical applications of NTPAM treatment in RR-HNSCC.

INTRODUCTION

Head and neck squamous cell carcinoma (HNSCC) ranks among the most prevalent cancers globally, with projections indicating a 30% increase in incidence by 2030 (Global Cancer Observatory) [1,2]. HNSCC is a multifactorial disease, the risk factors of which include exposure to tobacco products, alcohol consumption, and infection with high-risk human papillomavirus strains [3,4]. The prognosis for HNSCC remains poor, especially when the cancer has metastasized to regional cervical lymph nodes or distant organs [5-7].
The treatment options for advanced HNSCC are multimodal, encompassing surgery, radiotherapy, and chemotherapy. Radiotherapy notably improves overall survival (OS) when used as part of combined treatment modalities in advanced cases [8]. However, a subset of cancer cells may survive radiation therapy, exhibiting radioresistance [9]. These radioresistant tumor cells can cause local radiation treatment failures. Moergel et al. [10] demonstrated that P63 can predict radiation treatment failure and is associated with poor clinical outcomes in HNSCC. Furthermore, studies on cellular models have indicated that radioresistant cell subtypes are more invasive than their parental cells [11]. Therefore, radioresistance significantly impacts HNSCC treatment and is crucially linked to patient survival.
Changes in the signaling pathways within tumor cells are a hallmark of radioresistant HNSCC (RR-HNSCC). These cells may develop radioresistance by inhibiting cell death pathways that are activated by DNA damage from ionizing radiation [12]. Promotion of the reactive oxygen species (ROS) pathway by IFGBP3 results in cell death in HNSCC induced by ionizing radiation [13]. Liu et al. [14] reported that hyperbaric oxygen treatment can re-sensitize RR-HNSCC cells by regulating GPX4/ferroptosis. A deeper understanding of tumor resistance to radiation therapy has facilitated the development of treatments that can counteract radioresistance, potentially enabling the administration of lower doses of radiation.
Non-thermal plasma (NTP), produced at room temperature, is a gas mixture that includes ions, electrons, photons, and free radicals. It can be generated at specific locations and intensities [15]. Mounting evidence indicates that NTP can alter ROS in cells [16], induce immunogenic cell death [17], and mediate apoptosis [18] and ferroptosis in cancer cells [19]. The effectiveness of NTP-induced cancer cell death is contingent upon the tumor cell’s capacity to manage oxidative stress, which in turn activates cell death signaling [20]. NTP leads to cell death through the cell death pathway, causes DNA damage, and results in sub-G1 arrest via ATM signaling in HNSCC [21]. Lin et al. [22] found that NTP treatment promotes tumor radiation sensitivity and induces DNA damage both in vitro and in vivo. NTP therapy holds promise for treating RR-HNSCC and potentially reducing the radiation doses required during treatment.
While NTP therapy is still in its infancy, it holds promising potential for treating head and neck cancers [20]. However, the anticancer effects and mechanisms of NTP in radioresistant tumors are not yet well understood. This study aimed to investigate the regulatory role of NTP in signaling pathways of RR-HNSCC and to identify the signature genes involved in the NTP-mediated regulation of RR-HNSCC.

MATERIALS AND METHODS

Cell Lines and culture conditions

MSK-QLL1 and SCC15 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM): Nutrient Mixture F-12 (Gibco) supplemented with 10% fetal bovine serum (Gibco) and 1% penicillin/streptomycin (Gibco). All cells were cultured in a cell culture incubator at 37 °C with 5% CO2. The cells were irradiated with 6 Gy each time, then cultured for 2–3 days until they reached 70%–80% confluency, followed by re-irradiation. This procedure was repeated 10 times. Finally, a clonogenic assay was conducted under 4 Gy irradiation to compare parental cells with repeatedly irradiated cells to confirm the acquired radioresistance of the repeatedly treated cells (RR-MSK-QLL1 and RR-SCC15).

NTP-activated media treatment

DMEM/F12 was treated using an NTP jet device. The device was composed of a mass flow controller, gas distributor, DC (direct current) power supply, plasma inverter, and NTP reactor (Fig. 1). The mass flow controller controlled the gas flow with helium (4 standard liters per minute) and oxygen (1 standard cubic centimeter per minute). The two gases were mixtures in a gas distributor and entranced in the NTP reactor. The NTP reactor was powered by a DC power supply and plasma inverter, delivering energy to two electrodes at a few kilovolts and 20 kHz, with the discharge power ranging from 10 W to 24 W. The NTP reactor comprised a quartz tube (outer diameter, 6 mm; inner diameter, 4 mm) with two electrodes, an inner stainless-steel tube electrode, and an outer ground ring electrode. The gas was converted into a plasma jet via electrodes and directed vertically into the DMEM/F12 media inside the flask. The distance between the plasma device and the medium was maintained at approximately 1 cm. The plasma’s emission spectra were reported in our previous study with hydrogen peroxide and nitric oxide (NO3, NO2) [18]. Before each experiment, DMEM/F12 was exposed to the plasma jet for 2 hours to generate fresh NTP-activated media (NTPAM). Then, radioresistant cells were treated with 100% NTPAM for 6 and 12 hours, with the untreated control as the baseline for comparison.

Clonogenic assay for radiosensitivity evaluation

Parent and RR-HNSCC cells were seeded in 60 mm dishes with 3×104 cells. On the second day, the cells were irradiated with 4 Gy. After irradiation, irradiated cells and no-irradiated control were reseeded at 1,000 cells/well in 12-well plates. Each well was supplemented with 2 mL of medium, which was replaced every 2–3 days. After 10–14 days, cells were washed with cold PBS, fixed with 4% paraformaldehyde (Thermo Scientific), and stained with 1% crystal violet (Sigma-Aldrich). Finally, images were captured, and cell counting was performed using the ImageJ software. The vitality of the cells was tested to demonstrate their radiation resistance.

Cell proliferation assay

Parent and RR-HNSCC cells were seeded into 96-well plates at a density of 6×103 cells/well. After the radioresistant cells underwent a 12-hour treatment with 100% NTPAM, they were washed twice with cold PBS, and the medium was replaced with fresh medium. Then, each well was added 10 μL of Cell Counting Kit-8 solution (CCK-8, Dojindo). The culture plates were then incubated for 1–2 hours, and absorbance was measured at 450 nm using a spectrophotometer.

Transcriptome analysis

The NTPAM-intervened (12-hour) and untreated control RR-HNSCC cell lines were harvested, and RNA was isolated using the Qiagen RNeasy Kit (Qiagen) following the manufacturer’s protocol. All experiments were conducted under clean conditions, and the equipment was preautoclaved. Sequencing was performed by Macrogen, Inc. Briefly, the quality of extracted RNA was evaluated using an Agilent 2100 Bioanalyzer RNA Nano Chip (Agilent Technologies). RNA libraries were constructed with 1 μg of RNA using the TruSeq Stranded mRNA Sample Preparation Kit v2 (Illumina) according to the manufacturer’s protocol. Library quality was analyzed using an Agilent 2100 Bioanalyzer equipped with an Agilent DNA 1000 kit. All samples were sequenced on an Illumina HiSeq 2500 platform (Illumina), yielding an average of 38 million paired-end 100 base-pair reads. The study included four groups, with three replicates: RR-MSK-QLL1, RR-SCC15, NTP-MSK-QLL1, and NTP-RR-SCC15 (after 12-hour NTPAM intervention). Transcriptomic data were analyzed using the Deseq2 method to identify differentially expressed genes (DEGs). R (version 4.3.2) was used to generate volcano plots and differential heat maps.
Gene Set Variation Analysis (GSVA) assessed ROS and cell death pathway changes using the “GSVA” R package. The ROS pathway data were downloaded from MSigDB (https://www.gsea-msigdb.org/gsea/index.jsp). The gene set for cell death was based on relevant literature [23,24]. Radiotherapy resistance gene sets were referenced in studies [25-29]. Gene sets were downloaded from MSigDB (https://www.gsea-msigdb.org/gsea/index.jsp). Subsequently, Gene Set Enrichment Analysis (GSEA) was performed on the transcriptomic data of NTPAM treatment using the “fgsea” R package.

RNA extraction and real-time quantitative polymerase chain reaction

The process for extracting total cellular RNA is detailed in section “Transcriptome analysis.” The complementary DNA (cDNA) were synthesized with 5 μg of total RNA and TOPscript RT DryMIX (Enzynomics) according to the manufacturer’s instructions. Amplification was performed using the SYBR Green qPCR Master Mix (Thermo Fisher Scientific). The real-time quantitative polymerase chain reaction reactions were performed for 40 cycles of 95 °C for 15 seconds, 60 °C for 1 minute, and 72 °C for 1 minute. Melting curve analysis was performed. The qPCR primer sequences were as follows: human ABCC3-F: 5´-TAC GTG GAC CCA AAC AAT GTG-3´, ABCC3-R: 5´-GGA ATT GCT GGA TCC GTT TCA-3´; human DUSP16-F: 5´-GTT CTC TCG TTG TTT CCC TGG-3´, DUSP16-R: 5´-CAG CTC CTT GTT GAG GAC ATC-3´; human PDGFB-F: 5´-CCA TTC CCG AGG AGC TTT ATG-3´, PDGFB-R: 5´-GGT CAT GTT CAG GTC CAA CTC-3´; human RAF1-F: 5´-TCG ATG TCA GAC TTG TGG CTA-3´, RAF1-R: 5´-TCA AAG AAG GTA GTG CTG GGA-3´; and human THBS1-F: 5´-CAT CAG TGA GAC CGA TTT CCG-3´, THBS1-R: 5´-ATA ACC TAC AGC GAG TCC AGG-3´. We used the 2−ΔΔCt model for relative quantification of real-time qPCR fold change. The cycle threshold values obtained by real-time qPCR were imported into Microsoft Excel. The formulas for 2−ΔΔCt method are as follows.
ΔCtctrl=(Ctctrl−CtGAPDH), ΔCtNTPAM=(CtNTPAM−CtGAPDH)
ΔΔCtctrl=ΔCtctrl−ΔCtctrl average
ΔΔCtNTPAM=ΔCtNTPAM−ΔCtctrl average
The relative expression values are calculated as 2−ΔΔCtctrl, 2−ΔΔCtNTPAM.

The Cancer Genome Atlas and gene expression omnibus datasets assays

The Cancer Genome Atlas (TCGA)-HNSCC dataset was downloaded from the official TCGA website (https://portal.gdc.cancer.gov/). It includes transcriptomic data from 520 tumor tissues and 46 adjacent non-cancerous tissues. The TCGA clinical dataset was downloaded using the R package “TCGAbiolinks.” Based on the information from “additional radiation therapy,” “radiation therapy,” and “follow-up treatment success,” the outcomes of the patients who were treated with radiation can be determined. According to the outcomes of “complete remission/response” (CR), “partial remission/response” (PR), “stable disease” (SD), “persistent disease” (PED), and “progressive disease” (PRD), the patients were divided into two groups: “responders” (CR/PR) and “non-responders” (SD/PED/PRD) (PED patients were originally CR but developed new neoplasms, so they were grouped with SD and PRD) [30]. Survival analysis for specific selected genes was conducted using the “survival” and “survminer” R packages. The Kaplan-Meier method and log-rank test were used for survival analysis. The GSE213862 dataset includes 46 normal tissues, 43 negative-margin tissues, and 46 tumor tissues. This database was downloaded from the National Center for Biotechnology Information (NCBI) website (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi). According to the gene response to NTPAM intervention, we classified the genes into immediate-early genes (DUSP16, PDGFB, and THBS1) and delayed primary response genes (ABCC3 and RAF1) [31]. To verify the pathways associated with immediate-early genes, we categorized them into high- and low-expression groups based on the median expression levels of the genes [32]. Specifically, we performed a differential analysis between the DUSP16low+PDGFBhigh+THBS1high and DUSP16high+PDGFBlow+THBS1low groups to identify the pathways associated with specific immediate-early genes. Differential analysis of TCGA-HNSCC and Gene Expression Omnibus data was performed using the R package “Deseq2.” The R package “clusterProfiler,” “ReactomePA,” “fgsea” were utilized for GSEA analysis, applying gene sets from Kyoto Encyclopedia of Genes and Genomes (KEGG; https://www.genome.jp/kegg/), Gene Ontology (GO; https://geneontology.org/), and Reactome (https://reactome.org/).

Specific selected genes

A flowchart of the study is shown in Fig. 2. Significant gene pathways were selected using GSEA. We then identified the genes associated with these pathways. Genes with P<0.05, |log2(fold change)|>1.5 genes underwent survival analysis in TCGA-HNSCC. Genes showing statistically significant OS and disease-specific survival (DSS) were identified. Finally, considering the clinical information and NTPAM treatment outcomes, genes consistent with the selection criteria for both risk and protective factors were chosen.

Statistical analysis

Data were analyzed using SPSS version 26 (IBM Corp.) and R software version 4.3.2 (R Foundation for Statistical Computing). Normality was assessed for the pre-analyzed data. If the data exhibited a normal distribution, comparisons between the two groups were conducted using the t-test, whereas comparisons among multiple groups were performed using analysis of variance. Pairwise comparisons of normally distributed data were conducted using Dunnett’s t-test to compare multiple experimental groups with the control group. Non-normally distributed data were analyzed using non-parametric tests such as the Wilcoxon test. The results are expressed as mean±standard deviation. Statistical significance was set at α=0.05.

RESULTS

NTPAM treatment activates the ROS and cell death pathways in RR-HNSCC cells

We successfully established two RR-HNSCC cell lines by subjecting the parental HNSCC cells to chronic radiation stimulation. The clonogenic assay results indicated that the RR-MSK-QLL1 and RR-SCC15 cell lines exhibited higher cell viability than their respective parental cell lines following exposure to a 4 Gy radiation dose (Fig. 3A and B). To assess the impact of NTPAM on RR-HNSCC cells, we evaluated the viability of both untreated and NTPAM-treated RR-MSK-QLL1 and RR-SCC15 cells using the CCK8 assay. The treatment with NTPAM successfully inhibited the proliferation of these cell lines (Fig. 3C and D). Several studies have shown that NTPAM induces ROS-dependent cell death in various cancers. Consequently, we explored the changes in ROS and cell death signaling pathways in RR-HNSCC cell lines following NTPAM treatment through bioinformatics analysis. GSVA was employed to evaluate changes in ROS and cell death pathways based on RNA sequencing results. Using gene sets from the ROS pathway, NTPAM therapy was found to activate the ROS pathway in both RR-HNSCC cell lines (Fig. 3E and F). Post-NTPAM treatment, the RR-MSK-QLL1 cell line showed an increase in ROS-related pathways according to both the GO and Hallmark databases. In contrast, the RR-SCC15 cell line only demonstrated upregulation of ROS pathways in the GO database. The results of cell death analysis revealed that NTPAM treatment significantly activated the ferroptosis (t=10.15, P<0.001) and autophagy pathway (t=4.02, P=0.16) in RR-MSK-QLL1 cells (Fig. 3G). In contrast, NTPAM treatment cells had higher apoptosis (t=4.23, P=0.01), ferroptosis (t=5.51, P=0.005), and alkaliptosis (t=6.54, P=0.003) scores in RR-SCC15. (Fig. 3H). In RR-HNSCC cells, NTPAM treatment activated the ROS pathway and promoted cell death.

NTPAM treatment regulates pathways associated with radiation therapy resistance in RR-HNSCC cell lines

Combining 58 radiotherapy resistance pathways (TCGA) [25] with seven additional gene sets [26-28], along with a set of 21 radiotherapy-resistant genes [29], resulted in a total of 66 pathways associated with radiotherapy resistance. To investigate the regulation of pathways associated with radiotherapy resistance following interaction with NTPAM, GSEA was performed on the RNA sequencing data of RR-HNSCC cell lines that received or did not receive NTPAM treatment, focusing on 66 pathways associated with radiotherapy resistance. After NTPAM treatment of the RR-MSK-QLL1 cell line (Fig. 4A), 15 pathways related to radiotherapy resistance exhibited significant changes, whereas in the RR-SCC15 cell line, 8 pathways displayed significant results (Fig. 4B). In both cell lines, the KEGG ribosome, KEGG MAPK signaling, Reactome peptide ligand-binding receptors, and KEGG cytokine-cytokine receptor interaction pathways were upregulated after NTPAM treatment. NTPAM treatment reduced the expression of the Normalized Enrichment Score of Reactome heparan sulfate/heparin (HS-GAG) metabolism signaling pathway. The GSEA analysis revealed that 18 pathways related to radiotherapy resistance were correspondingly influenced by NTPAM treatment.
Subsequently, to identify hub genes affected by NTPAM treatment in RR-HNSCC cells, we analyzed genes that were significantly altered in pathways associated with resistance to radiation therapy. This analysis encompassed a total of 1,924 genes. DEG analysis showed that, of these genes, 290 were significantly upregulated and 184 were downregulated in the RR-MSK-QLL1 cell line (Fig. 4C and E). In the RR-SCC15 cell line, NTPAM treatment resulted in the significant upregulation of 181 genes and downregulation of 209 genes from the same pool of 1,924 genes (Fig. 4D and F). Across both RR-HNSCC cell lines, there were 117 genes commonly upregulated and 73 commonly downregulated following NTPAM therapy.

Discovery of hub genes for NTPAM therapy in RR-HNSCC cells

The TCGA-HNSCC database was utilized to perform survival regression analysis using 117 upregulated and 73 downregulated DEGs from both cell lines. This analysis helped identify key genes responsive to NTPAM therapy in RR-HNSCC cell lines. We obtained survival analysis results for eight genes that were consis tent with the effects of NTPAM treatment. The genes ABCC3 (hazard ratio for overall survival [HROS]=1.35, hazard ratio for DSS [HRDSS]=1.67), PDGFB (HROS=1.88, HRDSS=1.85), and THBS1 (HROS=1.67, HRDSS=2.13) were identified as risk factors for poor OS and DSS in patients with head and neck cancer (Table 1). After NTPAM treatment, the expression of these genes was downregulated in RR-MSK-QLL1 and RR-SCC15 cells (Table 1). Similarly, the protective genes DUSP16 (HROS=0.69, HRDSS=0.6), DUSP8 (HROS=0.76, HRDSS=0.65), LTA (HROS=0.74, HRDSS=0.67), PIK3R3 (HROS=0.64, HRDSS=0.59), and RAF1 (HROS=0.74, HRDSS=0.68) were upregulated in the NTPAM-treated RR-HNSCC cell lines (Table 1).
Subsequently, we evaluated the expression of eight genes in tumor and adjacent normal tissues using the TCGA-HNSCC database. The analysis revealed that the expression levels of ABCC3, PDGFB, and RAF1 were significantly different from those in normal tissues, aligning with the findings from the survival regression analysis (Fig. 5A, E, and G). Although DUSP16 (P=0.374) and THBS1 (P=0.098) did not reach statistical significance (Fig. 5B and H), the trends observed in the survival analysis were consistent with our data (Table 1). In contrast, despite the survival regression results, the expression of DUSP8, LTA, and PIK3R3 was higher in cancer tissues than in adjacent normal tissues (Fig. 5C, D, and F). Consequently, our analysis primarily focused on the expression of ABCC3, PDGFB, RAF1, DUSP16, and THBS1.

Expression profiles and characteristics of the five hub genes

Next, we explored the relationship between these five genes and radiotherapy prognosis. In the TCGA-HNSCC cohort, 309 patients underwent radiotherapy (CR, 163; PR, 5; SD, 4; PED, 6; PRD, 57; missing data, 74). Comparing the expression of these five genes between responders (CR and PR) and non-responders (SD, PED, and PRD), high expression levels of PDGFB and THBS1 were associated with a poor prognosis in radiotherapy for head and neck cancer (Fig. 6C and E) [30]. However, no differences in ABCC3, DUSP16, and RAF1 were observed among radiotherapy patients (Fig. 6A, B and D). These findings suggested that the PDGFB and THBS1 genes, which NTPAM regulated, were also associated with clinical radioresistance and prognosis in HNSCC. In the radioresistance pathway, ABCC3 and PDGFB were enriched in the WP NRF2 pathway. DUSP16, PDGFB, RAF1, and THBS1 were enriched in the KEGG MAPK signaling pathway, KEGG melanoma, KEGG focal adhesion, and REACTOME hemostasis pathways (Fig. 6F). We subsequently derived a risk-scoring formula using LASSO Cox regression. The risk ratio between the high-risk-score group and the low-risk-score group was 2.26 (P=0.001) (Fig. 6F). Additionally, the expression of ABCC3, PDGFB, and THBS1 was positively correlated with the risk score, whereas that of DUSP16 and RAF1 was negatively correlated (Fig. 6G).

NTPAM regulated the expression of genes in a time-dependent manner

To explore whether the therapeutic effects of NTPAM on RR-HNSCC cells depended on the time of manifestation, we treated RR-HNSCC cells with NTPAM for 6 and 12 hours and measured the mRNA expression of the five genes using PCR. Within the first 6 hours, NTPAM regulated the expression of DUSP16, PDGFB, and THBS1, and this regulation continued until 12 hours. However, the expression of ABCC3 and RAF1 only changed 12 hours after NTPAM treatment (Fig. 7). Consistent with previous results, the tumor candidate protective genes DUSP16 and RAF1 showed increased expression levels following the NTPAM intervention, whereas NTPAM suppressed the potential oncogenes ABCC3, PDGFB, and THBS1.
According to the PCR results, NTPAM intervention regulated the expression of DUSP16, PDGFB, and THBS1 in the primary response, whereas it regulated the expression of ABCC3 and RAF1 in the secondary response [31]. NTPAM may regulate subsequent cellular responses via DUSP16, PDGFB, and THBS1. To explore the pathways related to immediate-early genes, we divided the samples into two groups (DUSP16high+PDGFBlow+ THBS1low and DUSP16low+PDGFBhigh+THBS1high) based on their gene expression levels. These groups were then compared to predict the early regulation of signaling pathways by NTPAM. In the DUSP16low+PDGFBhigh+THBS1high group and the DUSP16high+ PDGFBlow+THBS1low group of tumors, the GSEA-GO results indicated an association between upregulation of the extracellular matrix (ECM) signaling pathway and downregulation of the T-cell receptor signaling pathway (Fig. 8A). In the GSEA-KEGG analysis, upregulation of the cellular processes-focal adhesion pathway was observed, accompanied by activation of ECM, phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT), and transforming growth factor-beta (TGF-β) signaling pathways, whereas some metabolism pathways, including arachidonic acid metabolism, linoleic acid metabolism, and arginine biosynthesis were downregulated (Fig. 8B). Consistent with the KEGG results, GSEA-REACTOME analysis revealed the upregulation of the ECM signaling pathway and activation of the mesenchymal-epithelial transition factor (MET) signaling pathway. Additionally, the downregulation of fatty acid metabolism was observed in these high-risk tumors (Fig. 8C). Overall, in the primary response of RR-OSCC to NTPAM, the genes DUSP16, PDGFB, and THBS1 were activated. The GSEA results indicated that the ECM, fatty acid metabolism, and PI3K-AKT, TGF-β, and MET signaling pathways may be responsible for the changes in the NTPAM immediate-early genes.

DISCUSSION

Radiotherapy is a double-edged sword that induces DNA damage through ROS, initiating cell death [33]. Although cancer patients benefit from radiation treatment, high doses of radiation can be toxic, causing physiological discomfort, reduced compliance, and even death [33]. Nonetheless, radiotherapy is essential for treating HNSCC. Yet, treatment resistance poses a significant clinical challenge [34]. Therefore, identifying predictive biomarkers for radioresistance and potential targets for radiosensitization is necessary for overcoming radioresistance in HNSCC.
In this study, we developed two radioresistant HNSCC cell lines, RR-MSK-QLL1 and RR-SCC15, by repeatedly exposing radiosensitive human parental cells to radiation. We then induced NTPAM-mediated cell death in these radioresistant cells to explore the regulatory molecules linked to radioresistance through RNA sequencing. The RNA sequencing data indicated that NTPAM therapy stimulated the ROS pathway and decreased cell viability in RR-HNSCC cells by enhancing pathways involved in regulated cell death. These results align with those reported by Vandamme et al. [16], who emphasized the critical role of the ROS pathway in NTP treatment of tumors. Therefore, NTPAM treatment appears promising for overcoming radiotherapy resistance in HNSCC.
While the study by Vandamme et al. [16] has linked the ROS pathway to the antitumor effects of NTP, it is evident that ROS alone cannot fully account for NTP’s ability to reverse the radioresistance of tumors. Previous studies have indicated that the ROS pathway regulates ribosomes [35], MAPK signaling [36], and cytokine-cytokine receptor interactions [37]. Additionally, ribosomes [28], MAPK signaling [38], peptide ligand-binding receptors [39], and cytokine-cytokine receptor interactions [40] are associated with the sensitivity of HNSCC to radiation therapy. Thus, NTP treatment could affect the ROS pathway in RR-HNSCC and modulate the pathways associated with radiation sensitivity.
Our transcriptomic analysis revealed that NTPAM processing regulates several key cellular components and pathways in two RR-HNSCC cell lines, including ribosomes, MAPK signaling, peptide ligand-binding receptors, cytokine-cytokine receptor interactions, and heparan sulfate/heparin metabolism. These findings suggest that NTPAM inhibits pathways associated with radioresistance in HNSCC and enhances the cytotoxic effectiveness of NTPAM treatment for RR-HNSCC. Additionally, we identified biomarkers linked to NTPAM treatment among genes involved in radioresistance pathways. Our research confirmed that the expression of three potential oncogenes (ABCC3, PDGFB, and THBS1) was downregulated due to the effect of NTPAM, while two candidate protective genes (DUSP16 and RAF1) were upregulated. Moreover, PDGFB and THBS1 were associated with poor prognosis in patients with TCGA-HNSCC undergoing radiotherapy. Our results suggest that ABCC3, DUSP16, PDGFB, RAF1, and THBS1 are potential biomarkers of NTPAM-regulated radioresistance.
Drobin et al. [41] showed that PDGFB is a biomarker for predicting radiation sensitivity in patients with head and neck cancers. Pal et al. [42] found that TGF-β stimulated THBS1, promoting the invasion of HNSCC and thereby affecting cancer prognosis. Research on the association between ABCC3, DUSP16, and RAF1 with radioresistance is relatively limited; however, these genes are crucial in tumor drug resistance and proliferation. ABCC3 has also been linked to drug resistance across various cancers [43]. In several solid tumor types, including nasopharyngeal carcinoma, colorectal cancer, gastric cancer, and breast cancer, DUSP16 targets the JNK and p38 pathways to regulate tumor cell death [44]. The role of RAF1 in cancer is contentious. It has been implicated in promoting lymphatic metastasis in hypopharyngeal cancer [45]. Moreover, RAF1 overexpression suppressed apoptosis in cancer cells [46]. Although these five hub genes were associated with NTPAM treatment in RR-HNSCC cell lines, their upstream and downstream relationships remain unclear.
We assessed gene expression at various time points to analyze the temporal dynamics of gene responses during NTPAM treatment. Understanding these dynamics is essential, as they can elucidate the mechanisms of action and the timing of gene regulation changes in response to treatment. In this study, we confirmed that the expression of DUSP16, PDGFB, and THBS1 in RR-HNSCC cells was regulated after 6 hours of NTPAM treatment, while the expression of ABCC3 and RAF1 was affected only after 12 hours. High expression levels of the candidate protective gene DUSP16, coupled with low expression levels of the potential oncogenes PDGFB and THBS1, were identified as early regulatory effects of NTPAM treatment. Our analysis indicated that these early regulatory effects were associated with ECM, PI3K-AKT, TGF-β, and MET signaling pathways. Jing et al. [47] reported that the PI3K-AKT pathway regulated PDGFB expression, thereby promoting the proliferation and metastasis of HNSCC.
Additionally, THBS1 acted as a critical physiological activator of TGF-β and a potent inducer of the epithelial-to-mesenchymal transition [48]. Reports on the PI3K-AKT signaling pathway in HNSCC have shown that THBS1 regulates this pathway. Our study identified temporal changes in gene expression in RR-HNSCC cells following NTPAM treatment. In addition, this analytical approach has deepened our understanding of the upstream-downstream relationships among genes. Nonetheless, conclusions drawn about the five genes in this study are constrained by the reliance on in vitro experiments and TCGA data, without validation through in vivo models or independent clinical cohorts. Therefore, animal studies and clinical research are essential to validate these hub genes and to further explore the associated signaling pathways in future studies.
NTP treatment is noninvasive and easy to administer, reducing the burden on patients undergoing treatment. Additionally, Bekeschus [20] discussed the safety of gas plasma, noting that NTP exhibited good safety and lacked severe side effects. Therefore, NTP therapy is a promising adjunctive treatment. Beyond its inhibitory effects on cancer [49]. NTP also reduces tumor radioresistance and drug resistance [50]. However, the specific effects of NTP treatment and its mechanisms of action on RR-HNSCC remain unclear. Our study validated the antitumor efficacy of NTPAM treatment and identified five hub genes that are associated with the ability of NTPAM treatment to reverse radiotherapy resistance in HNSCC.
In conclusion, our research demonstrates that NTPAM treatment can effectively counteract tumors in RR-HNSCC. Furthermore, ABCC3, DUSP16, PDGFB, RAF1, and THBS1 may serve as novel biomarkers for the NTPAM treatment of RR-HNSCC cells. These findings offer valuable insights into the mechanisms underlying the NTPAM treatment of radioresistant tumors.

HIGHLIGHTS

▪ Non-thermal plasma-activated media (NTPAM) therapy activated the reactive oxygen species and cell death pathways in radioresistant head and neck squamous cell carcinoma (RR-HNSCC) cells.
▪ NTPAM therapy regulated pathways associated with radiation therapy resistance in RR-HNSCC cell lines.
ABCC3, DUSP16, PDGFB, RAF1, and THBS1 may serve as novel biomarkers for NTPAM treatment in RR-HNSCC cells.
▪ NTPAM therapy targets the ECM, TGF-β, PI3K-AKT, and MET pathways, providing insights into reversing radiation resistance in RR-HNSCC.

CONFLICTS OF INTEREST

No potential conflict of interest relevant to this article was reported.

ACKNOWLEDGMENTS

This work was supported by research funding from Chungnam National University.

AUTHOR CONTRIBUTIONS

Methodology: SZ, YP, CO, MAL. Software: QN. Validation: YP, QN. Formal analysis: QN. Investigation: SZ. Data curation: SZ, MAL, QN, SS, SHP, SC, SP. Visualization: CO. Supervision: YIK, JWK, JWC, HRW, YS, LL. Project administration: YIK, JWK, JWC, HRW, YS, LL, BSK. Funding acquisition: BSK. Writing–original draft: SZ, SNJ, YP. Writing–review & editing: SNJ, BSK. All authors read and agreed to the published version of the manuscript.

Fig. 1.
Diagram of the non-thermal plasma (NTP)-activated media generation device. DC, direct current.
ceo-2024-00238f1.jpg
Fig. 2.
Overview of the flowchart for hub gene selection and validation, showing the cytotoxic effect of non-thermal plasma-activated media (NTPAM) on radioresistant head and neck squamous cell carcinoma (RR-HNSCC). GSEA, Gene Set Enrichment Analysis; TCGA, The Cancer Genome Atlas; RT-qPCR, real-time quantitative polymerase chain reaction; KEGG, Kyoto Encyclopedia of Genes and Genomes; GO, Gene Ontology.
ceo-2024-00238f2.jpg
Fig. 3.
The effects of non-thermal plasma-activated media (NTPAM) on radioresistant head and neck squamous cell carcinoma (RR-HNSCC) cells. Clonogenic assays using the RR-MSK-QLL1/MSK-QLL1 (A) and RR-SCC15/SCC15 cell lines (B). Viability of RR-MSK-QLL1 (C) and RR-SCC15 (D) cells with and without NTPAM treatment. NTPAM-induced reactive oxygen species pathways in RR-MSK-QLL1 (E) and RR-SCC15 (F) cell lines. NTPAM-induced cell death pathways in RR-MSK-QLL1 (G) and RR-SCC15 (H) cell lines. GOBP, Gene Ontology Biological Process. *P<0.05, **P<0.01, ***P<0.001.
ceo-2024-00238f3.jpg
Fig. 4.
The effects of non-thermal plasma-activated media (NTPAM) on radiation-related pathways and correlated genes in radioresistant head and neck squamous cell carcinoma (RR-HNSCC) cells. Gene Set Enrichment Analysis of the effect of NTPAM on radiation-related pathways in RR-MSK-QLL1 (A) and RR-SCC15 (B) cell lines. Volcano plots of radiation-related pathway genes in RR-MSK-QLL1 (C) and RR-SCC15 cells (D). Heat maps of the differentially expressed gene in NTPAM-RR-MSK-QLL1/RR-MSK-QLL1 (E) and NTPAM-RR-SCC15/RR-SCC15 (F) cells.
ceo-2024-00238f4.jpg
Fig. 5.
Comparison of the expression of eight selected genes between cancer tissues and surrounding normal tissues in The Cancer Genome Atlas (TCGA)-head and neck squamous cell carcinoma (HNSCC). The gene expression levels of ABCC3 (A), DUSP16 (B), DUSP8 (C), LTA (D), PDGFB (E), PIK3R3 (F), RAF1 (G), and THBS1 (H). *P<0.05, **P<0.01, ****P<0.0001.
ceo-2024-00238f5.jpg
Fig. 6.
The relationship between five selected genes and the prognosis of patients receiving radiotherapy. Gene expression levels of ABCC3 (A), DUSP16 (B), PDGFB (C), RAF1 (D), and THBS1 (E) in patients with The Cancer Genome Atlas (TCGA)-head and neck squamous cell carcinoma (HNSCC) receiving radiotherapy. (F) Sankey diagram showing the relationships between five genes and their associated pathways. (G) Distribution of risk scores and survival status in the TCGA-HNSCC cohort with the five hub genes and Kaplan-Meier survival curve showing the overall survival of TCGA-HNSCC patients. CR, complete remission/response; PR, partial remission/response; SD, stable disease; PED, persistent disease; PRD, progressive disease; HR, hazard ratio. *P<0.05, **P<0.01.
ceo-2024-00238f6.jpg
Fig. 7.
The expression of hub genes in radioresistant head and neck squamous cell carcinoma (RR-HNSCC) cell lines after non-thermal plasma-activated media (NTPAM) treatment. The mRNA expression levels of ABCC3, DUSP16, PDGFB, RAF1, and THBS1 in RR-MSK-QLL1 (A) and RR-SCC15 (B) cells. *P<0.05, **P<0.01, ***P<0.001.
ceo-2024-00238f7.jpg
Fig. 8.
Analysis of Gene Set Enrichment Analysis (GSEA) between two groups (DUSP16low+PDGFBhigh+THBS1high) and (DUSP16high+PDGFBlow+ THBS1low) in the Cancer Genome Atlas (TCGA)-head and neck squamous cell carcinoma (HNSCC) and GSE213862 databases. (A) GSEA- Gene Ontology (GO). (B) GSEA- Kyoto Encyclopedia of Genes and Genomes (KEGG). (C) GSEA-REACTOME. NES, normalized enrichment score; BP, biological process; CC, cellular component; MF, molecular function.
ceo-2024-00238f8.jpg
Table 1.
Key genes associated with TCGA-HNSCC survival affected by the NTPAM intervention
Gene OS HR OS P-valuea) DSS HR DSS P-valuea) NTPAM-SCC15-FCb) NTPAM-SCC15-Pc) NTPAM-MSK-QLL1-FCd) NTPAM-MSK-QLL1-Pe)
ABCC3 1.35 0.029 1.67 0.005 –1.69 0.004 –4.89 <0.001
DUSP16 0.69 0.007 0.60 0.005 2.83 <0.001 1.90 0.008
DUSP8 0.76 0.044 0.65 0.015 5.26 <0.001 1.77 0.027
LTA 0.74 0.025 0.67 0.025 9.07 <0.001 12.43 <0.001
PDGFB 1.37 0.022 1.53 0.019 –1.90 <0.001 –3.72 <0.001
PIK3R3 0.64 0.001 0.59 0.003 4.21 <0.001 3.93 <0.001
RAF1 0.74 0.029 0.68 0.032 1.86 <0.001 1.56 0.005
THBS1 1.50 0.003 1.81 0.001 –2.37 <0.001 –3.53 <0.001

TCGA, The Cancer Genome Atlas; HNSCC, head and neck squamous cell carcinoma; NTPAM, non-thermal plasma-activated media; OS, overall survival; HR, hazard ratio; DSS, disease-specific survival.

a) P-value for the HR.

b),c) RR-SCC15 cell line.

d),e) RR-MSK-QLL1 cell line.

b),d) The fold change of differentially expressed genes (DEGs) between NTPAM-treated and untreated radio-resistant cells.

c),e) The P-values of DEGs between NTPAM-treated and untreated radio-resistant cells.

REFERENCES

1. Johnson DE, Burtness B, Leemans CR, Lui VW, Bauman JE, Grandis JR. Head and neck squamous cell carcinoma. Nat Rev Dis Primers. 2020 Nov;6(1):92.
crossref pmid pmc pdf
2. Leemans CR, Snijders PJ, Brakenhoff RH. The molecular landscape of head and neck cancer. Nat Rev Cancer. 2018 May;18(5):269-82.
crossref pmid pdf
3. Ang KK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen-Tan PF, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010 Jul;363(1):24-35.
crossref pmid pmc
4. Zabolotnyi D, Kizim Y, Zabolotna D, Sulaieva O, Kizim V. Laryngopharyngeal reflux alters macrophage polarization in human papilloma virus-negative squamous cell carcinoma of the larynx in males. Clin Exp Otorhinolaryngol. 2021 May;14(2):240-3.
crossref pmid pmc pdf
5. Uz U, Eskiizmir G. Association between interleukin-6 and head and neck squamous cell carcinoma: a systematic review. Clin Exp Otorhinolaryngol. 2021 Feb;14(1):50-60.
crossref pmid pmc pdf
6. Yin CY, Zhang SS, Zhong JT, Zhou SH. Pepsin and laryngeal and hypopharyngeal carcinomas. Clin Exp Otorhinolaryngol. 2021 May;14(2):159-68.
crossref pmid pmc pdf
7. Lee DY, Hah JH, Jeong WJ, Chung EJ, Kwon TK, Ahn SH, et al. The expression of defensin-associated genes may be correlated with lymph node metastasis of early-stage tongue cancer. Clin Exp Otorhinolaryngol. 2022 Nov;15(4):372-9.
crossref pmid pmc pdf
8. Mucke T, Konen M, Wagenpfeil S, Kesting MR, Wolff KD, Holzle F. Low-dose preoperative chemoradiation therapy compared with surgery alone with or without postoperative radiotherapy in patients with head and neck carcinoma. Ann Surg Oncol. 2011 Oct;18(10):2739-47.
crossref pmid pdf
9. Wu TS, Lin BR, Chang HH, Chang CC. Radio resistance mechanisms of cancers: an overview and future perspectives. Biol Med. 2015 Jan;S2:002.
crossref
10. Moergel M, Abt E, Stockinger M, Kunkel M. Overexpression of p63 is associated with radiation resistance and prognosis in oral squamous cell carcinoma. Oral Oncol. 2010 Sep;46(9):667-71.
crossref pmid
11. You GR, Chang JT, Li YL, Chen YJ, Huang YC, Fan KH, et al. Molecular interplays between cell invasion and radioresistance that lead to poor prognosis in head-neck cancer. Front Oncol. 2021 Jul;11:681717.
crossref pmid pmc
12. Hutchinson MN, Mierzwa M, D’Silva NJ. Radiation resistance in head and neck squamous cell carcinoma: dire need for an appropriate sensitizer. Oncogene. 2020 Apr;39(18):3638-49.
crossref pmid pmc pdf
13. Wang SH, Chen YL, Hsiao JR, Tsai FY, Jiang SS, Lee AY, et al. Insulin-like growth factor binding protein 3 promotes radiosensitivity of oral squamous cell carcinoma cells via positive feedback on NF-κB/IL-6/ROS signaling. J Exp Clin Cancer Res. 2021 Mar;40(1):95.
crossref pmid pmc pdf
14. Liu J, An W, Zhao Q, Liu Z, Jiang Y, Li H, et al. Hyperbaric oxygen enhances X-ray induced ferroptosis in oral squamous cell carcinoma cells. Oral Dis. 2024 Mar;30(2):116-27.
pmid
15. Laroussi M, Akan T. Arc‐free atmospheric pressure cold plasma jets: a review. Plasma Process Polym. 2007 Nov;4(9):777-88.
crossref
16. Vandamme M, Robert E, Lerondel S, Sarron V, Ries D, Dozias S, et al. ROS implication in a new antitumor strategy based on non-thermal plasma. Int J Cancer. 2012 May;130(9):2185-94.
crossref pmid
17. Khalili M, Daniels L, Lin A, Krebs FC, Snook AE, Bekeschus S, et al. Non-thermal plasma-induced immunogenic cell death in cancer: a topical review. J Phys D Appl Phys. 2019 Oct;52(42):423001.
crossref pmid pmc pdf
18. Jung SN, Oh C, Chang JW, Liu L, Lim MA, Jin YL, et al. EGR1/GADD45α activation by ROS of non-thermal plasma mediates cell death in thyroid carcinoma. Cancers (Basel). 2021 Jan;13(2):351.
crossref pmid pmc
19. Lin A, Sahun M, Biscop E, Verswyvel H, De Waele J, De Backer J, et al. Acquired non-thermal plasma resistance mediates a shift towards aerobic glycolysis and ferroptotic cell death in melanoma. Drug Resist Updat. 2023 Mar;67:100914.
crossref pmid
20. Bekeschus S. Medical gas plasma technology: roadmap on cancer treatment and immunotherapy. Redox Biol. 2023 Sep;65:102798.
crossref pmid pmc
21. Chang JW, Kang SU, Shin YS, Kim KI, Seo SJ, Yang SS, et al. Non-thermal atmospheric pressure plasma induces apoptosis in oral cavity squamous cell carcinoma: involvement of DNA-damage-triggering sub-G(1) arrest via the ATM/p53 pathway. Arch Biochem Biophys. 2014 Mar;545:133-40.
crossref pmid
22. Lin L, Wang L, Liu Y, Xu C, Tu Y, Zhou J. Non‑thermal plasma inhibits tumor growth and proliferation and enhances the sensitivity to radiation in vitro and in vivo. Oncol Rep. 2018 Dec;40(6):3405-15.
crossref pmid pmc
23. Zou Y, Xie J, Zheng S, Liu W, Tang Y, Tian W, et al. Leveraging diverse cell-death patterns to predict the prognosis and drug sensitivity of triple-negative breast cancer patients after surgery. Int J Surg. 2022 Nov;107:106936.
crossref pmid
24. Luo Y, Liu L, Zhang C. Identification and analysis of diverse cell death patterns in diabetic kidney disease using microarray-based transcriptome profiling and single-nucleus RNA sequencing. Comput Biol Med. 2024 Feb;169:107780.
crossref pmid
25. Bornstein S, Schmidt M, Choonoo G, Levin T, Gray J, Thomas CR Jr, et al. IL-10 and integrin signaling pathways are associated with head and neck cancer progression. BMC Genomics. 2016 Jan;17:38.
crossref pmid pmc pdf
26. Eke I, Zscheppang K, Dickreuter E, Hickmann L, Mazzeo E, Unger K Jr, et al. Simultaneous β1 integrin-EGFR targeting and radiosensitization of human head and neck cancer. J Natl Cancer Inst. 2015 Feb;107(2):dju419.
crossref pmid
27. O’Cathail SM, Wu CH, Thomas R, Hawkins MA, Maughan TS, Lewis A. NRF2 mediates therapeutic resistance to chemoradiation in colorectal cancer through a metabolic switch. Antioxidants (Basel). 2021 Aug;10(9):1380.
crossref pmid pmc
28. Metge BJ, Alsheikh HA, Chen D, Elhamamsy AR, Hinshaw DC, Chen BR, et al. Ribosome biosynthesis and hedgehog activity are cooperative actionable signaling mechanisms in breast cancer following radiotherapy. NPJ Precis Oncol. 2023 Jun;7(1):61.
crossref pmid pmc pdf
29. Ye X, Liu J, Quan R, Lu Y, Zhang J. DKK1 affects survival of patients with head and neck squamous cell carcinoma by inducing resistance to radiotherapy and immunotherapy. Radiother Oncol. 2023 Apr;181:109485.
crossref pmid
30. Hua Y, Sun Z, Xiao Y, Li H, Ma X, Luo X, et al. Pretreatment CT-based machine learning radiomics model predicts response in unresectable hepatocellular carcinoma treated with lenvatinib plus PD-1 inhibitors and interventional therapy. J Immunother Cancer. 2024 Jul;12(7):e008953.
crossref pmid pmc
31. Tullai JW, Schaffer ME, Mullenbrock S, Sholder G, Kasif S, Cooper GM. Immediate-early and delayed primary response genes are distinct in function and genomic architecture. J Biol Chem. 2007 Aug;282(33):23981-95.
crossref pmid pmc
32. Chen D, Li G, Ji C, Lu Q, Qi Y, Tang C, et al. Enhanced B7-H4 expression in gliomas with low PD-L1 expression identifies super-cold tumors. J Immunother Cancer. 2020 May;8(1):e000154.
crossref pmid pmc
33. Wang Y, Qi H, Liu Y, Duan C, Liu X, Xia T, et al. The double-edged roles of ROS in cancer prevention and therapy. Theranostics. 2021 Mar;11(10):4839-57.
crossref pmid pmc
34. Anderson G, Ebadi M, Vo K, Novak J, Govindarajan A, Amini A. An updated review on head and neck cancer treatment with radiation therapy. Cancers (Basel). 2021 Sep;13(19):4912.
crossref pmid pmc
35. Shcherbik N, Pestov DG. The impact of oxidative stress on ribosomes: from injury to regulation. Cells. 2019 Nov;8(11):1379.
crossref pmid pmc
36. Jalmi SK, Sinha AK. ROS mediated MAPK signaling in abiotic and biotic stress-striking similarities and differences. Front Plant Sci. 2015 Sep;6:769.
crossref pmid pmc
37. Xiao Z, Deng S, Liu H, Wang R, Liu Y, Dai Z, et al. Glutamine deprivation induces ferroptosis in pancreatic cancer cells. Acta Biochim Biophys Sin (Shanghai). 2023 Mar;55(8):1288-300.
crossref pmid pmc pdf
38. Cheng Y, Chen J, Shi Y, Fang X, Tang Z. MAPK signaling pathway in oral squamous cell carcinoma: biological function and targeted therapy. Cancers (Basel). 2022 Sep;14(19):4625.
crossref pmid pmc
39. Long Y, Guo J, Chen J, Sun J, Wang H, Peng X, et al. GPR162 activates STING dependent DNA damage pathway as a novel tumor suppressor and radiation sensitizer. Signal Transduct Target Ther. 2023 Feb;8(1):48.
crossref pmid pmc pdf
40. Zhou W, Jiang Z, Li X, Xu Y, Shao Z. Cytokines: shifting the balance between glioma cells and tumor microenvironment after irradiation. J Cancer Res Clin Oncol. 2015 Apr;141(4):575-89.
crossref pmid pdf
41. Drobin K, Marczyk M, Halle M, Danielsson D, Papiez A, Sangsuwan T, et al. Molecular profiling for predictors of radiosensitivity in patients with breast or head-and-neck cancer. Cancers (Basel). 2020 Mar;12(3):753.
crossref pmid pmc
42. Pal SK, Nguyen CT, Morita KI, Miki Y, Kayamori K, Yamaguchi A, et al. THBS1 is induced by TGFB1 in the cancer stroma and promotes invasion of oral squamous cell carcinoma. J Oral Pathol Med. 2016 Nov;45(10):730-9.
pmid
43. Ramirez-Cosmes A, Reyes-Jimenez E, Zertuche-Martinez C, Hernandez-Hernandez CA, Garcia-Roman R, Romero-Diaz RI, et al. The implications of ABCC3 in cancer drug resistance: can we use it as a therapeutic target. Am J Cancer Res. 2021 Sep;11(9):4127-40.
pmid pmc
44. Low HB, Wong ZL, Wu B, Kong LR, Png CW, Cho YL, et al. DUSP16 promotes cancer chemoresistance through regulation of mitochondria-mediated cell death. Nat Commun. 2021 Apr;12(1):2284.
crossref pmid pmc pdf
45. Li Y, Pan M, Lu T, Yu D, Liu C, Wang Z, et al. RAF1 promotes lymphatic metastasis of hypopharyngeal carcinoma via regulating LAGE1: an experimental research. J Transl Med. 2022 Jun;20(1):255.
crossref pmid pmc pdf
46. Chen J, Fujii K, Zhang L, Roberts T, Fu H. Raf-1 promotes cell survival by antagonizing apoptosis signal-regulating kinase 1 through a MEK-ERK independent mechanism. Proc Natl Acad Sci U S A. 2001 Jul;98(14):7783-8.
crossref pmid pmc
47. Jing Y, Jin Y, Wang Y, Chen S, Zhang X, Song Y, et al. SPARC promotes the proliferation and metastasis of oral squamous cell carcinoma by PI3K/AKT/PDGFB/PDGFRβ axis. J Cell Physiol. 2019 Sep;234(9):15581-93.
crossref pmid pdf
48. Tsuchida R, Osawa T, Wang F, Nishii R, Das B, Tsuchida S, et al. BMP4/Thrombospondin-1 loop paracrinically inhibits tumor angiogenesis and suppresses the growth of solid tumors. Oncogene. 2014 Jul;33(29):3803-11.
crossref pmid pdf
49. Cheng YC, Chang KW, Pan JH, Chen CY, Chou CH, Tu HF, et al. Cold atmospheric plasma jet irradiation decreases the survival and the expression of oncogenic miRNAs of oral carcinoma cells. Int J Mol Sci. 2023 Nov;24(23):16662.
crossref pmid pmc
50. Dai X, Bazaka K, Richard DJ, Thompson ER, Ostrikov KK. The emerging role of gas plasma in oncotherapy. Trends Biotechnol. 2018 Nov;36(11):1183-98.
crossref pmid
Editorial Office
Korean Society of Otorhinolaryngology-Head and Neck Surgery
103-307 Park Tower officetel, Yongsan-dong 5-ga, Yongsan-gu, Seoul 04385, Korea
TEL: +82-2-711-9091   FAX: +82-2-3487-6603   E-mail: editor.eceo@gmail.com
Copyright © Korean Society of Otorhinolaryngology-Head and Neck Surgery.                 Developed in M2PI
Close layer
prev next