Abstract
Objectives
Disulfidptosis is a novel form of cell death, whose modulation in tumor cells may present a promising therapeutic strategy for cancer treatment. However, the role of disulfidptosis-related long non-coding RNAs (lncRNAs) in non-small cell lung carcinoma (NSCLC) remains poorly elucidated. This study aims to investigate the prognostic significance of disulfidptosis-related lncRNAs (DRLs) and reveal their relationship to the immune microenvironment of NSCLC.
Methods
DRLs were identified through co-expression analysis of NSCLC transcriptomic data obtained from the Genomic Data Commons (GDC) data portal. The DRLs prognostic signature (DRLPS) was established using the least absolute shrinkage and selection operator (LASSO) and Cox regression analyses. Samples were separated into high-DS and low-DS groups based on the median disulfidptosis score (DS) of DRLPS. Integrated analyses were then implemented to unveil the association between DRLs and NSCLC microenvironment. These involved the evaluation of functional enrichments, immune cell infiltrations, genetic alterations, and drug sensitivity.
Results
A prognostic signature was developed based on six prognostic DRLs, which are AL606489.1, LINC00857, AP003555.1, AP000695.1, AC113346.1, and LINC01615. The Kaplan–Meier survival curves demonstrated the significant association between DRLPS and NSCLC prognosis. The functional enrichment assessment revealed the pivotal involvement of DRLs in immune regulation and metabolism in NSCLC. The low-DS and high-DS subgroups of NSCLC patients exhibited distinct differences in terms of immune infiltration and tumor mutation burden. The potential to predict immunotherapy benefit and drug sensitivity in NSCLC treatments was observed in DRLPS.
Conclusions
In this study, disulfidptosis-related lncRNAs were identified and their roles in NSCLC were revealed. A novel prognostic signature with the potential to predict drug response in NSCLC treatment was developed.
Introduction
Non-small cell lung carcinoma (NSCLC), a prevalent and fatal form of lung cancer, accounts for around 15 % of global cancer-related fatalities. The five-year survival rate for individuals diagnosed with advanced-stage NSCLC is less than 20 % [1]. Conventionally, NSCLC patients who undergo treatments such as surgical resection, chemotherapy, and radiotherapy are susceptible to relapse, metastasis, and drug resistance. Emerging strategies have been focusing on the crucial impact of genomic aberrations on tumor cell proliferation and development. There is growing evidence indicating that the interactions between tumor cells and the immune microenvironment may provide a novel approach to NSCLC treatment [2]. In recent years, immunotherapy has shown remarkable clinical benefits for a minor proportion of patients. However, its widespread application remains limited, primarily due to tumor heterogeneity [3]. Thus, investigating effective biomarkers in NSCLC is of paramount significance.
In recent years, various regulatory modalities for cell death, such as ferroptosis, cuproptosis, necroptosis and pyroptosis have been reported [4], [5], [6]. These findings boosted the exploration of biomarkers for the prognosis and therapeutics in cancer treatment. Lately, a novel type of cellular programmed death, known as disulfidptosis, has been proposed [7]. Disulfide refers to an organic compound that contains disulfide bonds, which are primarily found in the cystine of living organisms. The accumulation of intracellular disulfide can result in elevated levels of reactive oxygen species (ROS) in tumor cell mitochondria, ultimately leading to cell death. SLC7A11 is responsible for promoting the cellular absorption of cysteine, which in turn helps to maintain appropriate levels of intracellular glutathione [8]. This process plays a crucial role in preventing apoptosis caused by oxidative stress. However, overexpression of SLC7A11 in tumor cells causes cystine to be converted to the more soluble cysteine, and this induces cell death. This process relies on the pentose phosphate metabolic pathway (PPP) and requires large amounts of nicotinamide adenine dinucleotide phosphate (NADPH). Restricting glucose supply to neoplastic cells increases the accumulation of cystine, in addition to intensifying the consumption of glutathione and NADPH [9]. Therefore, disulfidptosis modulates apoptosis by inducing the aberrant accumulation of disulfides in cells that overexpress SLC7A11 under glucose-deprived conditions. Regulating disulfidptosis in cancer cells could potentially serve as a promising therapeutic approach for NSCLC treatment.
Despite the increasing knowledge around the pathogenesis of NSCLC, there remains a dearth of research on gene interactions and their regulatory mechanisms. Long non-coding RNAs (lncRNAs) refer to non-coding RNAs with a transcript length exceeding 200 bp. They have an essential function in regulating gene expression at multiple levels. lncRNA interacts with DNA, RNA, and proteins to produce inhibitory or enhancing effects. Accumulating research revealed that lncRNAs are involved in various biological processes associated with tumorigenesis, and these include cell proliferation, apoptosis, chromatin modification, and DNA methylation [10], [11], [12]. Recently, numerous lncRNA biomarkers associated with cell death were identified to enhance prognosis and therapeutics in lung cancer. For instance, LINC00336 regulates the ferroptosis of lung cancer cells by posing as an endogenous sponge of MIR6852 [13]. The cuproptosis-related lncRNA MIR31HG shows oncogenic features in lung adenocarcinoma [14]. Diao et al. constructed a novel lncRNAs model associated with necroptosis to predict lung cancer prognosis [15]. However, the role of disulfidptosis-related lncRNAs of NSCLC is still yet to be investigated.
This study aims to evaluate the prognostic significance of disulfidptosis-related lncRNAs (DRLs) and elucidate their potential in relation to the tumor microenvironment in NSCLC. A machine learning approach was employed to develop the DRLs prognostic signature. The prognostic performance of this signature was estimated using ROC analysis, principal component analysis (PCA), nomogram construction, and calibration curve fitting. Further confirmation was carried out in both the validation and the entire cohort. Additionally, a comprehensive analysis that included functional assessment, immune infiltration, genomic mutation, and drug sensitivity was subsequently conducted to assess the association between DRLs and NSCLC. These findings lay a theoretical foundation for comprehending the role of DRLs in NSCLC, and may unleash promising biomarkers for predicting the prognosis of NSCLC and guide therapeutics for its treatment.
Methods
Data source and processing
A total of 1,096 patients diagnosed with NSCLC were enrolled in this study. The transcriptome profile and somatic mutation datasets were obtained from the Genomic Data Commons data portal (https://gdc.cancer.gov/access-data/gdc-data-portal), along with the clinical information, including age, sex, survival status, follow-up time, smoking history, pathological stage, and TNM stage [16]. The annotations for lncRNAs were retrieved from the National Center for Biotechnology Information (NCBI, https://www.ncbi.nlm.nih.gov/) [17]. Genes known to be markers for disulfidptosis (CAPZB, DSTN, ACTB, MYL6, TLN1, FLNB, FLNA, IQGAP1, MYH10, MYH9, ACTN4, PDLIM1, CD2AP, INF2, SLC7A11, NOX4, NOXA1, and ENOX2) were acquired from previous literature [7]. The overview for this study is depicted in Figure 1.

Overview of this study.
Screening disulfidptosis-related lncRNAs
The co-expression analysis between and disulfidptosis marker genes and lncRNAs was conducted using Pearson correlation to identify DRLs. The threshold was established where the correlation coefficient’s absolute value >0.4 and p-value <0.01. The Sankey plot based on these findings was visualized using the “ggalluvial” R package (version 0.12.5) [18].
Construction of DRLs prognostic signature (DRLPS)
The R package “caret” (version 6.0-94) was employed to randomly separate NSCLC samples into the training and validation cohorts. The prognostic significance of DRLs was assessed through a univariate Cox proportional hazards regression analysis, with a significance level set at p <0.05. Afterward, the LASSO algorithm was executed using the “glmnet” package (version 4.1-8) in the R software. A penalized function was developed, incorporating 10-fold cross-validation, to identify the optimal prognostic DRLs. Subsequently, the DRLs prognostic signature was developed using a multiple stepwise Cox regression analysis [19]. Meanwhile, the scoring system for disulfidptosis was established based on this prognostic signature, and the formula was as follows:
where βi represents the hazard ratio coefficient of Cox regression analysis and EXP (lncRNAi) means the expression of each lncRNA of DRLPS. Consequently, the patient was labelled with a DS.
Evaluation of prognostic ability
NSCLC patients were categorized into high- and low-DS groups based on the median DS. The survival analysis was conducted to assess the prognostic ability of DRLPS in NSCLC. ROC analysis using R package “survivalROC” (version 1.0.3.1) was employed to evaluate the predictive accuracy of overall survival (OS) at 1-, 3-, and 5-years. Validating the reliability of the prognostic signature was done using PCA. The “scatterplot3D” R package (version 0.3-44) was applied for visualization [20].
The independent prognostic performance of DRLPS was assessed using univariate and multivariate Cox regression methods among clinical features. A nomogram was constructed, incorporating the DS, age, sex, smoking history, as well as the TNM and pathological stages. The calibration curve was used to detect the accuracy of the nomogram.
Functional enrichment assessment
The gene set enrichment analysis (GSEA) was used to assess the differences in biological function between the low- and high-DS cohorts. Gene ontology (GO) and REACTOME pathways were involved in the analysis using R package “clusterProfiler” (version 4.6.2) [21]. The analysis of GO terms reveals the enriched functions of gene clusters in relation to the biological processes. The REACTOME analysis uncovers the potential signaling pathways that the genes interacted through. A p-value below 0.05 was deemed to represent statistical significance.
Association between DRLPS and immune microenvironment
ESTIMATE (https://bioinformatics.mdanderson.org/estimate/) is a commonly employed tool for evaluating the levels of stromal and immune cells in tumor tissues. The potential impact of DRLs on the tumor microenvironment was investigated by determining the stromal and immune score using the transcriptome profile of NSCLC [22]. The R package “immunodeconv” (version 2.1.0) was applied to calculate immune cell abundance between low- and high-DS groups [23]. The immune-related functions were examined using the “ssGSEA” algorithm (version 1.46.0) [24].
Prediction of immunotherapy and drug sensitivity
A practical web application known as Tumor Immune Dysfunction and Exclusion (TIDE, http://tide.dfci.harvard.edu/) has been developed to assess the efficacy of immunotherapy for individuals [25]. The TIDE score reflects the effectiveness of treatment with anti-PD1 and anti-CTLA4 drugs. The higher the TIDE scores, the greater the likelihood that cancer patients would benefit from immunotherapy. To evaluate the potential utility of DRLPS for NSCLC treatment, the response of the patients in the low- and high-DS groups to the commonly used medicines in NSCLC therapy was assessed. The half-maximal inhibitory concentration (IC50) for these drugs was calculated using the R package “oncoPredict” (version 0.2) [26].
Genomic alteration evaluation
The R package ‘maftools’ (version 2.14.0) was employed for the visualization of the landscape of genomic variations and calculation of tumor mutation burden (TMB) for NSCLC patients in high- and low-DS groups [27]. The joint analysis of DS and TMB was performed using Kaplan–Meier survival analysis to further evaluate the prognostic impact on the NSCLC patients.
RT-PCR assay
The expression levels of six prognostic DRLs in lung cancer cells were determined using Reverse Transcription-Polymerase Chain Reaction (RT-PCR). The primer sequences are provided in Table S1. The TRizol reagent (TIANGEN, Beijing, China) was utilized to extract total RNA from A549, NCI-H1975, and BEAS-2B cells. Subsequently, the obtained RNA was reverse transcribed into cDNA according to the manufacturer’s guidelines (Thermo Scientific, Waltham, USA). For PCR amplification, a reaction mixture (20 μL) was prepared with 10 μL of 2× PerfectStart Green qPCR SuperMix (TransGen, AQ601-04, Beijing, China), 2 μL of cDNA diluted in nuclease-free water, and 1 μL of a 10 μM primer with nuclease-free water. ABI Q1 sequencers were used for amplification and detection procedures. GAPDH was utilized as the reference gene. Quantifying the transcript was conducted using the 2−ΔΔCT method.
Statistical analysis
The R software (version 4.2.3) was used for performing the statistical analyses. The distribution of samples between the training and validation cohorts was assessed by the Chi-square test. Pearson correlation coefficient with a cutoff of |Cor| >0.4 and p-value <0.01 was used to identify DRLs. The quantitative comparison between the low- and high-DS groups was conducted using the Wilcoxon rank-sum test. The Kaplan–Meier survival analysis was implemented to estimate the predictive ability for NSCLC prognosis, followed by the log-rank p-value. The accuracy of prognostic prediction was assessed through ROC analysis. Univariate and multivariate regression analyses were performed using Cox proportional hazard models. Statistical significance was considered when the p-value fell below 0.05.
Results
Identifying disulfidptosis-related lncRNAs
The boxplot showed a significant differential expression of all disulfidptosis marker genes between the NSCLC tumor and adjacent normal tissue (Figure 2A). The cumulative rate at which these genes were altered reached 23.28 % (Figure 2B), and this highlights the critical role that disulfidptosis plays in tumorigenesis and cancer development. The Pearson correlation analysis revealed a strong association between 232 lncRNAs and 16 markers that are related to disulfidptosis. The relationship between these candidate DRLs and disulfidptosis marker genes was illustrated in the Sankey diagram (Figure 2C).

Identification of disulfidptosis-related lnRNAs in NSCLC. (A) The expression of disulfidoptosis marker genes in the tumor and adjacent normal tissue of NSCLC. (B) Mutational status of disulfidptosis marker genes of NSCLC. (C) Sankey diagram between 16 disulfidptosis marker genes and 232 disulfidptosis-associated lncRNAs. (D) The correlation between disulfidptosis marker genes and six lncRNAs of DRLPS.
Establishing the DRLs prognostic signature
A joint approach was employed in this analysis. The univariate Cox analysis screened 21 lncRNAs based on the extent to which they were associated with disulfidptosis, as well as their correlation with prognosis in NSCLC (Figure 3A). Subsequently, the LASSO regression analysis identified 14 optimal features among these lncRNAs (Figure 3B and C). Finally, a prognostic signature that consisted of six DRLs (AL606489.1, LINC00857, AP003555.1, AP000695.1, AC113346.1, and LINC01615) was established using the multiple stepwise Cox regression analysis. The survival analysis revealed a significant association between each lncRNA of DRLPS and the prognosis of NSCLC (Figure S1A–F). The RT-PCR assay demonstrated an obvious up-regulation of AL606489.1, LINC00857, AP000695.1, and AC113346.1 in NSCLC cells, while the downregulation of LINC01615 was observed in NCI-H1975 cells (Figure S2). Based on the heatmap of Pearson correlation coefficient, correlation was noted between disulfidptosis marker genes and these prognostic lncRNAs (Figure 2D). DS was calculated for each NSCLC patient based on the regression coefficient and expression of lncRNAs. DS=0.6578*EXPAL606489.1+0.8820 *EXPLINC00857+0.5217*EXPAP003555.1+0.8724*EXPAP000695.1+0.1019 *EXPAC113346.1−0.4471*EXPLINC01615.

Construction of disulfidptosis-related lncRNAs prognostic signature in NSCLC. (A) Identification of prognostic DRLs using univariate Cox regression analysis. (B, C) The LASSO algorithm was utilized to determine the optimal features among 21 prognostic DRLs. (D) Kaplan–Meier survival curve of low- and high-DS cohorts in the training set. (E) The survival statistics distribution with disulfidptosis scores in the training set. (F) Heatmap of prognostic six lncRNAs of DRLPS in the training set. (G) Kaplan–Meier survival curve of low- and high-DS groups in the validation set. (H) The survival statistics distribution with disulfidptosis scores in the validation set. (I) heatmap of DRLs of six lncRNAs of DRLPS in the validation set.
There were no significant differences (p>0.05, Table 1) observed in the clinical characteristics between the training and validation cohorts. This ensured the confidence of the classified groups in the subsequent analysis. Patients in the training cohort were categorized into low- and high-DS groups based on the median DS. The prognostic performance of DRLPS in NSCLC was evaluated through survival analysis. The survival curves indicated that the patients with high-DS were remarkably associated with poor overall survival (Figure 3D). The survival statistics distribution with disulfidptosis scores is illustrated in Figure 3E, which indicates a rise in deaths with increasing DS. Moreover, the expression level of DRLPS lncRNAs was obviously elevated in high-DS group (Figure 3F). These findings were consistently observed across the validation and entire cohorts (Figures 3G–I and 4A–C), thus confirming the accuracy of the established prognostic signature. Compared to disulfidptosis-related genes and lncRNAs, the prognostic DRLs exhibited superior efficacy in distinguishing between high- and low-DS populations (Figure 4D–G). This further validated the reliability of DRLPS.
Characteristics of non-small cell lung carcinoma patients.
Clinical features | Entire cohort (n=1096) | Training cohort (n=548) | Validation cohort (n=548) | p-Value |
---|---|---|---|---|
Age | 0.6465 | |||
>60 | 773 (70.6 %) | 392 (35.8 %) | 381 (34.8 %) | |
≤60 | 295 (26.9 %) | 141 (12.8 %) | 154 (14.1 %) | |
Unknown | 28 (2.5 %) | 15 (1.3 %) | 13 (1.2 %) | |
|
||||
Sex | 0.177 | |||
|
||||
Female | 451 (41.2 %) | 237 (21.6 %) | 214 (20.6 %) | |
Male | 645 (58.8 %) | 311 (28.4) | 334 (30.4 %) | |
|
||||
Stage | 0.596 | |||
|
||||
Stage I | 569 (51.9 %) | 287 (26.1 %) | 282 (25.8 %) | |
Stage II | 298 (27.2 %) | 156 (14.2 %) | 142 (13.0 %) | |
Stage III | 181 (16.5 %) | 85 (7.8 %) | 96 (8.7 %) | |
Stage IV | 35 (3.2 %) | 15 (1.4 %) | 20 (1.8 %) | |
Unknown | 13 (1.2 %) | 5 (0.5 %) | 8 (0.7 %) | |
|
||||
T stage | 0.228 | |||
|
||||
T1 | 308 (28.1 %) | 166 (15.1 %) | 142 (13.0 %) | |
T2 | 620 (56.5 %) | 291 (26.6 %) | 329 (29.9 %) | |
T3 | 122 (11.1 %) | 64 (5.8 %) | 58 (5.3 %) | |
T4 | 43 (3.9 %) | 25 (2.3 %) | 18 (1.6 %) | |
Unknown | 4 (0.4 %) | 2 (0.2 %) | 2 (0.2 %) | |
|
||||
M stage | 0.533 | |||
|
||||
M0 | 813 (74.2 %) | 406 (37.0 %) | 407 (37.2 %) | |
M1 | 34 (3.1 %) | 14 (1.3 %) | 20 (1.8 %) | |
Unknown | 249 (22.7 %) | 128 (11.7 %) | 121 (11.0 %) | |
|
||||
N stage | 0.283 | |||
|
||||
N0 | 698 (63.7 %) | 350 (31.9 %) | 348 (31.8 %) | |
N1 | 238 (21.7 %) | 118 (10.8 %) | 120 (10.9 %) | |
N2 | 123 (11.2 %) | 58 (5.3 %) | 65 (6.9 %) | |
N3 | 7 (0.7 %) | 2 (0.2 %) | 5 (0.5 %) | |
Unknown | 30 (2.7 %) | 20 (1.8 %) | 10 (0.9 %) | |
|
||||
Smoke | 0.779 | |||
|
||||
Never | 269 (24.5 %) | 137 (12.5 %) | 132 (12.0 %) | |
Ever | 827 (75.5 %) | 411 (37.5 %) | 416 (38.0 %) | |
|
||||
Histopathology | ||||
|
||||
LUAD | 594 (54.2 %) | 310 (28.3 %) | 284 (25.9 %) | |
LUSC | 502 (45.8 %) | 238 (21.7 %) | 264 (24.1 %) |
-
LUAD, lung adenocarcinoma. LUSC, lung squamous carcinoma.

Validation of the prognostic signature using the entire cohort and PCA. (A) Kaplan–Meier survival curve of the low- and high-DS groups in the entire cohort. (B) The survival statistics distribution with disulfidptosis scores in the entire set. (C) Heatmap of the DRLPS lncRNAs in the entire set. Scatter plot of PCA between low- and high-DS subgroups based on (D) all genes, (E) disulfidptosis-associated genes, (F) DRLs, and (G) prognostic DRLs.
Findings from the analysis of the association between DS and clinical factors revealed that DS significantly affects age, pathological stage, histopathology, as well as stages T and M (Table 2).
Correlation between disulfidptosis scores and clinical characteristics of NSCLC patients.
Characteristics | No. of patients | Disulfidptosis score | Chi-square value | p-Value | |
---|---|---|---|---|---|
Low | High | ||||
Age, year | 4.21 | 0.04 | |||
|
|||||
>60 | 773 | 402 | 371 | ||
≤60 | 295 | 132 | 163 | ||
|
|||||
Sex | 0.654 | 0.418 | |||
|
|||||
Male | 645 | 316 | 329 | ||
Female | 451 | 233 | 218 | ||
|
|||||
Stage | 17.6 | 0.00025 | |||
|
|||||
Stage I | 569 | 306 | 263 | ||
Stage II | 298 | 150 | 148 | ||
Stage III | 181 | 77 | 104 | ||
Stage IV | 35 | 8 | 27 | ||
|
|||||
T stage | 16.2 | 0.001 | |||
|
|||||
T1 | 308 | 178 | 130 | ||
T2 | 620 | 302 | 318 | ||
T3 | 122 | 54 | 68 | ||
T4 | 43 | 13 | 30 | ||
|
|||||
M stage | 10.9 | 0.00095 | |||
|
|||||
M0 | 813 | 415 | 398 | ||
M1 | 34 | 7 | 27 | ||
|
|||||
N stage | 5.02 | 0.17 | |||
|
|||||
N0 | 698 | 354 | 344 | ||
N1 | 238 | 119 | 119 | ||
N2 | 127 | 51 | 76 | ||
N3 | 7 | 4 | 3 | ||
|
|||||
Smoke | 0.769 | 0.381 | |||
|
|||||
Never | 269 | 128 | 141 | ||
Ever | 827 | 421 | 406 | ||
|
|||||
Histopathology | 36.8 | 1.3e-9 | |||
|
|||||
LUAD | 594 | 247 | 347 | ||
LUSC | 502 | 302 | 200 |
Disulfidptosis score serves as an independent risk factor
The ROC curves demonstrated the accuracy of DRLPS in predicting the overall survival at 1-, 3-, and 5-year periods, with the AUC values of 0.679, 0.711, and 0.740, respectively (Figure 5A). Meanwhile, the DS demonstrated a superior prognostic value than other clinical characteristics (Figure 5B). To further evaluate the prognostic significance of DRLPS in comparison with clinical characteristics, both univariate and multivariate Cox regression analyses were conducted. The univariate Cox analysis demonstrated a significant correlation between the DS (HR=1.36, Cox p=0.0018), pathological stage (HR=1.91, Cox p<0.001), N stage (HR=1.67, Cox p<0.001), T stage (HR=1.60, Cox p<0.001), age (HR=1.24, Cox p=0.029) and the prognosis of NSCLC (Figure 5C). The findings from the multivariate Cox regression approach were as follows: DS (HR=1.32, Cox p=0.0049), pathological stage (HR=1.59, Cox p=0.0025), T stage (HR=1.60, Cox p=0.041) and age (HR=1.32, Cox p=0.0055). These results also showed the predictive ability of NSCLC prognosis (Figure 5D). These findings revealed the significance of high DS as an independent risk indicator for unfavorable outcomes. Additionally, the nomogram (Figure 5E) was employed to quantify the prognostic performance of DRLPS based on the DS and clinical factors. The reliability of this nomogram was confirmed by the calibration plot (Figure 5F).

Prognostic performance of DRLPS in NSCLC. (A) Time-dependent ROC curves depicting overall survival at 1, 3, and 5 years in NSCLC. (B) ROC curves of overall survival for disulfidptosis score and clinical factors in NSCLC. (C) Univariate Cox regression analysis of disulfidptosis score and clinical characteristics in NSCLC. (D) Multivariate Cox regression analysis of disulfidptosis score and clinical characteristics in NSCLC. (E) establishment of a nomogram utilizing disulfidptosis score and clinical features. (F) The calibration curves of the nomogram.
DRLPS is related to the immune mediations and metabolism in NSCLC
The GO analysis revealed that the gene cluster of the low-DS group was primarily involved in the biological processes that are relevant for activating immune responses and T-cells, leukocyte-mediated immunity, as well as the regulation of lymphocytes. On the contrary, genes with high DS were found to be involved in the functions such as oxidative phosphorylation, nuclear chromosome segregation, mitochondrial respiratory chain complex assembly, meiotic cell cycle, and cytokinetic process (Figure 6A). With respect to the REACTOME pathways, T cell receptor (TCR) signaling, programmed death-1 receptor (PD-1) signaling, CTLA4 inhibitory signaling, interleukin-10 signaling, and signaling by interleukin were the prominent pathway terms in the low-DS cohort. It’s important to note that the enrichment of several pathways linked to metabolism was noted in the high-DS group (Figure 6B). These include selenoamino acid metabolism, O-like glycosylation of mucins, mitochondrial translation, respiratory electron transport, the citric acid TCA cycle, and termination of O-glycan biosynthesis. These results unveiled the correlation between DRLs and immune regulation as well as metabolism in the microenvironment of NSCLC. Therefore, it can be extrapolated that these DRLs may serve as the therapeutic targets for NSCLC treatment.

Functional enrichment analysis of DRLPS. (A) Enriched GO terms of biological process in the high- and low-DS groups. (B) Enriched REACTOME pathways in the high- and low-DS groups.
A significant association between DRLs and immune infiltration
Given the potential role of DRLs function in NSCLC immunity, the ESTIMATE and ssGSEA were performed to evaluate the intra-tumoral immune cell infiltration between the high- and low-DS groups. The ESTIMATE analysis revealed higher scores of immune and stromal cells in the low-DS group compared to the high-DS group (Figure 7A). ssGSEA results uncovered the immune cell populations of NSCLC patients (Figure 7B). A remarkable difference was observed in the two NSCLC subgroups with regard to naive B-cells, activated natural killer (NK) cells, activated/resting mast cells, macrophages M0/M1, activated dendritic cells, and various functional T-cells (Figure 7C). These findings demonstrated the significant correlation of DRLs with immune cell infiltration. Additionally, patients in the low-DS group exhibited heightened immune function activity, as evidenced by increased MHC class I level, type II interferon (IFN) response, cytolytic activity, inflammation promotion, CCR signaling, and co-stimulation/suppression of antigen-presenting cells (APCs) and T-cells (Figure 7D). This may contribute to anti-tumor potency in the NSCLC microenvironment.

Exploration of the association between DRLPS and immune microenvironment of NSCLC. (A) Comparison of stromal and immune infiltration levels between the high- and low-DS groups. (B) Immune cell population of NSCLC patients in the high- and low-DS groups. (C) Comparison of tumor-infiltrated immune cells between the high- and low-DS groups. (D) Heatmap of immune-related activity in the high- and low-DS groups of NSCLC patients. (E) Expression difference of the immune checkpoint markers between the high- and low-DS groups. (F) Comparison of TIDE score in the high- and low-DS cohorts. *p<0.05, **p<0.01, and ***p<0.001. ns, not significant.
Promising role of DRLPS in predicting immunotherapy and drug sensitivity
Comparisons on the expression levels of immune checkpoint genes in groups with low- and high-DS were done (Figure 7E). Within the cohort with a low-DS, the expression levels of 24 genes were significantly higher compared to those in the group with a high DS. Notably, PDCD1, CD274, and CTLA4, which are commonly used in clinical treatments, were among these 24 genes. To investigate the potential predictive significance of DRLPS in immunotherapy, the TIDE score was used to evaluate the impact of immunosuppressive factors on patients with NSCLC. The higher TIDE score observed in the group with a lower DS suggests that these patients exhibited heightened responsiveness to immunotherapy that involved anti-PD1 and anti-CTLA4 drugs (Figure 7F), suggesting the potential role of DRLPS in predicting the immunotherapeutic efficacy. Afterward, the relationship between the DS and drug sensitivity for the NSCLC treatment was also explored. The IC50s of 16 medicines, including bleomycin, doxorubicin, etoposide, gemcitabine, methotrexate, temozolomide, alectinib, dabrafenib, trametinib, cabozantinib, afatinib, panatinib, AZD6244, MS-275, CGP43231, and ruxolitinib exhibited significant negative correlation with the DS (Figure S3). Patients with a high DS exhibited enhanced drug susceptibility, as evidenced by lower IC50 values, in contrast to patients with low DS (Figure 8). The findings demonstrated the possibility of using DRLPS in guiding the clinical decisions of these medicines for NSCLC patients.

The significant association of drug sensitivity (IC50) with disulfidptosis score in NSCLC treatment. Notes: ** p value<0.01; *** p value <0.001.
Tumor mutation burden and genomic variation landscape
Increasing studies pointed out that genomic alterations are closely related to patients’ sensitivity to tumor treatment. Patients with a high tumor mutation burden may contain large amounts of neoantigens, which activate the immune system to target cancerous cells, thereby promoting apoptosis. In this study, the TMB levels and genomic variation landscape were involved in the analyses to investigate the relationship between DRLs and genomic alterations in NSCLC. A higher TMB level was noted among patients exhibiting low DS (p=0.029, Figure 9A). Apparently, higher TMB levels significantly correlated with an increased survival probability for NSCLC patients (p=0.02, Figure 9B). Additionally, the joint survival analysis combined with the DS and TMB level was also performed and the result revealed that these two indicators had a strong synergistic effect in predicting NSCLC prognosis (p=0.00071, Figure 9C). The waterfall plot of genetic variation displayed the mutation landscape for the top 20 altered-frequency genes in the low- and high-DS cohorts (Figure 9D and E). A higher alteration frequency was found in the group with a low DS, and TP53 exhibited the highest proportion of mutations in both subgroups of NSCLC.

The correlation between the disulfidptosis score and tumor mutation burden in NSCLC. (A) Comparison of TMB between the low- and high-DS groups in NSCLC. (B) Survival curve of the low- and high-TMB groups. (C) Survival analysis combined with DS and TMB level for NSCLC patients. (D, E) Genomic alteration landscape of the top 20 mutated genes in the low- and high-DS cohorts.
Discussion
The fatality rate of non-small cell lung cancer remains disproportionately high, considering that almost 50 % of individuals receive a diagnosis during a later stage. Effective biomarkers for NSCLC treatment are still lacking due to the limited understanding of the underlying mechanisms that drive the progression of malignancies. Therefore, discovering novel therapeutic targets holds significant value.
Metabolic alteration is a defining feature in cancer cells and also represents an exploitable vulnerability for tumor therapy. Given the intractable nature of drug-resistant cancer cells, metabolism-related cell death is increasingly gaining attention as a promising strategy for cancer treatment [28]. Previous studies demonstrated the involvement of pyroptosis, cuproptosis and ferroptosis in the NSCLC progression, with novel biomarkers continually being discovered based on these mechanisms [4, 5, 29]. It is noteworthy that disulfidptosis has recently emerged as a novel target for metabolic cancer therapy, thereby attracting significant attention. On the other hand, lncRNAs have emerged as important prognostic indicators in NSCLC and hold potential as promising molecular targets for therapeutic intervention. However, the regulatory mechanism of disulfidoptosis in NSCLC remains poorly understood, particularly with regard to the unexplored field of DRLs. Hence, the primary objective of this study is to explore associations between DRLs and NSCLC and uncover potential biomarkers. In this investigation, DRLs were identified using rigorous criteria, prior to establishing a 6-lncRNAs prognostic signature that consists of AL606489.1, LINC00857, AP003555.1, AP000695.1, AC113346.1, and LINC01615. The prognostic outcome of NSCLC showed a significant correlation with the disulfidptosis score of DRLPS, and the predictive accuracy was assessed using the AUC of ROC analysis. The analyses of univariate and multivariate Cox indicated that a high-DS independently predicts NSCLC prognosis. Afterward, the robustness of this prognostic model was also affirmed in both validation and entire cohorts. Among the six lncRNAs of DRLPS, previous studies have demonstrated the prognostic significance of AL606489.1 in relation to cuproptosis and ferroptosis in lung cancer [14, 30]. LINC00857 has been reported to promote pancreatic cancer tumorigenesis through the regulatory axis of miR-150-5p/E2F3 [31]. AP003555.1 is a prognostic lncRNA linked to oxidative stress in colon cancer. AP000695.1 can be potentially used as a prognostic indicator for forecasting survival results in individuals diagnosed with gastric cancer [32, 33]. LINC01615 is implicated in the progression of carcinogenesis and metastasis in several cancers, including hepatocellular carcinoma, colorectal cancer, and triple-negative breast cancer [34], [35], [36]. As for AC113346.1, there has been no research conducted on it yet, indicating a need for further exploration. Recently, Wang et al. [37] performed a multifaceted analysis of transcriptional and genetic modifications in regulators of disulfidptosis in lung adenocarcinoma using 10 distinct machine learning algorithms. Singh et al. [38] identified GPRIN2, KCNJ12, and TEKT4 as novel biomarkers in NSCLC through whole-exome sequencing. These studies motivated our initiative to explore potential therapeutic targets for NSCLC treatment, shedding light on their crucial role within the tumor microenvironment. In addition, the data from KnockTF and LncSEA were integrated to investigate potential transcription factors linked to disulfidptosis [39, 40]. The Venn diagram (Figure S4A) showed eight regulators (GTF2B, ELK3, ILF2, SRSF7, NCOR2, TOP2A, MYT1L, and ZFX) may be involved in the interaction network regarding disulfidptosis, which deserves further investigation (Figure S4B).
The crucial role of the immune microenvironment in the pathogenesis of NSCLC has been demonstrated. With a deepening comprehension of the correlation between the tumor microenvironment and immune response, novel targets for prognosis and therapeutics are being identified. The findings from this study indicated that DRLPS was clearly linked to immune functions. Distinct differences in the immune infiltration of NK cells, mast cells, macrophages, dendritic cells, T-cells, B-cells, between low- and high-DS groups were noted. More emerging evidence highlights the vital roles of these immune cells within the tumor microenvironment. Cytotoxic and helper T-cells, along with Tregs, are essential for promoting antitumor response, whereas exhausted T-cells promote cancer cell survival during tumor progression [41]. B-cells can function as APCs to augment the immune response of T-cells in the tumor microenvironment [42]. Dendritic cells possess the ability to internalize and present exogenous antigens to CD8 T-cells, thereby facilitating the elimination of tumor cells [43]. Previous studies uncovered the dichotomous roles of macrophages in the tumor microenvironment, with type-1 macrophage promoting immune effects through inflammation. Tumor-associated macrophages (TAMs), also referred to as Type-2 macrophages, promote tumor growth through the inhibition of inflammatory responses [44]. Some scientific reports show that mast cells have the capacity to inhibit the expression of MMP2 and MMP9, and this may serve as a potential mechanism for impeding tumor metastasis [45]. NK cells can establish direct contact with tumor cells and then use perforin and the tumor necrosis factor to induce cell lysis [46]. Exploiting the anti-tumor properties of these immune cells presents a promising direction for cancer therapy, though further investigation is needed.
Immunotherapy is an emerging approach for NSCLC treatment, which exerts anti-tumor effects by impairing the interactions between immune checkpoints, thereby preventing immune escape. Currently, several immune checkpoint blockades (ICBs) have been approved for the clinical management of NSCLC. These include inhibitors targeting PD-1, PD-L1, and CTLA4. Immunotherapy may become a superior option for NSCLC patients who undergo metastasis or relapse. However, the response of individuals to ICBs varies, and the immunotherapy benefit to each patient is contingent on the levels of expression of genes related to immune checkpoint. Therefore, exploring predictive signatures to guide decisions associated with immunotherapy has aroused considerable concern. In this study, it was found that patients with a low DS showed greater sensitivity to anti-PD1/anti-CTLA4 drug treatments, suggesting the potential of DRLPS as a predictor for assessing the effectiveness of immunotherapy in NSCLC. Furthermore, DRLPS also showed the potential to guide the clinical decisions for several medications regarding chemotherapy and targeted therapy. This may facilitate better treatment management for NSCLC patients. The study also revealed that the prognosis of NSCLC patients with EGFR mutations can be predicted by DRLPS (Figure S5), especially those who are responsive to tyrosine kinase inhibitors (TKIs) commonly used in NSCLC treatment.
Current research indicates that disulfidoptosis is associated with alterations in cellular redox status, which can trigger apoptosis in tumor cells by altering the conformation of cytoskeletal proteins [7]. Another study points out that cells expressing high levels of SLC7A11 can inhibit ferroptosis under the condition of glucose deprivation, and this may lead to disulfidptosis [47]. In this study, the functional assessment revealed the biological processes by which DRLs are implicated in NSCLC. Of significance, the dysfunctions of O-like glycosylation and O-glycan biosynthesis may impact the onset and progress of NSCLC. Previous studies highlighted the crucial role of glycosylation in cancer biology. Aberrant glycosylation is intricately linked to the malignant transformation of tumors, and certain anomalous glucose molecules have been identified as cancer biomarkers [48]. For instance, the overexpression of α-GalNAc and ST6GalNAc-I can stimulate the expression of STn and induce C1GALT1C1 mutation, thereby promoting tumor cell invasion [49]. The O-glycosylation pattern of integrins undergoes alterations during tumor cell migration, and the aberrant modulation of glycosylation between VEGFR and galectins may contribute to tumor angiogenesis [50]. Although these potential metabolism-related regulations of DRLs may shed light on their mechanism in NSCLC, there remains a vast unexplored territory between DRLs and tumor microenvironments. TP53 was found to be the most frequently altered gene in two subgroups of NSCLC patients. As a known tumor suppressor, TP53 encodes a protein that responds to diverse forms of cellular stress in order to regulate the expression of target genes, as well as induce apoptosis, DNA repair, and metabolic changes [51]. Therefore, studying the impact of genomic modifications on DRLs may unleash valuable insights into the underlying mechanism in NSCLC.
As a novel form of cell death, disulfidptosis can potentially be used in future cancer therapies. Exploring genes relevant to disulfidptosis may be a promising approach for identifying effective biomarkers for prognosis and therapeutics in NSCLC. The results of this research may offer insights into the mechanism of DRLs in NSCLC. Although the exceptional performance of the established DRLSP in predicting prognosis, guiding immunotherapy, and assessing drug response to chemotherapy and targeted therapy, there are certain limitations in this study. These limitations include analysis bias that may result from a single data source and limited robustness of the predictive signature due to its retrospective nature. Additionally, the causal relationship between DRLs and NSCLC remains unconfirmed; thus, future studies involving both in vivo and in vitro experiments will be necessary for validating these findings.
Conclusions
This study led to the establishment of a prognostic signature using six DRLs in NSCLC. This signature can also be applied in predicting the immunotherapeutic benefit and drug sensitivity. Furthermore, the findings from this study revealed the possible role of DRLs in the NSCLC microenvironment. This implies that these DRLs may serve as therapeutic targets for patients with NSCLC.
Funding source: Science and Technology Program Fund of Huaihua
Award Identifier / Grant number: 2021R3111
Funding source: Fund of Huaihua Technology Innovation platform
Award Identifier / Grant number: 2021R220
Acknowledgments
Not applicable.
-
Research ethics: The authors state that this article does not contain any studies with human participants or animals so exempt from institutional review board approval.
-
Informed consent: Not applicable.
-
Author contributions: Tan RM was responsible for the conception and design of the study. Liu H collected the data and drafted the manuscript. He SH analyzed the data and interpreted the results. Tan LM made revisions to the manuscript, Li MZ created the figures and tables, and Chen C performed the experiment. All authors read the manuscript and approved its submission.
-
Competing interests: The authors declare no potential conflicts of interest.
-
Research funding: This work was supported by the Science and Technology Program Fund of Huaihua (grant No. 2021R3111) and the Fund of Huaihua Technology Innovation platform (grant No. 2021R2206).
-
Data availability: The data presented in this study are available on request from the corresponding author.
References
1. Sung, H, Ferlay, J, Siegel, RL, Laversanne, M, Soerjomataram, I, Jemal, A, et al.. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209–49, https://doi.org/10.3322/caac.21660.Search in Google Scholar PubMed
2. Binnewies, M, Roberts, EW, Kersten, K, Chan, V, Fearon, D, Merad, M, et al.. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med 2018;24:541–50, https://doi.org/10.1038/s41591-018-0014-x.Search in Google Scholar PubMed PubMed Central
3. Lv, B, Wang, Y, Ma, D, Cheng, W, Liu, J, Yong, T, et al.. Immunotherapy: reshape the tumor immune microenvironment. Front Immunol 2022;13:844142, https://doi.org/10.3389/fimmu.2022.844142.Search in Google Scholar PubMed PubMed Central
4. Lei, G, Li, Z, Gan, B. Targeting ferroptosis as a vulnerability in cancer. Nat Rev Cancer 2022;22:381–96, https://doi.org/10.1038/s41568-022-00459-0.Search in Google Scholar PubMed PubMed Central
5. Tang, D, Chen, X, Kroemer, G. Cuproptosis: a copper-triggered modality of mitochondrial cell death. Cell Res 2022;32:417–8, https://doi.org/10.1038/s41422-022-00653-7.Search in Google Scholar PubMed PubMed Central
6. Bertheloot, D, Latz, E, Franklin, BS. Necroptosis, pyroptosis and apoptosis: an intricate game of cell death. Cell Mol Immunol 2021;18:1106–21, https://doi.org/10.1038/s41423-020-00630-3.Search in Google Scholar PubMed PubMed Central
7. Liu, X, Nie, L, Zhang, Y, Yan, Y, Wang, C, Colic, M, et al.. Actin cytoskeleton vulnerability to disulfide stress mediates disulfidptosis. Nat Cell Biol 2023;25:404–14, https://doi.org/10.1038/s41556-023-01091-2.Search in Google Scholar PubMed PubMed Central
8. Koppula, P, Zhang, Y, Zhuang, L, Gan, B. Amino acid transporter SLC7A11/xCT at the crossroads of regulating redox homeostasis and nutrient dependency of cancer. Cancer Commun 2018;38:1–13, https://doi.org/10.1186/s40880-018-0288-x.Search in Google Scholar PubMed PubMed Central
9. Liu, X, Olszewski, KL, Zhang, Y, Lim, EW, Shi, J, Zhang, X, et al.. Cystine transporter regulation of pentose phosphate pathway dependency and disulfide stress exposes a targetable metabolic vulnerability in cancer. Nat Cell Biol 2020;22:476–86, https://doi.org/10.1038/s41556-020-0496-x.Search in Google Scholar PubMed PubMed Central
10. Bridges, MC, Daulagala, AC, Kourtidis, A. LNCcation: lncRNA localization and function. J Cell Biol 2021;220:e202009045, https://doi.org/10.1083/jcb.202009045.Search in Google Scholar PubMed PubMed Central
11. Nojima, T, Proudfoot, NJ. Mechanisms of lncRNA biogenesis as revealed by nascent transcriptomics. Nat Rev Mol Cell Biol 2022;23:389–406, https://doi.org/10.1038/s41580-021-00447-6.Search in Google Scholar PubMed
12. Wang, W, Min, L, Qiu, X, Wu, X, Liu, C, Ma, J-x, et al.. Biological function of long non-coding RNA (LncRNA) Xist. Front Cell Dev Biol 2021;9:645647, https://doi.org/10.3389/fcell.2021.645647.Search in Google Scholar PubMed PubMed Central
13. Wang, M, Mao, C, Ouyang, L, Liu, Y, Lai, W, Liu, N, et al.. Long noncoding RNA LINC00336 inhibits ferroptosis in lung cancer by functioning as a competing endogenous RNA. Cell Death Differ 2019;26:2329–43, https://doi.org/10.1038/s41418-019-0304-y.Search in Google Scholar PubMed PubMed Central
14. Mo, X, Hu, D, Yang, P, Li, Y, Bashir, S, Nai, A, et al.. A novel cuproptosis-related prognostic lncRNA signature and lncRNA MIR31HG/miR-193a-3p/TNFRSF21 regulatory axis in lung adenocarcinoma. Front Oncol 2022;12:927706, https://doi.org/10.3389/fonc.2022.927706.Search in Google Scholar PubMed PubMed Central
15. Diao, X, Guo, C, Li, S. Construction of a novel prognostic signature in lung adenocarcinoma based on necroptosis-related lncRNAs. Front Genet 2022;13:833362, https://doi.org/10.3389/fgene.2022.833362.Search in Google Scholar PubMed PubMed Central
16. Tomczak, K, Czerwińska, P, Wiznerowicz, M. The Cancer Genome Atlas (TCGA): an immeasurable source of knowledge. Contemp Oncol 2015;19:A68–77, https://doi.org/10.5114/wo.2014.47136.Search in Google Scholar PubMed PubMed Central
17. O’Leary, NA, Wright, MW, Brister, JR, Ciufo, S, Haddad, D, McVeigh, R, et al.. Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion, and functional annotation. Nucleic Acids Res 2015;44:D733–45, https://doi.org/10.1093/nar/gkv1189.Search in Google Scholar PubMed PubMed Central
18. Brunson, J. ggalluvial: layered grammar for alluvial plots. J Open Source Softw 2020;5:2017. https://doi.org/10.21105/joss.02017.Search in Google Scholar PubMed PubMed Central
19. Yang, L, Yu, J, Tao, L, Huang, H-C, Gao, Y, Yao, J, et al.. Cuproptosis-related lncRNAs are biomarkers of prognosis and immune microenvironment in head and neck squamous cell carcinoma. Front Genet 2022;13:947551, https://doi.org/10.3389/fgene.2022.947551.Search in Google Scholar PubMed PubMed Central
20. Ligges, U, Maechler, M. Scatterplot3d – an R package for visualizing multivariate data. J Stat Software 2003;8:1–20, https://doi.org/10.18637/jss.v008.i11.Search in Google Scholar
21. Yu, G, Wang, L-G, Han, Y, He, QY. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics 2012;16:284–7, https://doi.org/10.1089/omi.2011.0118.Search in Google Scholar PubMed PubMed Central
22. Zuo, S, Wei, M, Wang, S, Dong, J, Wei, J. Pan-cancer analysis of immune cell infiltration identifies a prognostic immune-cell characteristic score (ICCS) in lung adenocarcinoma. Front Immunol 2020;11:01218, https://doi.org/10.3389/fimmu.2020.01218.Search in Google Scholar PubMed PubMed Central
23. Sturm, G, Finotello, F, List, M. Immunedeconv: an R package for unified access to computational methods for estimating immune cell fractions from bulk RNA-sequencing data. Methods Mol Biol 2020;2120:223–32, https://doi.org/10.1007/978-1-0716-0327-7_16.Search in Google Scholar PubMed
24. Hänzelmann, S, Castelo, R, Guinney, J. GSVA: gene set variation analysis for microarray and RNA-Seq data. BMC Bioinformatics 2013;14:7, https://doi.org/10.1186/1471-2105-14-7.Search in Google Scholar PubMed PubMed Central
25. Jiang, P, Gu, S, Pan, D, Fu, J, Sahu, AD, Hu, X, et al.. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med 2018;24:1550–8, https://doi.org/10.1038/s41591-018-0136-1.Search in Google Scholar PubMed PubMed Central
26. Maeser, D, Gruener, RF, Huang, RS. oncoPredict: an R package for predicting in vivo or cancer patient drug response and biomarkers from cell line screening data. Brief Bioinform 2021;22:bbab260, https://doi.org/10.1093/bib/bbab260.Search in Google Scholar PubMed PubMed Central
27. Mayakonda, A, Lin, D-C, Assenov, Y, Plass, C, Koeffler, HP. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res 2018;28:1747–56, https://doi.org/10.1101/gr.239244.118.Search in Google Scholar PubMed PubMed Central
28. Koren, E, Fuchs, Y. Modes of regulated cell death in cancer. Cancer Discov 2021;11:245–65, https://doi.org/10.1158/2159-8290.cd-20-0789.Search in Google Scholar
29. Wang, S, Liu, Y, Zhang, L, Sun, ZJCB. Methods for monitoring cancer cell pyroptosis. Cancer Biol Med 2021;19:398–414, https://doi.org/10.20892/j.issn.2095-3941.2021.0504.Search in Google Scholar PubMed PubMed Central
30. Guo, Y, Qu, Z, Li, D, Bai, F, Xing, J, Ding, Q, et al.. Identification of a prognostic ferroptosis-related lncRNA signature in the tumor microenvironment of lung adenocarcinoma. Cell Death Discov 2021;7:190, https://doi.org/10.1038/s41420-021-00576-z.Search in Google Scholar PubMed PubMed Central
31. Meng, X, Deng, Y, He, S, Niu, L, Zhu, H. m6A-mediated upregulation of LINC00857 promotes pancreatic cancer tumorigenesis by regulating the miR-150-5p/E2F3 axis. Front Oncol 2021;11:629947, https://doi.org/10.3389/fonc.2021.629947.Search in Google Scholar PubMed PubMed Central
32. Xu, S, Zhou, Y, Luo, J, Chen, S, Xie, J, Liu, H, et al.. Integrated analysis of a ferroptosis-related LncRNA signature for evaluating the prognosis of patients with colorectal cancer. Genes 2022;13:1094, https://doi.org/10.3390/genes13061094.Search in Google Scholar PubMed PubMed Central
33. Liu, J, Dai, Y, Lu, Y, Liu, X, Deng, J, Lu, W, et al.. Identification and validation of a new pyroptosis-associated lncRNA signature to predict survival outcomes, immunological responses and drug sensitivity in patients with gastric cancer. Math Biosci Eng 2023;20:1856–81, https://doi.org/10.3934/mbe.2023085.Search in Google Scholar PubMed
34. Xiang, Y, Feng, L, Liu, H, Liu, Y, Li, J, Su, L, et al.. SIPA1 regulates LINC01615 to promote metastasis in triple-negative breast cancer. Cancers 2022;14:4815, https://doi.org/10.3390/cancers14194815.Search in Google Scholar PubMed PubMed Central
35. Hu, Z, Yang, C, Guo, ST, Li, Y, Li, Y. LINC01615 activates ZEB2 through competitively binding with miR-3653-3p to promote the carcinogenesis of colon cancer cells. Cell Cycle 2021;21:228–46, https://doi.org/10.1080/15384101.2021.2015670.Search in Google Scholar PubMed PubMed Central
36. Ji, D, Chen, G-f, Liu, X, Zhu, J, Sun, J-Y, Zhang, X, et al.. Identification of LINC01615 as potential metastasis‐related long noncoding RNA in hepatocellular carcinoma. J Cell Physiol 2019;234:12964–70, https://doi.org/10.1002/jcp.27963.Search in Google Scholar PubMed
37. Wang, Y, Xu, Y, Liu, C, Yuan, C, Zhang, Y. Identification of disulfidptosis-related subgroups and prognostic signatures in lung adenocarcinoma using machine learning and experimental validation. Front Immunol 2023;14:1233260, https://doi.org/10.3389/fimmu.2023.1233260.Search in Google Scholar PubMed PubMed Central
38. Singh, V, Katiyar, A, Malik, PS, Kumar, S, Mohan, A, Singh, H, et al.. Identification of molecular biomarkers associated with non-small-cell lung carcinoma (NSCLC) using whole-exome sequencing. Cancer biomark 2023;1–18. https://doi.org/10.3233/cbm-220211.Search in Google Scholar PubMed
39. Feng, C, Song, C, Liu, Y, Qian, F, Gao, Y, Ning, Z, et al.. KnockTF: a comprehensive human gene expression profile database with knockdown/knockout of transcription factors. Nucleic Acids Res 2019;48:D93–100, https://doi.org/10.1093/nar/gkz881.Search in Google Scholar PubMed PubMed Central
40. Chen, J, Zhang, J, Gao, Y, Li, Y, Feng, C, Song, C, et al.. LncSEA: a platform for long non-coding RNA related sets and enrichment analysis. Nucleic Acids Res 2020;49:D969–80, https://doi.org/10.1093/nar/gkaa806.Search in Google Scholar PubMed PubMed Central
41. Waldman, AD, Fritz, JM, Lenardo, MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol 2020;20:651–68, https://doi.org/10.1038/s41577-020-0306-5.Search in Google Scholar PubMed PubMed Central
42. Michaud, D, Steward, CR, Mirlekar, B, Pylayeva-Gupta, Y. Regulatory B cells in cancer. Immunol Rev 2020;299:74–92, https://doi.org/10.1111/imr.12939.Search in Google Scholar PubMed PubMed Central
43. Kvedaraite, E, Ginhoux, F. Human dendritic cells in cancer. Sci Immunol 2022;7:eabm9409, https://doi.org/10.1126/sciimmunol.abm9409.Search in Google Scholar PubMed
44. Shapouri-Moghaddam, A, Mohammadian, S, Vazini, H, Taghadosi, M, Esmaeili, S-A, Mardani, F, et al.. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol 2018;233:6425–40, https://doi.org/10.1002/jcp.26429.Search in Google Scholar PubMed
45. Bagher, M, Rosmark, O, Elowsson Rendin, L, Nybom, A, Wasserstrom, S, Müller, C, et al.. Crosstalk between mast cells and lung fibroblasts is modified by alveolar extracellular matrix and influences epithelial migration. Int J Mol Sci 2021;22:506, https://doi.org/10.3390/ijms22020506.Search in Google Scholar PubMed PubMed Central
46. Shimasaki, N, Jain, A, Campana, D. NK cells for cancer immunotherapy. Nat Rev Drug Discov 2020;19:200–18, https://doi.org/10.1038/s41573-019-0052-1.Search in Google Scholar PubMed
47. Koppula, P, Zhuang, L, Gan, B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell 2020;12:599–620, https://doi.org/10.1007/s13238-020-00789-5.Search in Google Scholar PubMed PubMed Central
48. Pinho, SS, Reis, CA. Glycosylation in cancer: mechanisms and clinical implications. Nat Rev Cancer 2015;15:540–55, https://doi.org/10.1038/nrc3982.Search in Google Scholar PubMed
49. Pinho, S, Marcos, NT, Ferreira, B, Carvalho, AS, Oliveira, MJ, Santos-Silva, F, et al.. Biological significance of cancer-associated sialyl-Tn antigen: modulation of malignant phenotype in gastric carcinoma cells. Cancer Lett 2007;249:157–70, https://doi.org/10.1016/j.canlet.2006.08.010.Search in Google Scholar PubMed
50. Cecchi, F, Pajalunga, D, Fowler, C, Uren, A, Rabe, DC, Peruzzi, B, et al.. Targeted disruption of heparan sulfate interaction with hepatocyte and vascular endothelial growth factors blocks normal and oncogenic signaling. Cancer Cell 2012;22:250–62, https://doi.org/10.1016/j.ccr.2012.06.029.Search in Google Scholar PubMed PubMed Central
51. Bouaoun, L, Sonkin, D, Ardin, M, Hollstein, M, Byrnes, GB, Zavadil, J, et al.. TP53 variations in human cancers: new lessons from the IARC TP53 database and genomics data. Hum Mutat 2016;37:865–76, https://doi.org/10.1002/humu.23035.Search in Google Scholar PubMed
Supplementary Material
This article contains supplementary material (https://doi.org/10.1515/oncologie-2023-0384).
© 2023 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Review Articles
- Advances in ferroptosis of cancer therapy
- Immunotherapy in hepatocellular carcinoma: an overview of immune checkpoint inhibitors, drug resistance, and adverse effects
- The role of matrix metalloproteinase-2 in the metastatic cascade: a review
- The tumor microenvironment: a key player in multidrug resistance in cancer
- Robotic vs. laparoscopic approach in obese patients with endometrial cancer: which is the best? A mini-review
- SLC25 family with energy metabolism and immunity in malignant tumors
- Research Articles
- Catalase expression is an independent prognostic marker in liver hepatocellular carcinoma
- A novel immune-associated prognostic signature based on the immune cell infiltration analysis for hepatocellular carcinoma
- AKAP12 inhibits the proliferation of ovarian cancer by activating the Hippo pathway
- AQP1 as a novel biomarker to predict prognosis and tumor immunity in glioma patients
- Exosomal circular RNA NT5E driven by heterogeneous nuclear ribonucleoprotein A1 induces temozolomide resistance by targeting microRNA-153 in glioma cells
- miR‐30a‐3p inhibits the proliferation of laryngeal cancer cells by targeting DNMT3a through regulating DNA methylation of PTEN
- Disulfidptosis-related long non-coding RNAs predict prognosis and indicate therapeutic response in non-small cell lung carcinoma
- Case Report
- Primary retroperitoneal choriocarcinoma with lung and liver metastasis in a male patient: case report
- Short Commentary
- Clinical pharmacy services in cancer patients with hypertension
Articles in the same Issue
- Frontmatter
- Review Articles
- Advances in ferroptosis of cancer therapy
- Immunotherapy in hepatocellular carcinoma: an overview of immune checkpoint inhibitors, drug resistance, and adverse effects
- The role of matrix metalloproteinase-2 in the metastatic cascade: a review
- The tumor microenvironment: a key player in multidrug resistance in cancer
- Robotic vs. laparoscopic approach in obese patients with endometrial cancer: which is the best? A mini-review
- SLC25 family with energy metabolism and immunity in malignant tumors
- Research Articles
- Catalase expression is an independent prognostic marker in liver hepatocellular carcinoma
- A novel immune-associated prognostic signature based on the immune cell infiltration analysis for hepatocellular carcinoma
- AKAP12 inhibits the proliferation of ovarian cancer by activating the Hippo pathway
- AQP1 as a novel biomarker to predict prognosis and tumor immunity in glioma patients
- Exosomal circular RNA NT5E driven by heterogeneous nuclear ribonucleoprotein A1 induces temozolomide resistance by targeting microRNA-153 in glioma cells
- miR‐30a‐3p inhibits the proliferation of laryngeal cancer cells by targeting DNMT3a through regulating DNA methylation of PTEN
- Disulfidptosis-related long non-coding RNAs predict prognosis and indicate therapeutic response in non-small cell lung carcinoma
- Case Report
- Primary retroperitoneal choriocarcinoma with lung and liver metastasis in a male patient: case report
- Short Commentary
- Clinical pharmacy services in cancer patients with hypertension