Abstract

Background. Pyroptosis has a dual function in malignant tumor progression and management. The action of pyroptosis-related genes (PRGs) in pancreatic cancer (PC), however, remains uncertain. Methods. Differential expression analyses of 57 PRGs were conducted in the TCGA TARGET GTEx dataset. The candidate genes were determined using LASSO Cox regression and random forest analyses. A risk model was developed with the TCGA dataset and validated with the ICGC dataset. Results. Three prognosis-related PRGs (BAK1, GSDMC, and IL18) were chosen to create a risk model. High-risk patients from the TCGA and ICGC cohorts had an unfavorable overall survival (all ). The risk modelʼs accuracy and independent predictability were assessed by receiver operating characteristic curves and multivariate Cox regression analysis, respectively. High-risk patients possessed different molecular pathways, higher KRAS and TP53 mutations, increased expression of PD-L1, C1 immune subtype, and immunosuppressive microenvironment characterized by parainflammation compared to low-risk patients. KRAS and TP53 mutations participated in different inflammatory pathways and played different prognostic roles between the two risk groups. KRAS mutations in high-risk patients caused a more unfavorable prognosis than wild-type KRAS (), whereas TP53 mutations in low-risk patients exhibited a poorer outcome than wild-type TP53 (). Spearman correlation analyses revealed that the parainflammatory response in PC might be implicated in GSDMC-mediated pyroptosis via cytosolic DNA-sensing pathways under hypoxic conditions. Furthermore, the risk scores were significantly and positively related to the expression of HNRNPC, RBM15, YTHDF1, and YTHDF2, as well as sensitivity to gemcitabine, cisplatin, and erlotinib. Conclusions. This study created a novel pyroptosis-based risk model related to the parainflammatory immune microenvironment, which might help identify novel biomarkers, evaluate the tumor immune microenvironment, and develop management strategies for PC patients.

1. Introduction

Pancreatic cancer (PC) remains one of the most lethal malignancies worldwide. The disease ranks the fourth most common cancer-related death among both sexes in the United States [1] and sixth in China [2]. There were an estimated 458,918 new cases of PC diagnosed and 432,242 deaths from the disease globally in 2018 [3]. PC, a highly aggressive tumor, had a dismal prognosis [1]. Most PCs (more than 90%) are pancreatic ductal adenocarcinomas (PDACs) [4]. Surgical resection is regarded as the only potentially curative treatment for PC patients [5], but unfortunately, the majority of patients are either diagnosed at a late stage or are frequently ineligible for surgery, resulting in less than 20% of patients undergoing an operation [6]. Besides, even after radical resection, patients with PC are likely to experience high local recurrence rates and often have distant metastases [7]. Systemic chemotherapies are currently the standard regimen for treating advanced or metastatic PC, offering a slight overall survival (OS) benefit of only months [8, 9]. Immune therapy has been a hot topic in PC studies, but single-agent immune checkpoint inhibitors (ICIs) have generally been proven to be ineffective in clinical trials [10]. The immunosuppressive microenvironment is considered to cause PC resistance to immune therapies [11]. At present, the prognostic prediction of PC is mainly dependent on the clinicopathological staging system, which lacks accurate guidance for the prognosis and treatment of PC. Therefore, identifying novel predictive biomarkers for estimating the immune microenvironment is critically essential to predict survival and lay down personalized treatment regimens for PC patients.

Programed cell death (PCD), which is modulated by specific signaling pathways, is essential in maintaining homeostasis, host defense, cancer, and other pathologies [12]. PCD has been categorized into several distinct types, including apoptosis, autophagy, necrosis, ferroptosis, and pyroptosis [13]. Pyroptosis, different from other types of PCD, is generally regarded as a highly pro-inflammatory form of PCD [14]. Pyroptosis was observed for the first time in 1986 in mouse macrophages [15]. But until 2001, the first definition of pyroptosis introduced by Cookson and Brennan [16] distinguished it from apoptosis. In 2018, pyroptosis was described as a regulated cell death, which is mainly dependent on cytoplasmic membrane pore formation executed by gasdermin family members and is commonly (but not always) induced by inflammatory caspase activation [17].

Pyroptosis may be triggered by inflammasomes or without inflammasomes. Pyroptosis pathways dependent on inflammasomes comprise canonical caspase-1 pathway and noncanonical caspase-4/5/11 pathway, whereas inflammasome-independent pathways consist of granzyme proteases-dependent pathway and caspase-3-dependent pathway [18]. The canonical inflammasomes are started by pathogens or damage, which recruits pro-caspase-1 through inflammasome adaptor protein ASC (also called PYCARD), causing its self-cleavage and activating caspase-1 [19, 20]. Caspase-1 activation can mature pro-IL-1 and pro-IL18 [21] and cleave gasdermin D (GSDMD) protein to produce the GSDMD-N domain that forms the membrane pore to facilitate the release of IL-1β and IL18 [22]. The noncanonical pathway involves the activation of pro-caspase-4/5/11 [2325] to cleave GSDMD [26] and trigger pyroptosis. Activated caspase-11 induces NLRP3 inflammasome to start caspase-1, which promotes the maturation of IL-1β and IL18 [27, 28]. In the inflammasome-independent pathways, activated caspase-3 by chemotherapeutic drug cleaves gasdermin E (GSDME) into its N-terminal domain, causing cell membrane pores to form [29, 30]. In the granzyme proteases mediated pathway, the granzyme B produced by T cells or natural killer cells (NKs) leads to the target cell pyroptosis by direct cleavage of GSDME [31, 32].

Despite numerous recent studies exploring the functions of pyroptosis in cancers, it remains ambiguous how pyroptosis affects cancer prognosis and treatment. The reason for this may be the dual function of pyroptosis in cancer progression and management. Pyroptosis activation and cytokine release modify the tumor immune microenvironment (TIME) and facilitate tumor progression via evading immunosurveillance and inducing immune tolerance. Pyroptosis, however, can also improve immunotherapies through recruiting and activating immune cells to kill tumor cells [33, 34]. In some tumor types, including hepatocellular carcinoma [35], lung adenocarcinoma [36], and bladder cancer [37], pyroptosis-related genes (PRGs) have been assessed for their impact on prognosis and TIME. In PC, the functions of PRGs remain unclear and need to be further investigated. Hence, a novel pyroptosis-based risk model was established in the current study. We systematically explored the association of the risk model with prognosis, signaling pathways, somatic mutation, m6A-related gene expression, TIME, and drug sensitivity in PC. In addition, Increasing evidence demonstrates that pyroptosis is involved in inflammation and cancer immunity, but little is known about its correlation with parainflammation. Therefore, this study investigated and discussed the potential mechanisms of the influence of parainflammation on pyroptosis and TIME in PC. We found that cytosolic DNA-sensing pathways might participate in parainflammation-induced pyroptosis, which, to our knowledge, has not been reported previously.

2. Materials and Methods

2.1. Data Collection and Preprocessing

Since the TCGA dataset lacks sufficient data on normal pancreatic tissues, we downloaded the TCGA TARGET GTEx dataset (n = 19,109) with the Toil RNA-seq recompute expected_count version (accessed on November 19, 2021) from the UCSC Xena database [38]. Gene expression levels were presented as log2 (expected_count + 1). mRNA expression data for PC were derived from the TCGA TARGET GTEx dataset to determine differentially expressed PRGs. Data on 182 PC patients, including clinical information, RNA sequencing expression, and somatic mutations, were acquired from the TCGA database (accessed on December 4, 2021) as a training set. Eighty-two PC patients’ gene expression and clinicopathological data were available from the ICGC database (accessed on December 1, 2021) as a validation cohort. The transcripts per million (TPM) values for the TCGA and ICGC cohorts were converted from the fragments per kilobase of transcript per million mapped reads (FPKM) values according to the algorithm reported in previous studies [39, 40]. The TPM values were utilized for the subsequent analysis. The flow of our work is exhibited in Figure 1. Furthermore, 57 genes (Supplementary 1) associated with pyroptosis were collected from both a previous study [41] and the Molecular Signatures Database (MSigDB) [42].

2.2. Identifying Differentially Expressed PRGs

Differentially expressed genes (DEGs) in pancreatic tumor tissues and nontumor tissues from the TCGA TARGET GTEx cohort were analyzed using “DESeq2,” “edgeR,” and “limma” R packages with a threshold false discovery rate < 0.05 and |log2FC| ≥ 1. The intersection analysis of the three DEG sets obtained by the above methods found shared upregulated and downregulated DEGs. Subsequently, the intersection of PRGs with shared upregulated and downregulated DEGs identified differentially expressed PRGs for further analysis.

2.3. Constructing and Validating a Risk Model

Based on the STRING database (version 11.5) [43], an interaction network of the differentially expressed PRGs in the TCGA cohort was constructed. Using the R packages “igraph” and “reshape2,” a correlation network was created for candidate genes. Univariate Cox analysis using the “Survival” R package assessed the prognostic value of differentially expressed PRGs in the TCGA cohort. Least absolute shrinkage and selection operator (LASSO) Cox regression analysis (“glmnet” R package) and random forest analysis (“randomForestSRC” R package) were performed in the TCGA cohort to screen out candidate genes. The shared genes obtained by these two methods were selected as final signature genes to construct a risk model. Immunohistochemical (IHC) staining data for signature genes were obtained from the Human Protein Atlas (HPA) database. The expression levels of each of the signature genes and the corresponding exponentiated regression coefficients of the multivariate Cox regression model were used to compute PC patient risk scores.

The median risk score divided the patients into low- and high-risk groups. The “stats” and “Rtsne” R packages were used for principal component analysis (PCA) and t-distribution stochastic neighborhood embedding analysis (t-SNE), respectively. A Kaplan–Meier analysis with a log-rank test compared the OS between two risk groups. To investigate whether risk scores independently predicted patient prognosis, the risk score and clinicopathological features were analyzed with univariate and multivariate Cox regression analyses. Clinicopathological characteristics of both risk groups were compared using the χ2 test and presented as a heat map, including gender, age, TNM stage, T stage, N stage, M stage, and histological grade. The predictability of clinicopathological characteristics and the risk signature for 1-, 2-, and 3-year survival rates was estimated in the TCGA and ICGC cohorts using receiver operating characteristic (ROC) curves.

2.4. Function Analysis

Gene set enrichment analysis (GSEA) is a computational method for evaluating the distributional trend of a predefined gene set in the gene table ordered according to phenotypic relevance, thereby determining its impact on the phenotype [44]. The TCGA cohort was analyzed using GSEA to identify underlying pathways between the high- and low-risk groups. MsigDB provided KEGG gene sets (c2.cp.kegg.v7.4.symbols.gmt) and GO gene sets (c5.go.v7.4.symbols.gmt) as the reference gene sets to perform GSEA analysis. Determination of significance was performed with as the screening threshold. The gene sets of hypoxia (HALLMARK_HYPOXIA.v7.5.1.gmt) and the cGAS-STING pathway (REACTOME_STING_MEDIATED_INDUCTION_OF_HOST_IMMUNE_RESPONSES.v7.5.1.gmt) were also derived from MSigDB. The gene sets of the NF-kappaB pathway and the tumor necrosis factor (TNF) signaling pathway were acquired from a previous study [45]. To obtain enrichment scores for each pathway in each sample, single-sample GSEA (ssGSEA) scores for these four gene sets were computed using the gene set variation analysis (GSVA) package [46].

2.5. Immune Profile

The immune infiltration in tumor tissues was assessed with online tools and bioinformatics algorithms. TIMER2.0 provides users with multiple immune deconvolution methods, including XCEL, TIMER, QUANTISEQ, MCPCOUNTER, EPIC, CIBERSORT, and CIBERSORT-ABS algorithms [47]. The relationship of the risk score with immune infiltrating cells was evaluated using Spearman correlation analysis. Employing the Wilcoxon test in the R package “ggpubr,” we compared ssGSEA scores between the two risk groups for 14 immune-related pathways, including 13 pathways reported in a previous study [48] and the cGAS-STING pathway engaged in the induction of host immune responses. There may be an association between immune checkpoint gene expression and the efficacy of ICIs. Therefore, 47 immune checkpoint genes (Supplementary 1) were extracted from the training set and compared between the two risk groups. Moreover, we detected the association between the risk scores and six immune subtypes (C1–C6), namely C1 (wound healing), C2 (interferon (IFN)-gamma dominant), C3 (inflammatory), C4 (lymphocyte depleted), C5 (immunologically quiet), and C6 (TGF-beta dominant) [49].

2.6. Somatic Mutation and m6A-Related Modification Analyses

The mutation annotation format of somatic variants was available from the TCGA database, and visualization was done via the R package “maftools.” The somatic mutation count in the two risk groups was compared. In addition, we explored the effect of m6A-related modification on the risk signature by analyzing gene expression differences of m6A RNA methylation regulators between different risk groups.

2.7. Drug Sensitivities

The sensitivity of PC patients to targeted agents chemotherapeutic agents was assessed using the “pRRophetic” package [50], which predicts the half-maximal inhibitory concentration (IC50) of selected agents in both high- and low-risk groups. CellMiner database [51] was utilized to evaluate the association of individual prognostic genes with drug sensitivity. We extracted FDA-approved drugs from this database. An analysis of Pearson correlation was conducted with a screening threshold of .

2.8. Tumor Mutation Burden, Microsatellite Instability, and Immunotherapy

Data on the tumor mutation burden (TMB) and microsatellite instability (MSI) were obtained from the TCGA cohort through “maftools” and TCGAbiolinks” [52] R packages, respectively. Based on the reported dataset comprising 47 melanoma patients who had received ICIs therapy [53], we used the Subclass Mapping method [54, 55] to predict the response of different risk groups to ICIs treatment, including CTLA-4 and PD-1 inhibitors.

2.9. Statistical Analysis

All statistical analyses and data visualization were performed using R software (Version 4.1.0). Chi-square tests were employed to compare the different proportions of categorical variables. The Wilcoxon rank-sum tests and the Kruskal–Wallis test were utilized to compare two and multiple groups of nonnormally distributed continuous variables, respectively. Spearman correlation analysis was used to evaluate the correlations of the risk score with immune infiltrating cells and construct a Spearman correlation heatmap. The association of three prognosis-related PRGs expressions with drug sensitivity was assessed via Pearson correlation analysis. Two-tailed P values less than 0.05 were statistically significant in all statistical results.

3. Results

3.1. Identifying Differentially Expressed PRGs

In total, 5,241 upregulated genes and 2,884 downregulated genes, 5,182 upregulated genes and 2,941 downregulated genes, and 3,737 upregulated genes and 4,072 downregulated genes were identified between pancreatic tumor tissues and nontumor tissues of the TCGA TARGET GTEx cohort using “Deseq2,” “edgeR,” and “limma” packages, respectively (Figure 2(a)). Then, we obtained 6,494 DEGs shared by the above three methods, including 3,704 shared upregulated genes (Figure 2(b)) and 2,790 shared downregulated genes (Figure 2(c)). Twenty-six differently expressed PRGs were acquired by computing the intersection of 57 PRGs with 6,494 DEGs (Figure 2(d)). These 26 genes differently expressed between tumor and normal tissues were presented as a heatmap plot, with most of them highly expressed in PC tissues (Figure 2(e)). Except for two genes (CAPS5 and NLRP7), 24 of the 26 differentially expressed PRGs in the TCGA cohort were extracted for further analysis. A protein–protein interaction (PPI) analysis was performed for these 24 genes using the STRING database, as shown in Figure 2(f). In Figure 2(g), a correlation network was constructed between the 24 genes.

3.2. Constructing a Risk Signature

We used the TCGA-PAAD cohort as the training cohort to create a prognostic risk signature. A total of 177 PC samples with survival data were included in the survival analysis. A preliminary screening of prognosis-related genes was performed by univariate Cox regression analysis. Among the 24 differentially expressed PRGs, 10 genes significantly related to patient survival (HRs > 1 for all 10 genes) were selected for further study (Figure 3(a)). The LASSO Cox regression analysis identified four genes (BAK1, GSDMC, IL18, and TP63) by the optimum λ value (Figures 3(b) and 3(c)). Besides, we performed random forest analysis and screened out three genes (BAK1, GSDMC, and IL18) using a minimal depth approach (Figures 3(d) and 3(e)). Finally, three shared genes (BAK1, GSDMC, and IL18) of these two gene lists were retained as signature genes (Figure 3(f)). Significantly increased mRNA expression levels of these three signature genes were found in PC tissues compared to normal pancreatic tissues (Figure 2(e)). We further assessed the protein expressions of three signature genes in the HPA database and found that BAK1 and IL18 were overexpressed in PC tissues, whereas no GSDMC protein expression was observed in PC or normal tissues (Supplementary 2).

A three-gene prognostic signature was built according to the results of multiple Cox regression analysis, and each patient’s risk score was computed using the formula: the risk score = (1.496 × expression level of BAK1) + (1.255 × expression level of GSDMC) + (1.336 × expression level of IL18). The median risk score categorized PC patients into two groups: the high-risk group (n = 88) and the low-risk group (n = 89). An obvious correlation between the patients’ risk scores and the probability of death was observed, as evidenced by the distribution of risk scores and survival status (Figures 4(a) and 4(b)). PCA and t-SNE analyses confirmed that high- and low-risk patients were distributed in distinct directions (Figures 4(c) and 4(d)). Kaplan–Meier survival analysis demonstrated that high-risk patients had a significantly more dismal prognosis compared to the low-risk patients () (Figure 4(e)). The predictive accuracy of the risk signature was evaluated by time-dependent ROC curves, and the results indicated areas under the curve (AUC) values of 0.681, 0.641, and 0.701 for 1-, 2-, and 3-year survival, respectively (Figure 4(f)).

3.3. Validating the Risk Signature

The ICGC-PACA-AU dataset as external verification data further confirmed the robustness of the three gene signatures constructed in the TCGA dataset. In the same way, the calculation of risk scores for each patient in the ICGC-PACA-AU cohort was based on the expression levels of three signature genes and their exponentiated regression coefficients. The classification of the cases into the high- and low-risk groups was performed using the median risk score of the cohort. Similarly, death rates rose with higher risk scores (Figures 5(a) and 5(b)). Also, PCA and t-SNE analyses validated the risk score’s ability to classify (Figures 5(c) and 5(d)). According to Kaplan–Meier survival curves (Figure 5(e)), high-risk patients exhibited a decreased survival rate when compared to low-risk patients (). The AUC values for the risk signature at 1, 2, and 3 years were 0.597, 0.631, and 0.694, respectively (Figure 5(f)).

3.4. Independent Predictive Capability of the Risk Score

The available variables for the TCGA-PAAD cohort were assessed using univariate and multivariate Cox regression analyses to ascertain whether the risk score could independently predict patient prognosis. The results indicated that the risk score was significantly related to OS in both univariate and multivariate Cox regression analyses (hazard ratio (HR) = 1.225, 95% confidence interval (CI): 1.117–1.343, ; HR = 1.214, 95% CI: 1.098–1.342, , respectively) (Figures 6(a) and 6(b)). We also evaluated the associations of clinicopathological characteristics with the risk score, and the heatmap result showed a significant difference in histological grade between the two risk groups. Moreover, the three PRGs (BAK1, GSDMC, and IL18) were significantly overexpressed in the high-risk group as compared to the low-risk group. Nevertheless, no significant difference was found in the clinicopathological factors such as age, gender, T stage, N stage, M stage, and TNM stage between the two risk groups (Figure 6(c)). The time-dependent ROC curve depicted the 1-year AUC value of 0.681 for the risk score, and it exhibited superior predictive ability over clinicopathological characteristics (Figure 6(d)).

3.5. Function Analysis

The GSEA analysis of the TCGA cohort identified the underlying biological processes and enrichment pathways relevant to the risk model by analyzing GO terms and KEGG pathways. The top 5 KEGG pathways and GO terms were determined according to the p-value < 0.05. Based on the results of the GSEA analysis, the enriched KEGG pathways and GO terms in the high-risk group were mostly correlated with drug metabolism signaling pathways (drug metabolism-other enzymes) as well as tumor-related biological processes and signaling pathways, including cell cycle, P53 signaling pathway, DNA replication, pathways in cancer, mitotic nuclear division, epidermal cell differentiation, mitotic sister chromatid segregation, and epidermis development (Figures 7(a) and 7(c)), whereas the enriched KEGG pathways and GO terms in the low-risk group were mainly engaged in other pathways and biological processes, such as calcium signaling pathway, neuroactive ligand–receptor interaction, regulation of membrane potential, peptide hormone secretion, regulation of neurotransmitter level, and so on (Figures 7(b) and 7(d)). These findings suggested that PRGs associated with prognosis are probably involved in the drug treatment, occurrence, and progression of PC.

3.6. Immune Profile Analysis

Spearman correlation analysis was made to investigate the association of the risk score with immune infiltration using different algorithms, as depicted in Figure 8(a). According to the results, most of the immune cells, including B cells, Th2 CD4+ T cells, CD8+ T cells, regulatory T (Treg) cells, follicular helper T cells, neutrophils, M0 macrophages, M1 macrophages, M2 macrophages (from the CIBERSORT-ABS algorithm), NK cells, cancer-associated fibroblasts (CAFs), and myeloid dendritic cells (mDCs) (from the TIMER, CIBERSORT, and CIBERSORT-ABS algorithms) had a positive relation with the risk score. The ssGSEA scores of 14 immune-related pathways were calculated and subsequently compared between the two risk groups. High-risk individuals scored significantly higher in antigen-presenting cells (APCs) coinhibition, costimulation of APCs, Type I IFN responses, parainflammation, major histocompatibility complex (MHC) Class I and cGAS-STING pathway activation as compared to low-risk individuals (Figure 8(b)). The expression differences of immune checkpoints, crucial to immunotherapy, were compared in different groups. Compared with low-risk patients, the expression levels of TNFRSF14, TNFSF4, CD276, CD80, LGALS9, HHLA2, CD70, TNFSF9, TNFRSF25, TNFRSF4, CD40, TNFRSF18, TNFSF15, CD274, CD44, and TNFRSF9 in high-risk patients were significantly increased () (Figure 8(c)). Immune subtype analysis identified 56 patients with C1, 32 patients with C2, 40 patients with C3, one patient with C4, none with C5, and 21 patients with C6. Patients with C1, C2, C3, and C6 immune subtypes were included to be studied due to the absence of C5 patients and the extremely small number of C4 patients. A significant relation was discovered between the high-risk scores and C1 (wound healing), whereas the low-risk scores were positively linked with C3 (inflammatory) (Figure 8(d)).

3.7. Somatic Mutation and m6A-Related Modification Analyses

The 20 most frequently mutated genes in the two risk categories were depicted using waterfall plots based on somatic mutation analysis (Figures 9(a) and 9(b)). Although somatic mutation counts were not substantially different in the two risk groups () (Figure 9(c)), high-risk patients exhibited greater levels of KRAS and TP53 mutations than low-risk patients (Figures 9(d) and 9(e)). As a result, we examined the link between KRAS mutations and TP53 mutations in both high- and low-risk groups and observed a significant correlation (Figures 9(f) and 9(g)). Mutations in KRAS and TP53 were positively associated with the expressions of BAK1, GSDMC, IL18, and KRAS but not with PD-L1 or TP53 (Figures 9(h) and 9(i)). Besides, both KRAS mutations and TP53 mutations were positively related to the ssGSEA scores of hypoxia, the TNF signaling pathway, and the cGAS-STING pathway (Figures 9(j) and 9(k)). Patients with KRAS mutations had higher ssGSEA scores for the NF-kappaB pathway than those with wild-type KRAS, whereas patients with TP53 mutations had higher ssGSEA scores for MHC Class I, Type I IFN response, parainflammation, and pyroptosis than those with wild-type TP53 (Figures 9(j) and 9(k)). The results suggest that KRAS and TP53 mutations may dominate different inflammatory pathways. We also discovered that these two mutations had different prognostic effects between PC patients at high and low risk. High-risk patients with KRAS mutations suffered an inferior outcome than those with wild-type KRAS () (Figures 9(l) and 9(m)), whereas low-risk patients with mutant TP53 experienced a more adverse outcome than those with wild-type TP53 () (Figures 9(n) and 9(o)). In addition, the two risk groups were compared for gene expression levels of m6A RNA methylation regulators, and the high-risk groups exhibited significantly greater expression of HNRNPC, RBM15, YTHDF1, and YTHDF2 (Figure 9(p)).

3.8. Drug Sensitivities

The IC50 of three commonly used anticancer medications (gemcitabine, cisplatin, and erlotinib) in PC patients was estimated using the pRRophetic algorithm. IC50 denotes the concentration of an inhibitor needed to inhibit a biological or biochemical process by half. The high-risk group possessed significantly lower IC50 values than the low-risk group (Figure 10(a)10(c)), indicating that high-risk patients are more sensitive to these drugs. Spearman correlation analysis was employed to evaluate the connection between individual prognostic genes and drug sensitivity in NCI-60 cell lines from the CellMiner database. Statistical significant relationships () were selected and ranked according to their p-values. The top 16 results are presented in Supplementary 3. A negative relationship was observed between the expression of GSDMC and the drug sensitivity of cancer cells to lxazomib citrate, midostaurin, bortezomib, and pralatrexate. Similarly, upregulated IL18 expression increased cancer cell resistance to some drugs, such as pipamperone, bortezomib, actinomycin, and so on.

3.9. TMB, MSI, and Immunotherapy

The risk signature and TMB underwent a correlation analysis. High-TMB was defined as more than 20 muts/Mb, intermediate-TMB as 6–19 muts/Mb, and low-TMB as fewer than 5 muts/Mb [56]. Almost all patients had a low-TMB (median = 0.52 muts/Mb, range: 0–1.45 muts/Mb), except for one patient with a high-TMB (238.38 muts/Mb). The TMB did not differ significantly between the low- and high-risk categories (, Figure 10(d)), but the Spearman correlation analysis revealed a positive relationship of the risk scores with the TMB (R = 0.19, , Figure 10(e)). Also, MSI was estimated for the two risk groups. There were 140 patients with microsatellite stability (MSS), nine patients with low MSI (MSI-L), and none with high MSI (MSI-H). In comparison to the low-risk group, the high-risk group had a higher prevalence of MSI-L (Figure 10(f)). Subclass mapping analysis was then applied to predict the response of different risk groups to ICIs therapy. The result manifested that the high-risk group had a greater chance of responding to anti-CTLA-4 therapy (), but Bonferroni adjusted p-value was 0.08 (Figure 10(g)).

4. Discussion

The discovery of novel and effective molecular targets is crucial for diagnosing and treating PC due to its dismal prognosis and the limited traditional diagnostic and therapeutic strategies. The field of PCD has gained much attention in tumor biology research. Pyroptosis, an inflammatory form of PCD, has been demonstrated to participate in the progression of cancer, immunotherapies, and chemotherapies [57, 58]. However, pyroptosis in PC has not been fully elucidated in terms of its mechanisms and functions. Thus, we developed a pyroptosis-associated gene signature to explore its significance in PC patient prognosis, TIME, and treatment.

We obtained 57 genes associated with pyroptosis based on a previous study and the MSigDB. Because of the insufficient samples of the normal pancreas in the TCGA dataset, 26 PRGs with differential expression between PC and normal tissues were found in the TCGA TARGET GTEx dataset via three approaches. Twenty-four PRGs were enrolled in the TCGA cohort for further analysis, except for two genes without expression data. Ten PRGs were substantially associated with the prognosis of PC patients, as determined by univariate Cox regression. Our next step was to perform LASSO Cox regression and random forest analyses, and three PRGs (BAK1, GSDMC, and IL18) were selected as signature genes to construct a risk model. These three genes were more highly expressed at the mRNA level in PC tissues compared to normal pancreatic tissues, and their expressions were significantly associated with poor outcomes. IHC analysis from the HPA database confirmed that PC tissues overexpressed BAK1 and IL18 proteins compared to normal tissues, whereas neither PC tissues nor normal tissues had any detectable GSDMC protein. PC patients were divided into two categories, high- and low-risk, based on their median risk scores. Patients at higher risk had a worse outcome than those at lower risk. Both the univariate and multivariate Cox regression analysis revealed a significant relationship between the risk model and OS. The risk signature’s accuracy in predicting prognosis was confirmed by time-dependent ROC curves, which also showed that it was a more reliable prognostic predictor than clinicopathological characteristics in PC patients. More importantly, the risk model was verified to be robust and exhibited well-predictive capability in the ICGC cohort.

The BAK1 gene encodes the BAK protein. BAK, belonging to the BCL2 protein family, is a proapoptotic effector protein. Upon activation, BAK or BAX permeabilizes the outer membrane of mitochondria, allowing apoptogenic substances into the cytosol (such as cytochrome c) and activating caspase [5961]. Activation of BAX and BAK by chemotherapeutic agents can initiate the sequential activation of caspase-9 and caspase-3. The latter facilitates GSDME cleavage to produce N-termini, resulting in pyroptosis [57, 62]. Based on immunohistochemistry tests performed by Evans et al. [63] in PDAC and ampullary cancer, BAK expression was higher in malignant epithelia compared to acini and major ducts but lower in malignant epithelia compared to minor ducts. However, Graber [64] noted that although BAK mRNA expression in pancreatic tumor samples was 2.5-fold higher than that in the normal samples, elevated BAK expression in PC occurred in peritumoral tissue with chronic inflammation rather than in the tumor cells themselves.

GSDMC, a pore-forming protein’s precursor, is cleaved to liberate its N-terminal moiety, which induces membrane permeabilization and results in cell pyroptosis. Previous research has demonstrated that GSDMC acts as an oncogene to facilitate the proliferation and metastasis of tumor cells [65, 66]. In this study, GSDMC protein level was not detected in PC tissues or normal pancreatic tissues according to the results of the HPA database, which might be due to the low sensitivity of the selected IHC antibody. Xia et al. [67] verified by IHC staining analysis that elevated GSDMC expression is linked with a shorter OS in PDAC. Hou et al. [68] found that p-Stat3 increased PD-L1 nuclear translocation in hypoxic conditions, and these two factors combined boosted GSDMC transcription. Following TNF-alpha treatment, GSDMC is cleaved by activated caspase-8, causing tumor necrosis and pyroptosis. Notably, chronic tumor necrosis facilitates tumor growth and metastasis [69] and restrains antitumor immunity [68]. In addition, a poor prognosis was also discovered to be associated with increased GSDMC expression in breast cancer [68], lung adenocarcinoma [70], and kidney clear cell cancer [71].

IL18, as a pro-inflammatory cytokine, is constitutively expressed in monocytes, DCs, macrophages, and epithelial cells [72, 73]. The cytokine regulates innate and adaptive immune responses by influencing monocytes, DCs, NK cells, T cells, and B cells [74]. IL18 exhibits dual functions in regulating tumor cells and tumor microenvironment (TME). On the one hand, IL18 shows anticancer effects by facilitating the proliferation of NK cells and T lymphocytes [74]. On the other hand, IL18 participates in tumorigenesis and progression. It leads to tumor metastasis by proangiogenic action [75] and promotes the invasiveness of carcinoma cells by causing matrix metalloproteinase-9 (MMP-9) production [76]. Zhao et al. [77] discovered that IL18 derived from PDAC cells activates the PD-1/PD-L1 pathway by binding to its receptor on the surface of regulatory B cells, contributing to immune tolerance. In addition, IL18 enhances PC-cell proliferation and invasion via the NF-kappaB pathway [78]. The elevated level of IL18 induces Th1 immune responses characterized by hypersecretion of IFN-gamma and TNF-alpha, which are engaged in chronic inflammation [79]. A positive connection has been demonstrated between PC onset and chronic inflammation [80]. Chronic inflammation can generate conditions that promote malignant transformation by the gradual amassment of gene mutations and contribute to tumor cell proliferation and neovascularization [80]. IL18-binding protein (IL18BP), an antagonist to IL18 activity, possesses a high affinity for mature IL18 and blocks the interaction of IL18 with receptors on the cell surface [81]. Carbone et al. [79] found that the levels of free serum IL18 and IL18BP in PDAC patients were substantially higher than in healthy controls and that elevated serum levels of free IL18 were linked with an inferior outcome. This may be due to the imbalanced relationship between IL18 and IL18BP that influences the pathogenesis and progression of PC. However, one study by Guo et al. [78] observed that greater IL18 levels in PC tissues were linked with a shorter OS, whereas higher plasma IL18 levels were linked with a longer OS. In summary, IL18 overexpression might affect the development and progression of PC.

In this study, patients at high risk possessed relatively higher immune cell infiltration (including B cells, Th2 cells, CD 8+ T cells, Treg cells, Tfh cells, neutrophils, M0 macrophages, NK cells, M1 macrophages, mDCs, M2 macrophages, and CAFs) and more active immune functions or pathways (including APC costimulation, MHC Class I, parainflammation, APC coinhibition, Type I IFN response, and the cGAS-STING pathway) compared to patients at low risk. The immunosuppressive characteristics of the high-risk group verify the immune modulation ability of pyroptosis and affect PC patients’ prognosis. Underlying mechanisms are unclear, but parainflammation, a low inflammatory response between homeostasis and chronic inflammation [82], may participate in tumor initiation, invasion, metastasis, pyroptosis, and the formation of an immunosuppressive microenvironment in PC.

Parainflammation is characterized by Type I IFN response and TP53 mutation and is correlated with a worse prognosis [45]. Similarly, this study revealed that the patients at high risk exhibited a higher level of parainflammation, a stronger Type I IFN response, and higher TP53 mutations compared to patients at low risk. Parainflammation is caused by internal cell insults and is tightly associated with cellular senescence [83]. However, when TP53 mutations cause loss of p53 protein function, parainflammation shifts from cancer suppression to cancer promotion [83]. Thus, parainflammatory tumors tend to exhibit a high rate of TP53 mutations. Rapidly proliferating tumor cells are often afflicted with hypoxia, nutritional deficiencies, and energy shortages due to inadequate vasculature and blood supply [84]. Apoptosis will occur in cancer cells that cannot adapt to hypoxia [85]. As proapoptotic effector proteins, BAX and BAK modulate the permeability of the external mitochondrial membrane, which causes the translocation of cytochrome c and mitochondrial DNA (mtDNA) into the cytoplasm [86]. Multiple sensors of cytosolic DNA, namely absent in melanoma 2 (AIM2), Z-DNA binding protein 1(ZBP1), and cyclic GMP-AMP synthase (cGAS), exist for triggering innate immune responses in the cell [87]. mtDNA can effectively ignite the cGAS-STING-TBK1 signaling pathway, inducing the generation of Type I IFNs through activation of IFN-regulatory factor 3 (IRF3) and the production of other pro-inflammatory cytokines, which include IL-18 and IL-1β, via activating NF-kappaB pathway [88]. Type I IFNs attach to the IFN-alpha/beta receptor (IFNAR) complex and start the JAK/STAT downstream pathway, which triggers the transcription of IFN-stimulated genes [88] such as AIM2 [89] and ZBP1 [90]. Upon recognition of cytosolic DNA, ZBP1 initiated NLRP3 inflammasome activation through the RIPK1-RIPK3-caspase-8 axis [91]. Cytosolic DNA can also activate the AIM2 inflammasome. NLRP3 and AIM2 inflammasomes both initiate caspase 1, stimulating pro-IL-1 and pro-IL18 maturation. Under hypoxia, the PD-L1/p-Stat3 complex enhances GSDMC expression, which is subsequently cleaved with activated caspase-8, resulting in pyroptosis and the liberation of IL-1β and IL18 [68]. It is suggested that GSDMC expression is relevant to PD-L1 expression and hypoxic TME. Type I IFNs can increase PD-L1 expression [92]. Parainflammation characterized by Type I IFN response was observed to have a positive relationship with PD-L1 expression in PC (Figure 11). Hypoxia enhances tumor invasiveness, metastasis, and angiogenesis [93]. Of note, the risk score was positively related to NF-kappaB-mediated inflammatory response in the study. However, the ssGSEA score of pyroptosis did not correlate with the ssGSEA score of the NF-kappaB pathway; instead, it correlated significantly with the ssGSEA scores of Type I IFN response. In summary, the parainflammatory response in PC may be involved in GSDMC-induced pyroptosis through cytosolic DNA-sensing pathways under hypoxic conditions. The Spearman correlation heatmap (Figure 11) shows the relationship discussed above.

According to the somatic mutation analysis, patients at high risk presented a greater frequency of KRAS and TP53 mutations than patients at low risk. KRAS and TP53 mutations were positively correlated with the expressions of BAK1, GSDMC, IL18, and KRAS. Intriguingly, although KRAS mutations were significantly associated with TP53 mutations in two risk groups, these two gene mutations were involved in different inflammatory pathways and played different prognostic roles in distinct risk categories. Patients with KRAS mutations in the high-risk category possessed higher ssGSEA scores of the NF-kappaB pathway and a less favorable outcome than those with wild-type KRAS, while patients with TP53 mutations in the low-risk category possessed higher ssGSEA scores of parainflammation, Type I IFN response and pyroptosis and an unfavorable prognosis than those with wild-type TP53. The results also demonstrated that TP53 mutations rather than KRAS mutations played a significant part in parainflammatory response and pyroptosis in PC.

PC is characterized by desmoplasia and an inflammatory environment [94]. Cytokines participate in the interaction between immune cells and tumor cells [95]. IFNs encompass a broad family of cytokines that act as pivotal promoters of inflammation in the TME [96]. Type I IFNs are crucial for the TME of PC because they operate on immune and tumor cells, respectively, to prevent tumor development directly and indirectly [97]. They boost antitumor immunity by stimulating CD4+ helper T cells, CD8+ cytotoxic T cells, NK cells, DCs, and M1 macrophages while inhibiting Treg cells, myeloid-derived suppressor cells (MDSCs), and M2 macrophages [88]. Moreover, Type I IFNs activate DCs and enhance CD8+ T cell cross-priming. DCs, especially myeloid/conventional DC1 (cDC1), have a strong potential for processing antigens derived from tumor cells and cross-presenting these antigens through MHC class I to activate CD8+ T cells [98, 99]. However, chronic activation of the Type I IFN pathway or prolonged Type I IFN signaling may evoke immune suppressive mechanisms in cancer [100] and cause resistance to different cancer treatments [95], including ICIs resistance [101]. In the advanced phase of cancer progression, persistent Type I IFNs can upregulate indoleamine 2,3-dioxygenase and PD-L1 expressions on DCs and other myeloid cells, suppressing antitumor immunity [102]. This study also revealed a significantly positive connection between ssGSEA scores of Type I IFN response and PD-L1 expression (Figure 11). Therefore, parainflammation related to Type I IFN response may facilitate an immunosuppressive microenvironment in PC. NSAIDs have the potential to be of particular benefit to these PC patients with parainflammatory features, since NSAIDs attenuate human parainflammation.

MDSCs are a subset of bone marrow cells that express CD11b and Gr1 and are involved in immunosuppression [103]. The cells are typically categorized into two major cell subpopulations: monocytic (M)-MDSCs and granulocytic or polymorphonuclear (G/PMN)-MDSCs [104, 105]. Neutrophils resemble (G/PMN)-MDSCs in both phenotype and morphologyare. Neutrophils were a critical cell type in the TME of patients at high risk. It might be because PC cells attract neutrophils to the TME, inhibiting the immune response and promoting tumor progression. Neutrophils induce angiogenesis via MMP-9 [106] and promote epithelial-to-mesenchymal transition via elastase [107] in PDAC. Clinically, a high ratio of neutrophils to lymphocytes correlates positively with a poor prognosis in PDAC [108, 109]. Treg cells, known as antitumor immunosuppressants, can downregulate the activities of NK cells and T cells (CD4+ and CD8+). Transforming growth factor-beta and IL-10 produced by Treg cells have been demonstrated to suppress immune responses [110]. An elevated abundance of tumor-infiltrating Treg cells impairs the antitumor activity of CD8+ T cells and portends a worse prognosis in PDAC [111]. High-risk individuals also owned a greater abundance of T2 cells compared to low-risk individuals in this study. Increased Th2/Th1 ratio within the tumor-infiltrating cells indicated an inferior prognosis for PC patients with stage IIB/III [112]. The two main categories of tumor-related macrophages are “M1” and “M2” macrophages. M1 macrophages release pro-inflammatory cytokines that inhibit tumor growth, while M2 macrophages release anti-inflammatory cytokines that may promote tumor development [113]. Hu et al. [114] verified that M2 macrophage infiltration was negatively related to PC patients’ prognosis. Moreover, CAFs were crucial to the TIME of high-risk patients. CAFs are the principal cell type of the desmoplastic stroma in PC [115] and interact with tumor cells via various cellular pathways [116]. Once activated, myofibrotic CAFs or inflammatory CAFs undermine the anticancer immune response and accelerate the progression of PC [117].

Sixteen immune checkpoint molecules, which positively and negatively regulate immune responses, were overexpressed in high-risk individuals compared to high-risk individuals. It was demonstrated that high TNFSF9 expression was significantly linked with immune infiltration and poor OS in PC [118]. The research by Zhao et al. [119] revealed higher expression of B7-H3 (also called CD276) in PC patients than in normal controls, and its overexpression facilitates tumor progression. Also, increased expression of B7-H3 promotes cancer progression in primary hepatocellular carcinoma [120] and predicts a shorter OS in colorectal carcinoma [121]. PD-L1 (CD274) belongs to the B7 family [122] and is expressed on the cancerous cell membranes of various solid malignancies [123], including PC [124]. The interaction of PD-L1 with PD-1 protects cancerous cells from T-cell attack and contributes to their exhaustion [125]. Several studies have reached the same conclusion that increased PD-L1 expression in tumor tissues is implicated in the adverse prognosis of PC [124, 126, 127]. The function of other immune checkpoint molecules in PC has rarely been reported and requires further investigation.

An analysis of the relationship between risk scores and immune subtypes found that high-risk scores were strongly linked with C1 (wound healing), whereas low-risk scores were significantly linked with C3 (inflammatory). Thorsson et al. [49] identified 6 immuno-subtypes across carcinoma types and described their different features. C1 (wound healing) exhibited upregulation of angiogenic gene expressions, high KRAS and TP53 mutations, elevated proliferation rate, and high infiltration of Th2 cells [49]. Similarly, this study demonstrated that high-risk individuals had high frequencies of KRAS and TP53 mutations and increased Th2 cell infiltration in cancerous tissues. Oncogenic KRAS mutations have been confirmed to participate in fibro-inflammatory microenvironment development, tumorigenesis, and growth in PC [94, 128]. Wormann et al. [129] discovered that TP53 mutation or absence of p53 function in pancreatic tumors of mice activates the JAK2-STAT3 signaling pathway, which reduces fibrosis and the abundance of stellate cells in the stroma of pancreatic tumors and alters the types of immunocyte infiltration, promoting tumor growth, and stroma modification. C3 (inflammatory) had the most prominent Th17 signature, low to moderate neoplastic cell multiplication, increased Th1 genes, and decreased overall somatic copy number [49]. In addition, C3 had a better prognosis than other subtypes (including C1), which might be because C3 had a balanced immune response [49].

The m6A modification affects many malignancies by either increasing the expression of oncogenes or decreasing the expression of carcinoma suppressor genes [130, 131]. HNRNPC, RBM15, YTHDF1, and YTHDF2 expression levels were significantly higher in the current study’s high-risk individuals than in the low-risk individuals. Huang et al. [132] concluded that elevated expression of HNRNPC promotes tumor spread, rendering PDAC patients with a dismal prognosis. In hepatocellular carcinoma [133] and laryngeal squamous cell carcinoma [134], overexpression of RBM15 was reported to accelerate tumor growth and indicate a poor prognosis. YTHDF1 was discovered to boost antigen decomposition in the phagosome and prevent DCs from cross-presenting neoantigens, hence suppressing antitumor immunity [135, 136]. High YTHDF1 expression is related to an adverse prognosis in ovarian cancer [137] and breast cancer [138]. One research by Guo et al. [139] revealed that YTHDF2 is implicated in PC progression by cooperating with ALKBH5, an RNA demethylase.

The results of our estimation of the connection between these three gene expressions and medication sensitivity showed that GSDMC and IL18 expressions were adversely associated with the sensitivity of the medications presented in this study. Antitumor drugs such as gemcitabine, cisplatin, and erlotinib are generally recommended for PC treatment. As a result, we investigated the sensitivity of PC to these medications and discovered that high-risk tumors were more responsive to the medicines compared to low-risk tumors, implying that high-risk individuals may benefit from these therapies.

Moreover, ICIs therapy has exhibited impressive efficacy in the management of several solid tumors. In the present study, subclass mapping analysis suggested that high-risk individuals owned a higher likelihood of responding to anti-CTLA-4 treatment but with a Bonferroni-corrected p-value of 0.08. Increasing evidence indicates that ICIs therapy may be beneficial for patients with high-TMB and MSI-H [140, 141]. The association of risk scores with TMB and MSI was therefore examined. The risk scores and TMB showed a positive association, and MSI-L was more common in high-risk individuals than in low-risk individuals. However, almost all patients included in the study exhibited low-TMB and no MSI-H. Mismatch repair deficiency (dMMR)/MSI-H is presently the single reliable biomarker available for predicting immunotherapy response in PC patients [142]. As this population partially overlaps with high-TMB PC, and high-TMB/MSI-H PC represents a very small population, immunotherapy currently benefits only a tiny number of PC patients. The mainstay of treatment for most PC patients remains cytotoxic chemotherapy. Combining conventional therapy with targeted therapy and immunotherapy may create a promising PC therapeutic option.

However, this research has some restrictions to be noted. First, the risk signature was the lack of prospective data validation. Second, the exact function of these three gene combinations remains undetermined and requires additional experiments to confirm. Third, the risk score’s relationship with the tumor microenvironment requires to be determined experimentally. Fourth, the mechanism of parainflammation-induced pyroptosis needs to be validated in experiments.

5. Conclusions

A novel PC prognostic model with three PRGs was developed in this study. High-risk individuals possessed different molecular pathways, higher KRAS and TP53 mutations, increased expression of PD-L1, C1 immune subtype, and immunosuppressive microenvironment characterized by parainflammation compared to low-risk individuals. Besides, parainflammatatory response was crucial in PC pyroptosis. Consequently, the risk model may help discover novel biomarkers and offer fresh insights into predicting prognosis, exploring the TIME, and developing management strategies for PC patients.

Data Availability

The data supporting the findings of the present study are available upon request from the corresponding authors. All datasets in this study were downloaded from public databases.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Authors’ Contributions

Kong-kong Wei and Hao Chen designed this study. Kong-kong Wei and Zhi-xing Du collected the data. Kong-kong Wei, Zhi-xing Du, and Jun-ge Deng analyzed the data and interpreted the findings. Kong-kong Wei drafted the manuscript, which was revised by Jin-wei Yang and Hao Chen. Hao Chen obtained the funding and directed the research. The manuscript has been reviewed and approved by all authors.

Acknowledgments

The current study was funded by the Key Project of Science and Technology in Gansu province (no. 19ZD2WA001), the Key Talents Program of Gansu Province (no. 2019RCXM020), Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital (no. CY2017-ZD01), and Science and technology project of Chengguan District of Lanzhou City (nos. 2019RCCX0034, 2020SHFZ0039, and 2020JSCX0073). The platforms and datasets from UCSC Xena, TCGA, ICGC, MSigDB, HPA, STRING, TIMER2.0, and CellMiner are much appreciated.

Supplementary Materials

Supplementary 1. 57 pyroptosis-associated genes and 47 immune checkpoint genes.

Supplementary 2. IHC analysis of the protein expression of three signature genes in the Human Protein Atlas database.

Supplementary 3. Scatter plots of the connection between drug sensitivity and the expression level of each prognostic gene in the TCGA cohort.