Pyroptosis is an inflammation-mediated, programmed cell death (18). It can not only inhibit the occurrence and development of tumors but also develop a microenvironment that provides nutrition for cancer and accelerates its growth (8). In gastric cancer, a new pyroptosis-related gene signature has been identified to predict prognosis (19). However, the role of PRGs in prostate cancer (PCa) remains unknown, and we sought to elucidate this role.
PCa is a common malignant tumor found in elderly men worldwide (1). Unfortunately, approximately 27-53% of PCa patients develop local recurrence or distant metastasis within ten years of radical prostatectomy (20, 21). Biochemical recurrence (BCR) was defined as the second elevation of prostate-specific antigen (PSA) concentration above 0.2 µg/L, confirmed by two consecutive elevations. It is a determining risk factor for distant metastasis, prostate cancer specificity, and overall mortality (22). There is evidence that approximately 30% of patients with BCR develop distant metastases with clinical presentation, and 19–27% of patients may die of prostate cancer within ten years without receiving second treatment (23, 24). Therefore, stratifying patients with post-RP localized PCa into high-risk BCR patients is highly desirable, which may provide more frequent monitoring, early intervention, and even decision-making regarding adjuvant therapy.
It is an effective method to classify samples based on a predetermined gene expression signature (25). Our classification strategy is based on this approach and classifies PCa based on 52 PRGs expression patterns. We found that the expression of these PRGs was completely different between the two clusters due to heterogeneity. Besides, the prognosis of different clusters varies significantly. Several agreements emerged from our analysis: 1. The expression level of most PRGs was higher in cluster 2; 2. Most DEG expression levels among different clusters were higher in cluster 2; 3. Cluster 1, as a separate subtype, has a worse prognosis; 4. A combination of clinical information and RNA transcriptome data is more likely to reflect cell phenotype. After quantifying immune cells of different clusters, we discovered that many of them had a higher content of cluster 1, particularly T regulatory cells (Tregs). Tregs are the key barrier for tumor immunotherapy, as they actively mediate autoimmune tolerance (26, 27). In recent years, as medicine has advanced, immune checkpoint inhibitors have become the key treatment measures for many malignant tumors (28). PD-1, PD-L1, PD-L2, and CTLA4 are common immune checkpoints, where PD-L1 and PD-L2 are the two ligands of PD-1 and belong to B7 family (29, 30). PD-L1 is widely expressed throughout the body, particularly in immune and cancer cells, whereas PD-L2 expression is relatively limited to professional antigen-presenting cells and increases in response to congenital receptor signals (31). In addition, CTLA4 antibodies have been demonstrated to reverse T-cell allergy, leading to an antitumor response (32). We quantified immune checkpoints for different clusters and found cluster 2 had higher levels of these immune checkpoints, indicating that patients with cluster 2 were more likely to benefit from immunotherapy.
Clinical trials have tested anti-tumor molecular targeting drugs in all PCa subtypes, regardless of the underlying molecular subtypes. For instance, immune checkpoint molecules are expressed differently in different subtypes, and immunotherapy should be distinguished accordingly. To enhance clinical utility, we developed a scoring model (PRG-score) to quantify prognostic risk based on two clusters. This study provided strong evidence for clinical management of PCa. First, PRG-score considers the heterogeneity of patients, and PCA results indicate that scoring models can significantly distinguish patients from different risk subgroups. Second, the score can be associated with pyroptosis and prognosis. Specifically, PRG-score characterizes and assigns different weights to both tumor suppressor and tumor promoter genes. The signature included eight genes: CENPA, LCN2, COL7A1, ALB, UBXN10, SPZ1, SCNN1A, and TFF3. The coefficients of UBXN10, SCNN1A, LCN2, and TFF3 are negative, indicating that they can be used as protective factors for patients. Increased expression of these genes improves the prognosis of patients. The coefficients of ALB, SPZ1, CENPA, and COL7A1 are positive, indicating that increased expression increases the risk of poor prognosis in patients. After a careful review of relevant literature, we found that these genes were strongly associated with the occurrence and development of inflammation or malignant tumor. For instance, Masayuki Watanabe stated that Transcription factor SPZ1 might promote TWIST-mediated epithelial-mesenchymal transition in thoracic malignancies (33). By targeting SLPI, Xu et al. found that LCN2 mediated by IL-17 affects proliferation, migration, invasion, and cell cycle of gastric cancer cells (34). Stefan et al. found that COL7A1 editing via CRISPR/Cas9 in recessive dystrophic epidermolysis bullosa (35). Lin et al. found that TFF3 contributes to epithelial-mesenchymal transition (EMT) in papillary thyroid carcinoma cells via MAPK/ERK signaling pathway (36). Third, PRG-score can significantly distinguish the clinical characteristics of different patients, indicating that as the score increases, the proportion of PCa patients with T3, T4, and N1 increases significantly. PRG-Score predicts progression-free survival and, to a certain extent, the overall survival rate. Fourth, data on immune cell infiltration indicate that PRG-score has significant immunotherapeutic value. The results of ssGSEA reveal that the content of most immune cells in the low score group is higher than that in the high score group, which results in an overactive immune system that responds better to immunotherapy. There are significant differences in the content of immune checkpoints among different subgroups, which can provide important information for future research on immune checkpoints associated with PCa, particularly PD-L2 and CTLA4. Finally, PRG-score focuses directly on PCa cell death patterns compared to other models. Researchers have studied prognosis models of PCa with modification conditions such as ferroptosis, m6A, and immune score, such as seven-gene signature (15) discovered by Liu et al., eleven-gene signature (16) discovered by Zhang et al., and seven-gene signature (17) discovered by Lv et al. Through comparison, we found that the accuracy of PRG-score signature was superior to other prognostic models, and our research focused more on factors that directly contributed to tumor cell death and changed tumor microenvironment. As a result, our model is more valuable for facilitating treatment.
In our study, pyroptosis-related genes are used as the starting point, PCa patients are divided into different subtypes, DEGs are identified, and a prognosis model is constructed that can accurately predict tumor progression-free survival rate and overall survival rate of patients. At present, research progress on pyroptosis is limited, and the relationship between prostate cancer and pyroptosis has not been studied. Although we explored and verified it from multiple perspectives and different databases, this research still has certain limitations. Although our findings were validated using external data sets, additional in vivo and in vitro experiments were required due to limited sample size of data set for prostate cancer. In addition, we could not confirm whether these regulatory factors also play corresponding roles in the pyroptosis pathway of PCa, which warrants further investigation.