Data preprocessing
For training and validation cohorts, we collected 379 patients from the TCGA, 285 patients from GSE9891, 185 patients from GSE26712, 101 patients from GSE63885, and 266 patients from GSE49997, among which, 76 patients from the TCGA, 285 patients from GSE9891, 185 patients from GSE26712, 101 patients from GSE63885, and 266 patients from GSE49997 who underwent surgery and have survival features met the inclusion criteria. As there was a lack of normal ovarian patient data in the TCGA database, we obtained transcriptome data from 88 ovarian normal tissues in the GTEx database. After integration, we identified 680 programmed cell death genes that were expressed in both the training and validation cohorts, which were then used for further analysis. Additionally, we obtained single-cell RNA transcriptome data from 10 patients, which included 3 adjacent samples and 7 tumor samples from GSE210347.
Genotyping and clinical features of OV patients after surgery
Given the adequacy of sample size and the comprehensiveness of corresponding clinical information, we employed GSE9891 for phenotyping ovarian cancer patients. By performing Nonnegative Matrix Factorization (NMF) analysis using the expression matrix of 285 prognosis associated cell death-related genes defined by FSbyCox algorithm, we obtained two different subtypes of OV (Fig. 1A). These OV clusters exhibited significant differences in overall survival (OS, P < 0.001). Among which, cluster 2 showed favorable survival, while cluster 1 showed the worst survival (Fig. 1B). Additionally, we assessed the clinical characteristics of patients in different clusters. We found that 64% of patients in cluster 1 were diagnosed with grade 3. Meanwhile, the 97% of patients in cluster 1 were diagnosed with stage III/IV, while stage I/II predominated in patients in clusters 1. Besides, all patients in cluster 1 were diagnosed with Malignant and all low malignant potential (LMP) patients were in cluster 2, indicating that cluster 1 was characterized by advanced pathological stages, which is consistent with a faster decline in its survival curve (Fig. 1C). We further investigated Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathways between these clusters. We found that cluster 1 showed significantly enriched in pathways associated with tumor invasion and metastasis containing PI3K-Akt signaling pathway, JAK-STAT signaling pathway, Wnt signaling pathway, TGF-beta signaling pathway, TNF signaling pathway and NF-kappa B signaling pathway (Fig. 1D). Additionally, we performed a GSEA analysis based on differentially expressed genes between C1 and C2 groups. Pathways associated with treatment resistance and tumor invasion (e.g., Hypoxia, Mitotic Spindle, EMT and Myogenesis) were significantly upregulated in the cluster 1 group (Fig. 1E). Taken together, our results suggested that this classification demonstrates excellent prognostic potential for predicting postoperative outcomes in OV patients undergo surgery. Enrichment of pathways associated with treatment resistance and tumor invasion in the cluster 1 group is potential cause of its poor prognosis.
Tumor microenvironment and chemotherapy response in different clusters
Given the prominent role of immunotherapy as a highly promising therapeutic strategy for ovarian cancer, and recognizing the crucial involvement of cell death in activating antitumor immune responses, we conducted an analysis of the tumor microenvironment (TME) within these two identified clusters. Tertiary lymphoid structures (TLS) are acknowledged as germinal centers for immune cells in the TME, and thus we assessed the expression levels of a range of chemokines implicated in TLS formation. Intriguingly, we observed significantly higher expression of most chemokines responsible for regulating immune infiltration in cluster 1 (Fig. 2A). Furthermore, several interferons, including their receptors (e.g., IFNAR2, IFNGR2), and the majority of interleukins and their receptors were associated with transcripts linked to immune activation. Consistent with the higher expression of chemokines and increased immune cell infiltration within the TME of cluster 1, we noted elevated levels of these interferons, interleukins, and their receptors in this particular cluster (Fig. 2B, 2C).
Subsequently, we proceeded to calculate the immune score and ESTIMATE score using the ESTIMATE algorithm within the different clusters. A higher immune score signifies greater infiltration of immune cells, while a higher ESTIMATE score indicates lower tumor purity. Notably, we observed significantly elevated immune scores and ESTIMATE scores in cluster 1 compared to cluster 2, indicative of increased immune cell infiltration within cluster 1 (Fig. 2D). Additionally, we analyzed the immune-related function scores for each sample, revealing that most immune-related function scores were lower in cluster 2, suggesting suppression of these functions within this cluster (Fig. 2E).
Given that the expression of immune checkpoints serves as a crucial foundation for immune checkpoint inhibitors (ICIs) therapy, we proceeded to evaluate the expression levels of immune checkpoints within the two identified clusters. Remarkably, cluster 1 exhibited higher expression of most immune checkpoints, including CD274/PD-L1, CTLA4, HAVCR2/TIM-3, ICOS, IDO1, PD-L2, LAG3, and TIGIT, in comparison to cluster 2. This observation suggests that cluster 1 may potentially derive greater benefit from immunotherapy interventions (Fig. 2F). Furthermore, we conducted an additional assessment of the response to immune checkpoint inhibitors (ICIs) across the different clusters. Notably, patients in cluster 1 displayed higher TIDE scores, indicating a more favorable response to immunotherapy, whereas cluster 2 exhibited relative resistance. This finding aligns with the expression patterns of immune checkpoints observed (Figure 2G). Overall, the tumor microenvironment (TME) in cluster 1 appears to support anti-tumor immunity and exhibits higher expression of immune checkpoints, thereby suggesting potential benefits from ICIs therapy.
Variant landscape of programmed cell death genes in OV patients
Then, we used the TCGA-OV and GTEx cohort and applied the Limma algorithm to identify 239 differentially expressed genes (DEGs) associated with 12 programmed cell death (PCD) patterns. The DEGs were selected based on an adjusted p-value less than 0.05 and |LogFC| > 1. A comprehensive list of these DEGs can be found in Table S1, while their corresponding volcano plot is depicted in Fig. S1A. The KEGG was used to perform an enrichment analysis of the differentially expressed genes (DEGs), revealing their important role in regulating pathways associated with cancer processes. Specifically, the DEGs were found to be involved in the NF-kappa B signaling pathway, PI3K-Akt signaling pathway, and mTOR signaling pathway. Furthermore, the Gene Ontology (GO) enrichment analysis revealed that the differentially expressed genes (DEGs) are involved in crucial biological pathways such as regulating apoptotic signaling, autophagy, cellular response to hydrogen peroxide, and cell death in response to oxidative stress (Fig. S1B). Subsequent analysis using the UniCox method revealed that 28 of the identified genes were significantly correlated with the prognosis of OV patients Supplementary (Fig. S1C). The Circos plot of the expression of these 28 PCD genes demonstrated that CTSB was expressed at higher levels in OV prognostic effecta patients who underwent surgery (Fig. S1D). Additionally, we screened the CNV level of these features (Fig. S1E).
Establishment and evaluation of a programmed cell-death index in OV
To examine the prognosis of PCD-related mRNAs, we created a programmed cell-death index (PCDI) using Lasso and multiple Cox regression analysis. In contrast to conventional approaches, we employed multiple iterations (1000 times) to mitigate the impact of random errors in Lasso regression and reduce contingency issues. The process involved five steps: (i) screening of 28 PCD-related genes associated with prognosis, and removal of those with low variance (Var < 5), (ii) dimensional reduction of selected PCD-related genes using Lasso regression, repeated for 1000 times, and counting the total number of occurrences of each gene, (iii) retention of genes with a total number of occurrences of more than 999 times among the PCD-genes, (iv) the model with the maximum AUC value was selected as final prognostic model through multiple repetitions, (v) calculate the programmed cell-death index (PCDI) for each sample.
Through this process, we finally selected 8 PCD-related genes to establish model corresponding to the maximum AUC (5-year AUC = 0.81) (Fig. 3A - 3C), including FBXO7, NFATC4, PIK3R1, PPP1R15A, SIAH1, STXBP1, TIMP3 and UNC5B. Among them, FBXO7 and SIAH1 were positively correlated with survival rate while NFATC4, PIK3R1, PPP1R15A, SIAH1, STXBP1, TIMP3 and UNC5B were negatively correlated with survival rate (Fig. S2A). The programmed cell death index (PCDI) of each patient was exported using the following formula: PCDI = (-0.120880 * FBXO7 exp.) + (0.020864 * NFATC4 exp.) + (0.069234 * PIK3R1 exp.) + (0.008061 * PPP1R15A exp.) + (-0.179442 * SIAH1 exp.) + (0.032142 * STXBP1 exp.) + (0.066505 * TIMP3 exp.) + (0.013387 * UNC5B exp.). The PCDI was calculated using the formula above and patients were categorized into a high or low-PCDI group based on the best cut-off defined by the 'survminer' R package. In the TCGA cohort, 51 OV patients were in the high-PCDI group and 25 OV patients were in the low-PCDI group. Results from K-M analysis indicated that patients in the high-PCDI group had a lower overall survival rate compared to those in the low-PCDI group (P < 0.05) (Fig. 3B). Additionally, a low PCDI was associated with a prolonged survival time (Fig. 3D). Time-dependent receiver operating characteristic (ROC) curves were drawn using R software, and the area under the curve (AUC) was calculated at various time points to estimate the predictive performance of the prognostic model in the TCGA-OV cohorts. The ROC curve indicated that the PCDI possessed notable credibility and predictive value, with an AUC of 0.810 at 3 years and 0.774 at 5 years in the TCGA cohorts (Fig. 3C). Univariate Cox analysis revealed that only PCDI was associated with the survival rate of ovarian patients who underwent surgery (Fig. 3E). Furthermore, the multivariate Cox model identified PCDI as the only independent predictive factor with a p-value less than 0.001 (Fig. 3F).
External validation of the accuracy of programmed cell-death index
To assess the effectiveness and precision of the prognostic model in practical clinical use, we proceeded with external validation analysis. Our validation cohorts included GSE9891, GSE26712, GSE63885, and GSE49997. We determined the best cut-off using the 'survminer' R package and divided the patients into two groups. Specifically, 285 patients from GSE9891, 185 patients from GSE26712, 101 patients from GSE63885, and 266 patients from GSE49997 were included in the analysis. The results of the Kaplan-Meier analysis indicated that patients in the high-PCDI group had a significantly shorter overall survival and higher death rates (P < 0.05), as shown in Fig. 4A. Finally, multivariable Cox and logistic regression analyses were involved to establish a nomogram model in the OV cohort to estimate the overall survival. Sample type, Stage, Grade, and PCDI were included in the model (Fig. 4B).
Immune and functional landscapes of low- and high- PCDI groups
The immune system plays a crucial role in fighting tumors, and its effectiveness is influenced by various components within the TME. These components include the major histocompatibility complex (MHC) molecules that are responsible for presenting antigens to the immune system and interferons (IFNs) that help eradicate cancer cells. Furthermore, immune checkpoints, interleukins (ILs), cytokines, and their receptors represent essential TME components, with chemokines responsible for immune cell migration into the TME, and IFNs regulating the anti-tumor activity of effector T cells[27-29]. To explore the reasons behind the lower survival rate of patients with high-PCDI, a comprehensive analysis of the TCGA cohort was carried out. The study involved comparing immune functions of both groups to identify any discrepancies.
GSVA analysis was performed to examine the correlation between HALLMARK pathways and PCDI. Our results revealed that patients with high-PCDI exhibited an enrichment of pathways that support tumor progression and metastasis, including Epithelial mesenchymal transition, Hypoxia, Myogenesis and TGF BETA signaling (Fig. 5A). On the other hand, immune associated pathways such as IFN-α response, IFN-γ response, and inflammatory response were found to be enriched in the low-PCDI group (Fig. 5B).
Additionally, we found a strong negative correlation between PCDI and the expression of various chemokines and receptors, such as CCL5, CCL8, CCL18, CCR7, CXCL9, CXCL10, CXCL11, CXCL13, and XCL1 (Fig. 5C). Additionally, we observed a negative correlation between PCDI and the expression of chemokines and their receptors, including IFNs and their receptors (Fig. 5D and 5E). Moreover, our findings indicate that low-PCDI patients exhibited a significant improvement in classical immune function associated signatures, such as cytolytic activity, inflammation-promoting, T cell co-stimulation, and TIL. This suggests that these patients may have an increased response to immunotherapy, as shown in Figure 5F.
Dissection of tumor microenvironment based on single cell transcriptome
To investigate the differences in the tumor microenvironment between low- and high-PCDI groups, we analyzed single-cell mRNA profiles of ovarian carcinoma tissues from the GSE210347 dataset. We applied a filter to only include cells that expressed at-least 200 genes and removed cells with over 20% expression of mitochondrial genes. Following quality control, the filtered cells were classified into seven distinct cell types based on established biomarkers. These types include epithelial, identified by KRT8, KRT18, and EPCAM, T & NK, identified by CD3D, IL7R, NKG7 and GZMK cells, Cancer associated fibroblast (CAF), identified by COL1A1, DCN and COL1A2, Plasma, identified by JCHAIN, MZB1, and IGKC, Perivascular-like (PVL) cell, identified by TAGLN, MYH11, and MCAM, Macrophage, identified by SPP1, C1QC, S100A9 and CD14, Monocytes, identified by HLA-DRA and FCGR3A, Cycling cell, identified by MKI67 and CDK1, Cycling cell, identified by MKI67 and CDK1, Endothelial cell, identified by VWF and PECAM1, and B cell, identified by CD79A and MS4A1 (Fig. 6A, 6B and S3B). We utilized the lasso algorithm to evaluate the score of individual cells, using 8 PCD genes (Fig. 6C). Our findings indicate that the PCDI was significantly higher in the adjacent tissues as compared to the tumor tissues (Fig. 6D).
To overcome the limitations of our scRNA-seq dataset's sample size, we employed the use of BisqueRNA[30], a deconvolution algorithm, to simulate cell-type-specific gene expression profiles. This allowed us to predict the abundance of each cell type, as quantified by scRNA-seq, in larger scale datasets from four independent OV cohorts sourced from the GEO. The BisqueRNA tool's ability to accurately predict gene expression profiles specific to certain cell types was developed through training with our own single-cell RNA sequencing data. In order to better understand the relationships between various cell populations in the ovarian microenvironment, we conducted an analysis of pairwise Spearman correlations within infiltration patterns of ten major cell types across four independent ovarian cancer cohorts. We observed a strong negative correlation between cycling cells and PCDI across all groups studied and considered a spearman correlation coefficient [|Rs|] > 0.2 and P Value < 0.05 to indicate significant correlation. The correlation ranged from -0.2 in the GSE26712 dataset with 185 OV samples to -0.4 in the GSE49997 dataset with 266 OV samples (Fig. 6E). In addition, we found that the PCDI was higher in CAF, PVL, and endothelial cells, whereas it was almost zero in epithelial, T cell, plasma, and monocytes (Fig. S3A). Then, we utilized the Wilcoxon test to compare the proportion of Endothelial and CAF in low-PCDI samples to that in high-PCDI samples. To further analyze the changes in the immune microenvironment between the two groups, T&NK, macrophage, and monocytes were reaggregated (Fig. S3D, S3E). Additionally, infer-CNV was utilized to distinguish between malignant and normal epithelial cells (Fig. S3F).
Efficacy of programmed cell death index in predicting drug sensitivity
To investigate potential treatment strategies for high- and low-PCDI groups, we first explored the expression of immune checkpoints in the high- and low- PCDI groups, and revealed that OV patients with low-PCDI were more sensitive to immunotherapy drug, supported by the higher expression of immunotherapy targets such as BTLA, PD-L1 (CD274), CTLA4, ICOS and TIGIT in low-PCDI group (Fig. 7A). What’s more, by using TIDE webserver, we found that patients with high-PCDI responded poorly to checkpoint inhibitors (CPIs) therapy treatment, whereas the proportion of patients with low-PCDI responded to CPIs treatment was higher than patients with high-PCDI (Fig. 7B). In line with this, the TIDE score in OV patients was analyzed, revealing a higher TIDE score in high-PCDI patients and suggesting a significant positive correlation between the TIDE score and PCDI (Fig. 7B, 7C). Our results imply that OV patients with low-PCDI may benefit from immunotherapy.
To further investigate the correlation between the PCDI and drug sensitivity, we analyzed the IC50 values of each drug in OV samples to identify any significant variations. Our study revealed that patients in the high-PCDI group exhibited greater sensitivity to tyrosine kinase inhibitors, such as BMS.754807, AP.24534, OSI.906 and KIN001.135, as illustrated in Fig. 7D. Additionally, our findings indicate that OV patients with low-PCDI were more sensitive to Etoposide, a cell cycle specific antitumor agent, supported by the lower IC50 values of these drugs in the high-PCDI group (Fig. 7E).