Skip to main content

Identification of comutation in signaling pathways to predict the clinical outcomes of immunotherapy

Abstract

Background

Immune checkpoint blockades (ICBs) have emerged as a promising treatment for cancer. Recently, tumour mutational burden (TMB) and neoantigen load (NAL) have been proposed to be potential biomarkers to predict the efficacy of ICB; however, they were limited by difficulties in defining the cut-off values and inconsistent detection platforms. Therefore, it is critical to identify more effective predictive biomarkers for screening patients who will potentially benefit from immunotherapy. In this study, we aimed to identify comutated signaling pathways to predict the clinical outcomes of immunotherapy.

Methods

Here, we comprehensively analysed the signaling pathway mutation status of 9763 samples across 33 different cancer types from The Cancer Genome Atlas (TCGA) by mapping the somatic mutations to the pathways. We then explored the comutated pathways that were associated with increased TMB and NAL by using receiver operating characteristic (ROC) curve analysis and multiple linear regressions.

Results

Our results revealed that comutation of the Spliceosome (Sp) pathway and Hedgehog (He) signaling pathway (defined as SpHe-comut+) could be used as a predictor of increased TMB and NAL and was associated with increased levels of immune-related signatures. In seven independent immunotherapy cohorts, we validated that SpHe-comut+ patients exhibited a longer overall survival (OS) or progression-free survival (PFS) and a higher objective response rate (ORR) than SpHe-comut patients. Moreover, a combination of SpHe-comut status with PD-L1 expression further improved the predictive value for ICB therapy.

Conclusion

Overall, SpHe-comut+ was demonstrated to be an effective predictor of immunotherapeutic benefit in seven independent immunotherapy cohorts and may serve as a potential and convenient biomarker for the clinical application of ICB therapy.

Introduction

In recent years, immunotherapy with immune checkpoint blockades (ICBs) has emerged as a promising treatment for advanced-stage cancer. There are three main types of ICB therapies, including inhibition of programmed cell death 1 (PD-1) or ligand 1 (PD-L1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4), which have been approved by the FDA for a range of solid tumours [1, 2]. However, only a small subset of patients can benefit from ICBs, and the clinical application of ICBs is still limited. Thus, it is necessary to identify new biomarkers to screen patients who may respond to ICB therapy.

Tumour mutational burden (TMB), defined as the total number of somatic mutations per coding area of the genome in a tumour [3], has gained increasing recognition as a clinically predictive biomarker for immunotherapy response in several cancer types, including melanoma, non-small cell lung cancer (NSCLC), and urothelial carcinoma [4, 5]. However, TMB varies across different cancer types because of the heterogeneity of cancer. Moreover, increasing evidence indicates that TMB assessment and bioinformatics interpretation are also different across various targeted sequencing panels [6]. Therefore, an optimal threshold for classifying a cancer patient into TMB-high or TMB-low in ICB treatment remains unclear [7], which limits TMB as a biomarker for clinical application. Moreover, neoantigen load (NAL) is another potential ICB biomarker: Anagnostou et al. reported that ICB had significant therapeutic effects against tumours with increased mutation-associated neoantigen load [8]. Recent studies have shown that somatic mutations can give rise to neoepitopes, which may serve as neoantigens allowing for enhanced immunogenicity [9, 10]. However, NAL has the same limitation as TMB, which also restricts it as an effective biomarker for ICB [11].

Previous studies have demonstrated that mutations in specific genes may be associated with ICB efficacy [12, 13]. For example, Boichard et al. found that APOBEC-related mutagenesis increases immunogenicity, which was correlated with responses to immunotherapy [14]. Alterations in DNA damage response (DDR) genes are associated with increased TMB and tumour infiltration by T cells [15]. Pan et al. revealed that a 52-gene mutation signature could predict immunotherapy benefits in NSCLC [13]. These studies are mainly based on the gene level; nevertheless, exploring the impacts of mutations on immunotherapy at the pathway level will provide us with more robust results than at the gene level because of tumour heterogeneity. This may be because even if patients have different genetic mutations, these mutated genes tend to be ultimately involved in certain pathways [16, 17].

Thus, several pathway-based biomarker identification methods have been developed. Li et al. revealed that substantial alterations of genes in the Notch pathway may predict the prognosis of NSCLC patients [18]. Another study found that mutations in the PI3K-AKT-mTOR pathway can serve as predictors of the efficacy of immunotherapy in gastric adenocarcinoma [19]. Wang et al. suggested that the co-occurrence of alterations in the related pathways may help elucidate functionally relevant mechanisms that might inform treatment options, and they thus identified that comutation in some DDR pathways can serve as a potential biomarker for ICB [20]. However, they only focused on DDR-related pathways and did not consider other essential pathways, such as signaling pathways. Signaling pathways control essential biological progression, including molecular mechanisms, disease states, and drug responses [21,22,23,24,25,26]. Mutations in specific signaling pathways, for example, the NOTCH signaling pathway, have been proposed to be associated with the response to ICB in many cancer patients. Nevertheless, comutations in the signaling pathways of immunotherapy remain to be elucidated.

In this study, we comprehensively analysed the signaling pathway mutation status of 9763 samples across 33 different cancer types from The Cancer Genome Atlas (TCGA) to explore the association between the comutated pathway and immunotherapy response. ROC curve and multiple linear regression analyses were performed to determine the optimal combinations of comutated signaling pathways associated with increased TMB and NAL. Moreover, we further explored the associations between comutation in signaling pathways and the efficacy of immunotherapy. Collectively, we aimed to identify comutated signaling pathways to predict the clinical outcomes of immunotherapy, which may provide a potential and convenient biomarker for the future clinical application of immunotherapy.

Materials and methods

Data collection

We first collected 9763 cancer patients across 33 cancer types from the GDC TCGA data portal (https://portal.gdc.cancer.gov/), and their somatic mutations (whole-exome sequencing), gene expression (RNA-seq), and clinical data (sex, age, survival information, etc.) were downloaded accordingly. Then, the corresponding neoantigen data of 5446 TCGA cancer patients across 20 cancer types were downloaded from the TCIA (https://tcia.at/neoantigens) [27], and the microsatellite instability (MSI) data from 5930 TCGA cancer patients across 18 cancer types were obtained from the research of Hause et al. [28]. To further validate the association between comutated signaling pathways and the clinical benefit of ICBs, we retrieved seven clinical cohorts treated with ICBs from the published studies: (1) the Inova cohort includes 50 advanced melanoma patients treated with anti-PD-(L)1 or plus anti-CTLA-4 therapy [29]; (2) the Rizvi cohort includes 34 patients with NSCLC treated with anti-PD-1 therapy, with the tumour tissues profiled with the MSKCC panel [30]; (3) the Allen and Snyder cohorts respectively include 110 and 64 patients with melanoma who received anti-CTLA-4 therapy [31, 32]; (4) the Hellmann cohort consists of 75 patients with NSCLC treated with anti-PD-1 plus anti-CTLA-4 therapy [33]; (5) the Miao cohort contains 284 patients with mixed solid tumours (including non-small cell lung cancer, melanoma, head and neck cancer, bladder cancer, and clear cell renal cell carcinoma) treated with anti-CTLA-4 or PD-1/PD-L1 inhibitors [34]; (6) the MSKCC cohort, which was part of a pan-cancer study for targeted sequencing, including 1,661 characterized patients treated with anti-CTLA-4 or PD-1/PD-L1 inhibitors [35]. Patients’ responses to immunotherapy were based on definitions consistent with how they were evaluated in the above cohorts. For the Inova, Allen, Snyder, and Miao cohorts, the tumour immunotherapy response was defined by the Response Evaluation Criteria in Solid Tumours 1.1 (RECIST 1.1), and patients who experienced a complete response (CR) or partial response (PR) were classified as responders; patients who experienced stable disease (SD) or progressive disease (PD) were classified as non-responders. For the Rizvi and Hellmann cohorts, patients were classified as having a durable clinical benefit (DCB) (complete response [CR]/partial response [PR] or stable disease [SD] that lasted > 6 months) or no durable benefit (NDB) (progression of the disease [PD] or SD that lasted ≤ 6 months). Detailed information regarding the above cohorts is shown in Additional file 1: Table S1. We also summarized the details for the time period and dose selection of the above cohorts in Additional file 2: Table S2.

We then downloaded the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways from the MsigDB database [36] and manually extracted 68 canonical signaling pathways.

Identifying the comutated pathways associated with high TMB and high NAL

Using TCGA somatic mutation data, we constructed a non-silent gene binary mutation matrix, in which each column represents a sample and each row represents a mutated gene. If a gene mutation occurred in one sample, the element was 1; otherwise, it was 0. In our study, we extracted only non-silent somatic mutations in the genomic coding regions, including missense mutations, nonsense mutations, insertions, deletions, and splice mutations. Genes with mutation frequencies greater than 1% were considered. Then, we mapped the mutated genes into signaling pathways, and if a pathway involved at least one mutated gene, the pathway was defined as mutated. Pathways with a mutation frequency of at least 1% were retained. For each pathway, we used the multivariate Cox proportional hazard model to estimate the association between the pathway mutation status and overall survival (OS) by accounting for known covariates such as patient age, sex, and cancer type. The statistical significance levels (p values) of pathways were then corrected with the false discovery rate (FDR) method proposed by Benjamini and Hochberg [37]. Then, the pathways with FDR < 0.05 were retained for further analysis.

High TMB and NAL were proposed to be associated with an improved response to ICB treatment [38]. In this study, TMB was calculated as the total number of non-silent somatic mutations per coding area of the genome in a tumour, and the NAL data were downloaded from the TCIA (https://tcia.at/neoantigens). We intended to identify the comutated pathways associated with high TMB and NAL. To do this, we first divided the TCGA cancer patients into high TMB/NAL and low TMB/NAL groups according to the top quartile of TMB/NAL levels. Receiver operating characteristic (ROC) curve analysis was then used to determine the cut-off points for pathway mutation counts based on the highest Youden index. According to the cut-off of pathway mutation counts, we further screened which pathways were comutated. Multiple linear regression analysis was used to test the correlation between the survival-associated pathways and TMB/NAL. Considering the consistency of TMB and NAL data, the pathways with a suitable β coefficient were retained as comutated (comut+) pathways.

Survival analysis (OS or PFS) for the pathway comutation status was performed by Kaplan–Meier curves, and their significance was assessed by the log-rank method. Fisher’s exact test was used to identify the association between the pathway comutation status and ORR.

Differential analysis of immune-related features between pathway comut+ and comut patients

We first collected immune response-related genes, including immune checkpoints, T-cell receptor genes, tumour microenvironment (TME)-related genes, interferon-gamma (IFNγ)-associated genes, etc., from the published articles [39,40,41], which include 47 genes and were listed in Additional file 1: Table S3. For gene expression data of TCGA cancer patients, FPKM-normalized profiles were used, and all expression values were then log2(value + 1) transformed. We used the Wilcoxon rank-sum test to compare the differential expression levels of these immune response-related genes between comut+ and comut patients. The resulting p values were corrected using the FDR method.

Then, we compared the differences in immune-related signature features, including immune score, cytolytic activity (CYT) score, major histocompatibility complex (MHC)-I score, and T-cell-inflamed immune gene expression profile (GEP) score, between comut+ and comut patients. The immune score was estimated by the “ESTIMATE” R package [42]. The CYT score [43] and MHC-I score [44] were calculated by taking the mean expression of their signature genes. The GEP score [45] was estimated by performing ssGSEA using the GEP gene list.

Furthermore, the GSEA method was applied to identify the pathways associated with pathway comutation status. Specifically, a ranked gene list was produced based on the t-score of expression values between comut+ and comut patients, and then the functional gene sets or pathways were mapped to the ranked list. We performed these analyses using the “clusterProfiler” package [46].

Meta-analysis of ORR and OS or PFS based on pathway comutation status across multiple immunotherapy cohorts

To pool outcomes across multiple immunotherapy cohorts, the random-effect inverse-variance weighted approach was employed, which is computed by hazard ratio (HR) and its 95% confidence interval (CI) to assess the magnitude of influence on therapeutic benefits such as ORR, PFS, and OS based on pathway comut+ status. We used I2 statistics to assess the heterogeneity across different immunotherapy cohorts. Pheterogeneity > 0.1 and I2 < 50% indicated that there was no significant inter-cohort heterogeneity, and the results were consistent in these cohorts.

Results

Identification of comutated signaling pathways to estimate TMB and NAL

A detailed flow chart of our study is shown in Fig. 1. In the TCGA cohort of 9763 patients across 33 cancer types, genes with a non-silent mutation in at least 1% of cancer patients were retained. Then, the genes were mapped to the 68 signaling pathways, and if any one of the genes involved in the pathway was mutated, the pathway was defined as mutated. To explore whether the mutations in signaling pathways could predict higher TMB and NAL, we grouped patients into two subgroups: “Pathway+”, those with mutations in one or more signaling pathways; “Pathway”, those without mutations in any of the signaling pathways. Comparing the TMB and NAL levels between the two subgroups, we found that both TMB and NAL levels were significantly higher in the Pathway+ patient subgroup than in the Pathway patient subgroup by analysing the sequencing data from TCGA (Additional file 1: Fig. S1A, B), suggesting that tumours with more pathway mutations may generate and express more tumour-specific peptides, which are defined as neoantigens. Neoantigens are subsequently expressed on the surface of tumour cells via the major histocompatibility complex (MHC), which can activate a T-cell-mediated cytotoxic anti-tumour immune response and drive T-cell population expansion, thereby potentially affecting the immunotherapy response. These results indicate that comutations of multiple signaling pathways can be used to predict higher TMB and NAL levels.

Fig. 1
figure 1

Flowchart to identify comutations in signaling pathways

As not all signaling pathways are associated with cancer, we screened the survival-associated pathways. Specifically, for each pathway, we used the multivariate Cox proportional hazard model to estimate the association between the pathway mutation status and overall survival (OS) by accounting for known covariates, including patient age, sex, and cancer type. With FDR < 0.05, six significant pathways, such as the ECM receptor interaction (ECMRI) pathway, Spliceosome (Sp) pathway, Hedgehog (He) signaling pathway, and RNA degradation pathway, etc., were used for the following analysis (Additional file 1: Table S4). To further analyse how many pathways could be jointly used to predict higher TMB and NAL levels, receiver operating characteristic (ROC) curves were used to determine the optimal number of pathways in the pathway combination. To do this, we first classified the TCGA cancer patients into high TMB/NAL (TMB-H/NAL-H) and low TMB/NAL (TMB-L/NAL-L) groups according to the top quartile of TMB/NAL levels. When compared with other combinations of pathways in TMB-H estimation, mutations covering two pathways demonstrated the best Youden index, with 61.9% sensitivity and 88.5% specificity (Fig. 2A). Furthermore, the TCGA NAL data showed a similar result, with a sensitivity of 64.7% and specificity of 87.5% (Fig. 2B). These results indicated that the comutation of two signaling pathways may better predict higher TMB and NAL levels.

Fig. 2
figure 2

Correlations between SpHe-comut status and TMB, NAL, and MSI-H. A, B ROC curves of the number of comutated signaling pathways to predict higher TMB (A) and higher NAL levels (B) from TCGA. C, D Multiple linear regression β coefficients between six signaling pathways and TMB (C) and NAL (D). EF Comparison of TMB (E) and NAL (F) between patients with Sp-ECMRI comutation, He-ECMRI comutation, Sp-He comutation, and other comutations. G, H Comparison of TMB (G) and NAL (H) between SpHe-comut+ and SpHe-comut groups in tumors with the approved indication of immune therapy. I Overlapping of patients with SpHe-comut+ and MSI-H. J Comparison of TMB in different combinations of SpHe-comut status (SpHe-comut+ and SpHe-comut) and MSI status (MSI-H and MSS) groups. K Waterfall plot of two signature mutation pathways and the mutation genes involved in these pathways. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001; *****p < 0.00001. BLCA bladder urothelial carcinoma, HNSC head and neck squamous cell carcinoma, KIRC kidney renal clear cell carcinoma, LIHC liver hepatocellular carcinoma, SKCM skin cutaneous melanoma, STAD stomach adenocarcinoma, LUAD lung adenocarcinoma, LUSC lung squamous cell carcinoma

Considering latent interactions between different signaling pathways, we then used a multiple linear regression approach to determine the most efficient mode of pathway combination to research the relationships of signaling pathway mutations with TMB or NAL. As a result, the Spliceosome (Sp) pathway, Hedgehog (He) signaling pathway, and the ECMRI pathway, which have suitable β coefficients and contribute more to both TMB and NAL, were identified (Fig. 2C, D). To obtain the optimal two pathways, we compared the TMB and NAL for each pair of comutation pathway combinations. Despite not being able to completely exclude the effects of other comutation combinations, patients who had comutation of Sp-He illustrated significantly higher TMB and NAL than those with comutation of Sp-ECMRI and He-ECMRI or other comutations (Fig. 2E, F). Therefore, we defined the comutation in Sp-He pathways as “SpHe-comut+”; otherwise, we defined it as “SpHe-comut“. By comparing the TMB and NAL levels between the “SpHe-comut+” and “SpHe-comut” groups, we discovered that SpHe-comut+ patients showed significantly higher TMB and NAL than SpHe-comut patients (Additional file 1: Fig. S1C, D).

To explore the distribution of SpHe-comut status, we found that 702 patients were defined as SpHe-comut+ (7.19%) among the 9763 cases in the TCGA database. The number of SpHe-comut+ patients varied among different cancer types, ranging from 0% in KICH to 25.89% in SKCM (Additional file 1: Fig. S1E). We found that the distribution of TMB and NAL levels also varied among cancer types (Additional file 1: Fig. S1F, G), similar to the distribution of SpHe-comut+ among cancer types. We then analysed whether the presence of SpHe-comut+ was associated with an increased level of TMB or NAL. The results showed that there were significantly higher TMB (Fig. 2G) and NAL (Fig. 2H) levels in SpHe-comut+ patients than in SpHe-comut patients across cancer types with an approved indication of immune checkpoint inhibitors (such as SKCM, LUAD, HNSC, and BLCA, among others), except for the NAL levels of KIRC and LIHC. This may be due to the relatively low NAL levels in KIRC and LIHC (Fig. 2H), resulting in the difference not reaching statistical significance. Additionally, similar results were obtained for other cancer types (Additional file 1: Tables S5 and S6). Moreover, researchers found that MSI was strongly associated with TMB, and we then analysed whether the SpHe-comut+ status could be used to predict MSI-high (MSI-H) status. The results showed that SpHe-comut+ contributed significantly to MSI-H (Fisher’s exact test, P < 0.001) and predicted MSI-H with a sensitivity of 63.22% and a specificity of 94.15% (Fig. 2I). We then found that patients in the MSI-H/SpHe-comut+ group had significantly higher TMB than those in the MSI-H/SpHe-comut group, and in comparing microsatellite stability (MSS)/SpHe-comut+ to MSS/SpHe-comut, a significant difference in TMB was also observed (Fig. 2J). Furthermore, the waterfall plot showed that the patients in the SpHe-comut+ group had higher TMB and NAL levels, which covered more cancer patients than the MSI-H group (Fig. 2K). These findings suggested that SpHe-comut+ could be used to substitute for TMB estimation and could predict more TMB-H patients than MSI-H patients.

Correlations between SpHe-comut+ and immune-related features

Previous studies have reported a connection between TMB and immune-related features, including immune score, CYT score, major histocompatibility complex (MHC)-I score, and T-cell-inflamed immune GEP score [43,44,45]. To explore whether the SpHe-comut statuses are also associated with those immune-related features, we analysed 9272 samples of 33 cancers from TCGA with both gene expression and somatic mutation data. The Immune Score was first calculated using the “ESTIMATE” algorithm, and our results showed that the patients with SpHe-comut+ had a significantly higher immune score than patients with SpHe-comut over a pan-cancer analysis (Fig. 3A). The CYT score was then calculated based on the mean transcript levels of granzyme A (GZMA) and perforin 1 (PRF1), which are significantly upregulated with cytotoxic T-cell activation [43]. The MHC-I score was calculated using the mean expression of MHC-I core genes, and the T-cell-inflamed immune GEP score was estimated by performing ssGSEA using the GEP gene list (Additional file 1: Table S4). We observed obviously higher CYT scores, MHC-I scores, and GEP scores in the SpHe-comut+ patient group than in the SpHe-comut patient group (Fig. 3B–D). It was reported that the production of MHC is a crucial component of the immune system, which presents invading pathogen peptides to the surface membrane of T cells and triggers the immune response [47]. The GEP score was reported as a biomarker to classify patients who can benefit from anti-PD-1 therapy [45]. These findings indicated that SpHe-comut+ was associated with immune-related features, suggesting that SpHe-comut+ may affect the efficacy of immunotherapy.

Fig. 3
figure 3

Correlation between SpHe-comut status and immune-related features. AD Comparison of the immune score (A), CYT score (B), MHC-I score (C), and GEP score (D) between the SpHe-comut+ and SpHe-comut groups. E GSEA plot of significant hallmark pathways in comparison between the SpHe-comut+ and SpHe-comut groups. ****p < 0.0001

To further explore the biological functions affected by SpHe-comut+, GSEA was performed. A ranked gene list was constructed based on the gene differential expression levels (T-score) between the SpHe-comut+ and SpHe-comut groups across all cancer patients, and the gene sets of hallmark pathways from the MsigDB database were mapped to the ranked list. The results revealed that several immune-related hallmark pathways were significantly enriched in the SpHe-comut+ patient group, such as the interferon-gamma (IFNγ) response, DNA repair, and inflammatory response pathways (Fig. 3E). IFNγ was proposed to be an important marker of response to ICB in lung cancer and melanoma patients [48]. Deficiency in the DNA damage response has recently emerged as a major driver of tumour immunogenicity [49]. The inflammatory response (inflammation) has been demonstrated to be closely associated with all stages of development as well as with the efficacy of anti-cancer immunotherapy [50].

Additionally, we investigated the differential expression levels of immune response-related genes between the SpHe-comut+ and SpHe-comut groups over a pan-cancer analysis, and 39 of 47 genes showed significant differences (Wilcoxon rank-sum test, FDR < 0.05). We then compared the mean expression levels of these 39 genes between the SpHe-comut+ and SpHe-comut groups in each cancer type. Interestingly, the heatmap of mean difference expression levels of these genes showed that the cancer types with an approved indication of immune therapy (such as SKCM, LUAD, HNSC, and BLCA, among others) clustered together, and these genes presented higher mean expression levels in SpHe-comut+ patients in these cancer types (Fig. 4A). Moreover, in the pan-cancer analysis, these 39 genes, including five tumour microenvironment genes (Fig. 4B), five immune checkpoints (Fig. 4C), 11 T-cell receptors (Fig. 4D), and 18 T-effector and IFNγ pathway-relevant genes (Fig. 4E), were significantly up-regulated in the SpHe-comut+ group compared with the SpHe-comut group. These genes, such as IL18, IL1β, IL6, IL2RB, and IL15RA, are involved in the interleukin 1 (IL-1) family, which has been reported to promote the innate immune response to shape and improve the patient’s adaptive immune response [51]. These findings indicated that SpHe-comut+ patients may tend to respond to immunotherapy.

Fig. 4
figure 4

Correlations between SpHe-comut status and immune response-related gene mRNA expression. A Heatmap of mean expression differences in immune response-related genes between SpHe-comut+ and SpHe-comut patients across different cancer types. BE Comparison of the expression levels of genes related to the tumour microenvironment (B), immune checkpoints (C), T-cell receptor (D), and INFγ pathway and T-effector (E) signature between SpHe-comut+ and SpHe-comut groups in the pan-cancer analysis. *FDR < 0.05; **FDR < 0.01; ***FDR < 0.001; ****FDR < 0.0001; *****FDR < 0.00001.

SpHe-comut+ is a predictive and prognostic biomarker of ICBs

To explore whether the SpHe-comut+ status can be used to predict the efficacy of immunotherapy, three immunotherapy cohorts were obtained from published studies for this analysis, including the Inova cohort (melanoma), the Rizvi cohort (NSCLC), and the Miao cohort (pan-cancer) (see “Materials and methods”). The SpHe comutation status was examined in these three independent immunotherapy cohorts, and then the patients were divided into SpHe-comut+ and SpHe-comut groups. Our results showed that patients with SpHe-comut+ status had significantly higher TMB and NAL levels in these three cohorts, which was consistent with the results previously observed in TCGA pan-cancer analyses (Additional file 1: Fig. S2A–C). Moreover, we also found that the SpHe-comut+ status can be used as a biomarker to predict the efficacy of immunotherapy in three cohorts. Specifically, in the Inova cohort with 50 melanoma patients treated with anti-PD-(L)1 plus anti-CTLA4, 25 patients were defined as SpHe-comut+, and those patients had a significantly longer PFS (median PFS, SpHe-comut+ not reached (NR) vs. SpHe-comut 7.5 months; HR, 0.27; 95% CI, 0.11–0.64; log-rank test p = 0.002; Fig. 5A). An obviously higher ORR was found in the SpHe-comut+ patient group (SpHe-comut+ 88.0% vs. SpHe-comut 59.1%; Fisher’s exact test, p = 0.042; Fig. 5B). In the NSCLC Rizvi cohort, a significant association between SpHe-comut+ and longer PFS was observed (median PFS, SpHe-comut+ not reached (NR) vs. SpHe-comut 5.2 months; HR, 0.31; 95% CI 0.091–1.1; log-rank p = 0.047; Fig. 5C), as well as higher ORR (SpHe-comut+ 75% vs. SpHe-comut 30.7%; Fisher’s exact test, p = 0.042; Fig. 5D). The same results were observed in the Miao cohort, in which a longer OS (median OS, SpHe-comut+ 26.68 vs. SpHe-comut 20.39 months; HR, 0.69; 95% CI 0.47–1; log-rank p = 0.050; Fig. 5E) and a higher ORR (SpHe-comut+ 39.4% vs. SpHe-comut 21.5%; Fisher’s exact test, p = 0.002; Fig. 5F) was found in the SpHe-comut+ patient group. These results suggested that SpHe-comut+ had a good prognostic predictive ability in three immunotherapy cohorts and can be used as a predictor for the efficacy of immunotherapy.

Fig. 5
figure 5

Comparisons of PFS (or OS) and ORR between SpHe-comutand SpHe-comut groups across different cohorts. A Kaplan–Meier survival curves of PFS comparing SpHe-comut+ and SpHe-comut groups among patients with melanoma treated with anti-PD-(L)1 or anti-CTLA-4 therapy from the Inova cohort. B Comparison of ORR between the SpHe-comut+ and SpHe-comut groups from the Inova cohort. C Kaplan–Meier survival curves of PFS comparing the SpHe-comut+ and SpHe-comut groups in patients with NSCLC treated with anti-PD-1 therapy from the Rizvi cohort. D Comparison of ORR between the SpHe-comut+ and SpHe-comut groups from the Rizvi cohort. E Kaplan–Meier survival curves of OS comparing SpHe-comut+ and SpHe-comut groups in patients with pan-cancer treated with anti-CTLA4 or anti-PD-(L)1 therapy from the Miao cohorts. F Comparison of ORR between the SpHe-comut+ and SpHe-comut groups from the Miao cohort

To further verify the prognostic power of SpHe-comut+, the other four public immunotherapy cohorts with sufficient information on genomic alterations and survival were analysed. The four public immunotherapy cohorts included (1) the Hellmann cohort of 75 NSCLC patients treated with anti-PD-1 plus anti-CTLA4; (2) the Allen and Snyder cohort, a pooled analysis of two cohorts of 174 metastatic melanoma patients treated with anti-CTLA4; and (3) the MSKCC cohort of 1661 pan-cancer patients treated with anti-PD-(L)1 or anti-CTLA4. Our analysis revealed a significant positive association between SpHe-comut+ and TMB and NAL levels (Additional file 1: Fig. S2D–F).

Next, a survival analysis was performed in the Hellmann, Allen and Snyder, and MSKCC immunotherapy cohorts to explore the impacts of SpHe-comut status on PFS and OS. The survival analysis showed that SpHe-comut+ was significantly associated with a better prognosis in these immunotherapy cohorts (Fig. 6A–C). The SpHe-comut+ patients showed a higher ORR than the SpHe-comut patients in the Hellmann and Allen and Snyder cohorts (Additional file 1: Fig. S3A, B). According to a pooled analysis for the above cohorts, compared to the SpHe-comut group, the SpHe-comut+ patient group showed a higher ORR [pooled risk ratio (RR), 1.74; 95% CI 1.41–2.15; p < 0.001, Fig. 6D], a longer PFS (pooled HR, 0.56; 95% CI 0.39–0.79; p = 0.001, Fig. 6E), and a longer OS (pooled HR, 0.76; 95% CI 0.64–0.91; p = 0.002, Fig. 6F). The results of the statistical analysis for heterogeneity in all pooled estimates were insignificant (Pheterogeneity > 0.10 and I2 < 50%), indicating the consistency of the association between SpHe-comut+ status and favourable benefits to ICBs across these cohorts.

Fig. 6
figure 6

Pooled analysis of PFS, OS, and ORR comparing the SpHe-comut+ and SpHe-comut patients across different cohorts. A Kaplan–Meier survival curves of PFS comparing the SpHe-comut+ and SpHe-comut groups in the Hellmann cohort. B, C Kaplan–Meier survival curves of OS comparing the SpHe-comut+ and SpHe-comut groups in the Allen and Snyder cohort (B) and the MSKCC cohort (C). DF Pooled estimates of ORR (D), PFS (E), and OS (F) comparing SpHe-comut+ and SpHe-comut patients across different cohorts

Furthermore, we performed multivariate Cox regression analyses of SpHe-comut status and survival by adjusting for clinical features (such as patient age, sex, etc.) in the Inova cohort, Rizvi cohort, and Miao cohort. In each cohort, we found that SpHe-comut was an independent prognostic factor (Additional file 1: Fig. S4A–C).

Comparison of SpHe-comut with other immunotherapy biomarkers

Previous studies highlighted the relevance of TMB to the response to ICB therapy across a wide variety of cancer types [35]. Recently, Wang et al. found that comutations in DDR pathways can also serve as potential biomarkers for ICB [20]. We used the concordance index (C-index) and ROC curve analysis to evaluate the predictive value of SpHe-comut status for response to ICB therapy and compared it with TMB and DDR-comut status. In the Inova cohort (melanoma), the C-index and ROC curve analyses demonstrated that SpHe-comut status was a predictive biomarker of immunotherapy clinical benefit (C-index = 0.69, AUROC = 0.69), and its predictive power was superior to that of TMB and DDR-comut status (Additional file 1: Fig. S5A, B). Similar results were observed in the Rizvi cohort (NSCLC) and Miao cohort (pan-cancer) (Additional file 1: Fig. S5A, B).

Considering that SpHe-comut and DDR-comut are both immunotherapy predictive biomarkers based on TMB-screened pathway mutations, we then explored the joint utility of these two biomarkers for patient stratification. Survival analysis in the Rizvi cohort showed that dual-positive SpHe-comut and DDR-comut (both comut+) patients had longer PFS than either single-positive (single comut+) or dual-negative (both comut) patients (median PFS, not reached (NR) vs. not reached (NR) vs. 3.8 months, log-rank test, p = 0.012, Fig. 7A). Additionally, there was an increased proportion of DCB in both comut+ groups compared to the other two groups (80% vs. 55.6% vs. 25%, p = 0.046, Fig. 7B). We obtained similar results for the Inova cohort (Additional file 1: Fig. S6A, B) and Miao cohort (Additional file 1: Fig. S6C, D), which heralded the possibility of integrating SpHe-comut and DDR-comut as a novel marker for predicting the immune response.

Fig. 7
figure 7

Combining SpHe-comut status with DDR-comut status and PD-L1 expression for the prediction of ICB therapy. A Kaplan–Meier survival curves of PFS comparing the patients within each of the three indicated subgroups classified by SpHe-comut status and DDR-comut status in the Rizvi cohort. B Comparison of proportion of patients with DCB calculated within each of the three indicated subgroups classified by SpHe-comut status and DDR-comut status in the Rizvi cohort. C Kaplan–Meier survival curves of PFS comparing the patients within each of the three indicated subgroups classified by SpHe-comut status and PD-L1 expression in the Rizvi cohort. D Comparison of proportion of patients with DCB calculated within each of the three indicated subgroups classified by SpHe-comut status and PD-L1 expression in the Rizvi cohort

A number of recent studies have illustrated the importance of PD-L1 expression in response to PD-(L)1 blockade immunotherapy [52]. In the Rizvi cohort, we collected data across the board from patients with valid PD-L1 expression information. According to the cut-off point of 50%, the patients were classified into PD-L1high and PD-L1low groups. Based on multivariable Cox analysis, SpHe-comut status (HR, 0.16; 95% CI 0.04–0.67; p = 0.012) and PD-L1 expression (HR, 0.091; 95% CI 0.021–0.39; p = 0.001) were two independent predictor variables for anti-PD-(L)1 treatment (Additional file 1: Fig. S4B). We thus examined the relationship between these two biomarkers. A Kaplan–Meier analysis of the Rizvi cohort revealed that patients with dual positive status for SpHe-comut and PD-L1 (SpHe-comut+ and PD-L1high) obtained more favourable PFS than either single positive (SpHe-comut+ or PD-L1high) or dual negative patients (SpHe-comut and PD-L1low, median PFS, not reached (NR) vs. 14.5 vs. 2.1 months, log-rank test, p < 0.001, Fig. 7C). In addition, the objective response analysis showed an increased proportion of DCB in the SpHe-comut+ and PD-L1high groups compared to the two other groups (100% vs. 64.3% vs. 13.3%, p = 0.005, Fig. 7D). Based on these results, we speculated that the addition of SpHe-comut to PD-L1 might further improve the predictive value for anti-PD-(L)1 therapy.

In addition, we collected some mutated markers that have been regarded as stable immunotherapy markers in NSCLC, including those of the Li et al. method [18] and Bai et al. method [53]. Li et al. suggested that highly mutated NOTCH signaling may serve as a biomarker for the prediction of the prognosis of NSCLC patients treated with ICIs, and Bai et al. developed an eight-gene GMS risk model to predict the immunotherapeutic benefit in non-squamous NSCLC. We applied the Li et al. method and Bai et al. method in the Rizvi cohort, and the patients were then divided into GMS-high and GMS-low, NOTCH-high and NOTCH-low groups. We found that there was no significant difference in patients’ progression-free survival and immunotherapy response between each of the two patient groups (GMS-high vs. GMS-low or NOTCH-high vs. NOTCH-low, Additional file 1: Fig. S7A–D), while our method could classify patients into two groups with significant distinctive progression-free survival and immunotherapy response (Fig. 5C, D). These observations indicate that SpHe-comut had a better prognostic predictive ability than the Li et al. and Bai et al. methods.

Discussion

Recently, immunotherapy with immune checkpoint blockades (ICBs) has emerged as a promising treatment for cancer. TMB is a biomarker that can be used to predict patient response to cancer immunotherapy, indicating that patients with high TMB have better responses to ICB therapy. An increased neoantigen load is associated with a better response to ICB treatment as well. However, these indicators have limited use due to the lack of a validated cut-off value. Cancer is a disease in which cells acquire the ability to divide and proliferate uncontrollably, usually through genetic changes in specific genes. Mutation profiles differ dramatically between individuals with cancer. They may be used as indicators for treatment response and disease progression. Applying a molecular pathway-based approach to cancer mutations can dramatically boost their biomarker potential. Therefore, investigation of genomic mutation pathways may lead to the discovery of effective biomarkers for immunotherapy.

Alterations in human signaling pathways have been shown to be related to the development of cancer and its immunotherapy by many studies [21]. As we used the clinical immunotherapy data of cancer patients to identify the comutations in signaling pathways, the results may be easier to translate into clinical applications. In the study, we demonstrated that the presence of comutations in the Spliceosome (Sp) pathway and Hedgehog (He) signaling pathway could be used as a novel biomarker for ICB therapy. The prognostic and predictive values of SpHe-comut+ were validated in seven independent immunotherapy cohorts. In the melanoma cohort, the SpHe-comut+ group exhibited longer median PFS (Fig. 5A) and better ORR (Fig. 5B). In the NSCLC cohort, both the median PFS (Fig. 5C) and ORR (Fig. 5D) were improved in the SpHe-comut+ group. Moreover, in the pan-cancer cohort, the median OS and ORR were improved in the SpHe-comut+ group (Fig. 5E, F). Meanwhile, the predictive power of the response to immunotherapy of SpHe-comut+ was demonstrated to be superior to those of DDR-comut+ and TMB (Additional file 1: Fig. S5A, B). To further verify the prognostic power of SpHe-comut+, four other public immunotherapy cohorts were adopted. According to a pooled analysis for the above cohorts, the results showed that the statistical analysis for heterogeneity in the ORR, OS, and PFS pooled estimates were insignificant (Pheterogeneity > 0.10 and I2 < 50%), indicating the consistency of the association between SpHe-comut+ status and clinical results of ICBs across these cohorts (Fig. 6D–F).

As there are still multiple cellular and molecular mechanisms involved in immunotherapy, we thus combined SpHe-comut+ with other biomarkers, such as DDR-comut+ and PD-L1 expression. The results showed that patients with both comut+ statuses (SpHe-comut+ and DDR-comut+) had longer survival and higher response proportions to immunotherapy than either single-positive (single comut+) or dual-negative (both comut) patients in the Rizvi, Inova, and Miao cohorts, respectively (Fig. 7A, B, Additional file 1: Fig. S6A–D). Moreover, in the Rizvi cohort with PD-L1 expression information, we combined SpHe-comut and PD-L1 expression and demonstrated that patients with dual-positive status (SpHe-comut+ and PD-L1high) or single-positive status (SpHe-comut+ or PD-L1high) obtained more favourable PFS and DCB than patients with dual-negative status (SpHe-comut and PD-L1low status) (Fig. 7C, D). Based on these results, we speculated that the combination of SpHe-comut and DDR-comut or PD-L1 may operate as a synergistic component in immunotherapy prediction.

The advantage of our method is that it provides an optional and cost-effective approach by offering a small panel of genes in the signature pathways that can easily be translated into an easy-to-use clinical assay to identify potential immunotherapy responders. However, there are still limitations in our research. For example, we did not differentiate with respect to whether the gene mutations in the pathway were functional. Otto Warburg suggested that defects in mitochondrial respiration may be the underlying cause of cancer [54], and it would be better to add the role of comutation of mitochondrial genes to the prediction of the clinical outcomes of immunotherapy in our study. Our attempt to recruit functional mutations into our comut+ pattern was handicapped by the limited information available regarding the functions of different mutations. This suggests that the development of immunotherapeutic biomarkers requires more advanced sequencing techniques and nanomaterials, such as carbon nanotubes [55, 56]. If there is more information available regarding the functions of different mutations, the comut+ pattern may provide higher accuracy for efficacy prediction for ICB delivery.

Conclusion

In summary, the pan-cancer analysis revealed that comutation of Spliceosome (Sp) pathways and the Hedgehog (He) signaling pathway (defined as SpHe-comut+) was associated with increased TMB and NAL levels, as well as increased levels of immune-related signatures. Furthermore, the patients with SpHe-comut+ exhibited a higher ORR and a longer OS or PFS than SpHe-comut patients in seven independent immunotherapy cohorts. Our results suggest that SpHe-comut+ might provide an optional and cost-effective approach for identifying potential immunotherapy responders, which might complement recent immunotherapy biomarkers.

Availability of data and materials

The TCGA cancer patients’ data is downloaded from the GDC TCGA data portal (https://portal.gdc.cancer.gov/). The seven immunotherapy cohorts are available in the publications cited in the manuscript.

Abbreviations

ACC:

Adrenocortical carcinoma

BLCA:

Bladder urothelial carcinoma

BRCA:

Breast invasive carcinoma

CESC:

Cervical squamous cell carcinoma and endocervical adenocarcinoma

CHOL:

Cholangiocarcinoma

COAD:

Colon adenocarcinoma

CTLA4:

Cytotoxic T-lymphocyte antigen-4

C-index:

Concordance index

CI:

Confidence interval

CR:

Complete response

CYT:

Cytolytic activity

DLBC:

Lymphoid neoplasm diffuse large B-cell lymphoma

DCB:

Durable clinical benefit

DDR:

DNA damage response

ESCA:

Esophageal carcinoma

ECMRI pathway:

ECM receptor interaction pathway

GEP:

Gene expression profile

FPKM:

Fragments per kilobase of exon model per million mapped fragments

FDR:

False discovery rate

GBM:

Glioblastoma multiforme

HNSC:

Head and neck squamous cell carcinoma

IFNγ:

Interferon-gamma

HR:

Hazard ratio

ICB:

Immune checkpoint blockades

IL-1:

Interleukin 1

KICH:

Kidney chromophobe

KIRC:

Kidney renal clear cell carcinoma

KIRP:

Kidney renal papillary cell carcinoma

KEGG:

Kyoto encyclopedia of genes and genomes

LAML:

Acute myeloid leukemia

LGG:

Brain lower grade glioma

LIHC:

Liver hepatocellular carcinoma

LUAD:

Lung adenocarcinoma

LUSC:

Lung squamous cell carcinoma

MESO:

Mesothelioma

MSI:

Microsatellite instability

MSS:

Microsatellite stability

MHC:

Major histocompatibility complex

NDB:

No durable benefit

NAL:

Neoantigen load

NSCLC:

Non-small cell lung cancer

OV:

Ovarian serous cystadenocarcinoma

OS:

Overall survival

ORR:

Objective response rate

PD:

Progressive disease

PR:

Partial response

PAAD:

Pancreatic adenocarcinoma

PCPG:

Pheochromocytoma and paraganglioma

PRAD:

Prostate adenocarcinoma

PD-1:

Programmed cell death 1

PFS:

Progression-free survival

READ:

Rectum adenocarcinoma

ROC:

Receiver operating characteristic

SARC:

Sarcoma

SKCM:

Skin cutaneous melanoma

STAD:

Stomach adenocarcinoma

STES:

Stomach and esophageal carcinoma

SD:

Stable disease

ssGSEA:

Single-sample GSEA

TMB:

Tumor mutational burden

GSEA:

Gene set enrichment analysis

TGCT:

Testicular germ cell tumors

THCA:

Thyroid carcinoma

THYM:

Thymoma

TCIA:

The cancer immunome atlas

TCGA:

The cancer genome atlas

TME:

Tumor microenvironment

UCEC:

Uterine corpus endometrial carcinoma

UCS:

Uterine carcinosarcoma

UVM:

Uveal melanoma

WES:

Whole-exome sequencing

References

  1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373:23–34.

    Article  Google Scholar 

  2. Lemery S, Keegan P, Pazdur R. First FDA approval agnostic of cancer site—when a biomarker defines the indication. N Engl J Med. 2017;377:1409–12.

    Article  Google Scholar 

  3. Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, Schrock A, Campbell B, Shlien A, Chmielecki J, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9:34.

    Article  Google Scholar 

  4. Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, Stephens PJ, Daniels GA, Kurzrock R. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017;16:2598–608.

    Article  CAS  Google Scholar 

  5. Sha D, Jin Z, Budczies J, Kluck K, Stenzinger A, Sinicrope FA. Tumor mutational burden as a predictive biomarker in solid tumors. Cancer Discov. 2020;10:1808–25.

    Article  CAS  Google Scholar 

  6. Strickler JH, Hanks BA, Khasraw M. Tumor mutational burden as a predictor of immunotherapy response: is more always better? Clin Cancer Res. 2021;27:1236–41.

    Article  CAS  Google Scholar 

  7. Merino DM, McShane LM, Fabrizio D, Funari V, Chen SJ, White JR, Wenz P, Baden J, Barrett JC, Chaudhary R, et al. Establishing guidelines to harmonize tumor mutational burden (TMB): in silico assessment of variation in TMB quantification across diagnostic platforms: phase I of the friends of Cancer Research TMB Harmonization Project. J Immunother Cancer. 2020. https://doi.org/10.1136/jitc-2019-000147.

    Article  Google Scholar 

  8. Anagnostou V, Smith KN, Forde PM, Niknafs N, Bhattacharya R, White J, Zhang T, Adleff V, Phallen J, Wali N, et al. Evolution of neoantigen landscape during immune checkpoint blockade in non-small cell lung cancer. Cancer Discov. 2017;7:264–76.

    Article  CAS  Google Scholar 

  9. Matsushita H, Vesely MD, Koboldt DC, Rickert CG, Uppaluri R, Magrini VJ, Arthur CD, White JM, Chen YS, Shea LK, et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature. 2012;482:400–4.

    Article  CAS  Google Scholar 

  10. Tran E, Turcotte S, Gros A, Robbins PF, Lu YC, Dudley ME, Wunderlich JR, Somerville RP, Hogan K, Hinrichs CS, et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science. 2014;344:641–5.

    Article  CAS  Google Scholar 

  11. Gibney GT, Weiner LM, Atkins MB. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016;17:e542-51.

    Article  Google Scholar 

  12. Zhang K, Hong X, Song Z, Xu Y, Li C, Wang G, Zhang Y, Zhao X, Zhao Z, Zhao J, et al. Identification of deleterious NOTCH mutation as novel predictor to efficacious immunotherapy in NSCLC. Clin Cancer Res. 2020;26:3649–61.

    Article  CAS  Google Scholar 

  13. Pan D, Hu AY, Antonia SJ, Li CY. A gene mutation signature predicting immunotherapy benefits in patients with NSCLC. J Thorac Oncol. 2021;16:419–27.

    Article  CAS  Google Scholar 

  14. Boichard A, Pham TV, Yeerna H, Goodman A, Tamayo P, Lippman S, Frampton GM, Tsigelny IF, Kurzrock R. APOBEC-related mutagenesis and neo-peptide hydrophobicity: implications for response to immunotherapy. Oncoimmunology. 2019;8:1550341.

    Article  Google Scholar 

  15. Chae YK, Anker JF, Oh MS, Bais P, Namburi S, Agte S, Giles FJ, Chuang JH. Mutations in DNA repair genes are associated with increased neoantigen burden and a distinct immunophenotype in lung squamous cell carcinoma. Sci Rep. 2019;9:3235.

    Article  Google Scholar 

  16. Garraway LA, Lander ES. Lessons from the cancer genome. Cell. 2013;153:17–37.

    Article  CAS  Google Scholar 

  17. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr, Kinzler KW. Cancer genome landscapes. Science. 2013;339:1546–58.

    Article  CAS  Google Scholar 

  18. Li X, Wang Y, Li X, Feng G, Hu S, Bai Y. The impact of NOTCH pathway alteration on tumor microenvironment and clinical survival of immune checkpoint inhibitors in NSCLC. Front Immunol. 2021;12:638763.

    Article  CAS  Google Scholar 

  19. Wang Z, Wang X, Xu Y, Li J, Zhang X, Peng Z, Hu Y, Zhao X, Dong K, Zhang B, et al. Mutations of PI3K-AKT-mTOR pathway as predictors for immune cell infiltration and immunotherapy efficacy in dMMR/MSI-H gastric adenocarcinoma. BMC Med. 2022;20:133.

    Article  CAS  Google Scholar 

  20. Wang Z, Zhao J, Wang G, Zhang F, Zhang Z, Zhang F, Zhang Y, Dong H, Zhao X, Duan J, et al. Comutations in DNA damage response pathways serve as potential biomarkers for Immune checkpoint blockade. Cancer Res. 2018;78:6486–96.

    Article  CAS  Google Scholar 

  21. Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, Dimitriadoy S, Liu DL, Kantheti HS, Saghafinia S, et al. Oncogenic signaling pathways in the cancer genome atlas. Cell. 2018;173:321–37.e310.

    Article  CAS  Google Scholar 

  22. Han J, Han X, Kong Q, Cheng L. psSubpathway: a software package for flexible identification of phenotype-specific subpathways in cancer progression. Bioinformatics. 2020;36:2303–5.

    Article  CAS  Google Scholar 

  23. Han X, Kong Q, Liu C, Cheng L, Han J. SubtypeDrug: a software package for prioritization of candidate cancer subtype-specific drugs. Bioinformatics. 2021;37(16):2491–3.

    Article  CAS  Google Scholar 

  24. Li X, He Y, Wu J, Qiu J, Li J, Wang Q, Jiang Y, Han J. A novel pathway mutation perturbation score predicts the clinical outcomes of immunotherapy. Brief Bioinform. 2022;23(5):bbac360.

    Article  Google Scholar 

  25. Sheng Y, Jiang Y, Yang Y, Li X, Qiu J, Wu J, Cheng L, Han J. CNA2Subpathway: identification of dysregulated subpathway driven by copy number alterations in cancer. Brief Bioinform. 2021;22(5):bbaa413.

    Article  Google Scholar 

  26. Wu J, Li X, Wang Q, Han J. DRviaSPCN: a software package for drug repurposing in cancer via a subpathway crosstalk network. Bioinformatics. 2022;38:4975–7.

    Article  Google Scholar 

  27. Charoentong P, Finotello F, Angelova M, Mayer C, Efremova M, Rieder D, Hackl H, Trajanoski Z. Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep. 2017;18:248–62.

    Article  CAS  Google Scholar 

  28. Hause RJ, Pritchard CC, Shendure J, Salipante SJ. Classification and characterization of microsatellite instability across 18 cancer types. Nat Med. 2016;22:1342–50.

    Article  CAS  Google Scholar 

  29. Abbott CW, Boyle SM, Pyke RM, McDaniel LD, Levy E, Navarro FCP, Mellacheruvu D, Zhang SV, Tan M, Santiago R, et al. Prediction of immunotherapy response in melanoma through combined modeling of neoantigen burden and immune-related resistance mechanisms. Clin Cancer Res. 2021;27:4265–76.

    Article  CAS  Google Scholar 

  30. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–8.

    Article  CAS  Google Scholar 

  31. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, Walsh LA, Postow MA, Wong P, Ho TS, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189–99.

    Article  Google Scholar 

  32. Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, Sucker A, Hillen U, Foppen MHG, Goldinger SM, et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science. 2015;350:207–11.

    Article  Google Scholar 

  33. Hellmann MD, Nathanson T, Rizvi H, Creelan BC, Sanchez-Vega F, Ahuja A, Ni A, Novik JB, Mangarin LMB, Abu-Akeel M, et al. Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell. 2018;33:843–52.e844.

    Article  CAS  Google Scholar 

  34. Miao D, Margolis CA, Vokes NI, Liu D, Taylor-Weiner A, Wankowicz SM, Adeegbe D, Keliher D, Schilling B, Tracy A, et al. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat Genet. 2018;50:1271–81.

    Article  CAS  Google Scholar 

  35. Samstein RM, Lee C-H, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, Barron DA, Zehir A, Jordan EJ, Omuro A, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51:202–6.

    Article  CAS  Google Scholar 

  36. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, Mesirov JP. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–50.

    Article  CAS  Google Scholar 

  37. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B (Methodol). 1995;57(1):289–300.

    Google Scholar 

  38. Carbone DP, Reck M, Paz-Ares L, Creelan B, Horn L, Steins M, Felip E, van den Heuvel MM, Ciuleanu TE, Badin F, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 2017;376:2415–26.

    Article  CAS  Google Scholar 

  39. Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, Albright A, Cheng JD, Kang SP, Shankaran V, et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest. 2017;127:2930–40.

    Article  Google Scholar 

  40. Dong ZY, Zhong WZ, Zhang XC, Su J, Xie Z, Liu SY, Tu HY, Chen HJ, Sun YL, Zhou Q, et al. Potential predictive value of TP53 and KRAS mutation status for response to PD-1 blockade immunotherapy in lung adenocarcinoma. Clin Cancer Res. 2017;23:3012–24.

    Article  CAS  Google Scholar 

  41. Higgs BW, Morehouse CA, Streicher K, Brohawn PZ, Pilataxi F, Gupta A, Ranade K. Interferon gamma messenger RNA signature in tumor biopsies predicts outcomes in patients with non-small cell lung carcinoma or urothelial cancer treated with durvalumab. Clin Cancer Res. 2018;24:3857–66.

    Article  CAS  Google Scholar 

  42. Aran D, Sirota M, Butte AJ. Systematic pan-cancer analysis of tumour purity. Nat Commun. 2015;6:8971.

    Article  CAS  Google Scholar 

  43. Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160:48–61.

    Article  CAS  Google Scholar 

  44. Liu D, Schilling B, Liu D, Sucker A, Livingstone E, Jerby-Arnon L, Zimmer L, Gutzmer R, Satzger I, Loquai C, et al. Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat Med. 2019;25:1916–27.

    Article  CAS  Google Scholar 

  45. Cristescu R, Mogg R, Ayers M, Albright A, Murphy E, Yearley J, Sher X, Liu XQ, Lu H, Nebozhyn M, et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science. 2018;362:eaar3593.

    Article  Google Scholar 

  46. Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, Feng T, Zhou L, Tang W, Zhan L, et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation. 2021;2:100141.

    CAS  Google Scholar 

  47. Radwan J, Babik W, Kaufman J, Lenz TL, Winternitz J. Advances in the evolutionary understanding of MHC polymorphism. Trends Genet. 2020;36:298–311.

    Article  CAS  Google Scholar 

  48. Karachaliou N, Gonzalez-Cao M, Crespo G, Drozdowskyj A, Aldeguer E, Gimenez-Capitan A, Teixido C, Molina-Vila MA, Viteri S, De Los Llanos Gil M, et al. Interferon gamma, an important marker of response to immune checkpoint blockade in non-small cell lung cancer and melanoma patients. Ther Adv Med Oncol. 2018;10:1758834017749748.

    Article  Google Scholar 

  49. Chabanon RM, Rouanne M, Lord CJ, Soria JC, Pasero P, Postel-Vinay S. Targeting the DNA damage response in immuno-oncology: developments and opportunities. Nat Rev Cancer. 2021;21:701–17.

    Article  CAS  Google Scholar 

  50. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–99.

    Article  CAS  Google Scholar 

  51. Sims JE, Smith DE. The IL-1 family: regulators of immunity. Nat Rev Immunol. 2010;10:89–102.

    Article  CAS  Google Scholar 

  52. Loke P, Allison JP. PD-L1 and PD-L2 are differentially regulated by Th1 and Th2 cells. Proc Natl Acad Sci USA. 2003;100:5336–41.

    Article  CAS  Google Scholar 

  53. Bai X, Wu DH, Ma SC, Wang J, Tang XR, Kang S, Fu QJ, Cao CH, Luo HS, Chen YH, et al. Development and validation of a genomic mutation signature to predict response to PD-1 inhibitors in non-squamous NSCLC: a multicohort study. J Immunother Cancer. 2020;8:e000381.

    Article  Google Scholar 

  54. Warburg O. On respiratory impairment in cancer cells. Science. 1956;124:269–70.

    Article  CAS  Google Scholar 

  55. Ahmadian E, Janas D, Eftekhari A, Zare N. Application of carbon nanotubes in sensing/monitoring of pancreas and liver cancer. Chemosphere. 2022;302:134826.

    Article  CAS  Google Scholar 

  56. Eftekhari A, Ahmadian E, Salatin S, Sharifi S, Dizaj SM, Khalilov R, Hasanzadeh M. Current analytical approaches in diagnosis of melanoma. TRAC Trends Anal Chem. 2019;116:122–35.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported in part by the National Natural Science Foundation of China (Grant No. 62072145), and the Natural Science Foundation of Heilongjiang Province (Grant No. LH2019C042).

Author information

Authors and Affiliations

Authors

Contributions

JH conceived and designed the study. JQ, XL, and YH developed the method. JQ, QW, and JL analyzed the data and implemented the methodology. JW and YJ provided constructive discussions. JH and JQ drafted the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Ying Jiang or Junwei Han.

Ethics declarations

Ethics approval and consent to participate

The patient data analyzed in this work were acquired from publicly available datasets.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Figure S1.

Correlations between co-mutations of signaling pathways and tumor mutational burden, neoantigen load. Figure S2. Correlations between SpHe-comut status and Tumor mutational burden or neoantigen load. Figure S3. Comparison of the objective response rate between the SpHe-comut+ and SpHe-comut groups. Figure S4. Univariable and multivariable Cox analysis of SpHe-comut status and clinicopathological factors (Age, Sex, and PD-L1 expression) for PFS or OS in the ICBs cohorts. Figure S5. Comparison of performance for predicting ORR among SpHe-comut, DDR-comut and TMB as a predictive biomarker. Figure S6. Combining SpHe-comut status with DDR-comut status for the prediction of ICB therapy. Figure S7. Comparison of SpHe-comut status with NOTCH and GMS for the prediction of ICB therapy in the Rizvi cohort. Table S1. Data source. Table S3. Gene list of immune-related gene signature. Table S4. The significant survival-associated signaling pathways identified by the multivariate Cox regression model adjusted by clinical factors. Table S5. Comparison of tumor mutational burden between SpHe-comut+ and SpHe-comut groups across different cancer types. Table S6. Comparison of neoantigen load between SpHe-comut+ and SpHe-comut groups across different cancer types.

Additional file 2: Table S2.

The Information for time period and dose selection of involved cohorts.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qiu, J., Li, X., He, Y. et al. Identification of comutation in signaling pathways to predict the clinical outcomes of immunotherapy. J Transl Med 20, 613 (2022). https://doi.org/10.1186/s12967-022-03836-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-022-03836-3

Keywords