Landscape of genetic variation of m6A-related exosome genes in colon cancer
Figure 1a summarized the relationship between m6A regulators and exosome-related genes. First of all, a colossal amount of differential expression genes were selected from exosomes in serum between colon cancer patients and normal people, which named as exosome related genes. And then, those associated to m6A regulators were sorted out and we obtained 59 m6A-related exosome genes. Initially, it is commonly acknowledged that somatic mutation and CNV (copy number variation) are linked with cancers [14, 15]. Therefore, we prefer to acquire a preliminary knowledge of frequency of somatic variation and CNV of 59 m6A-related exosome genes in CC patients. In 399 patients, mutations of m6A-related exosome genes occurred on 159 patients, accounting for 39.85%. The histogram displayed that RNF43 was ranked first on mutation frequency and ZNF423 subsequently followed. But there are not any mutations exhibiting in TFAM, ALKBH5, C12orf57, GOLGA8A, GOLGA8N, GTF2F2 and NPIPA1 (Fig. 1b). In the following analyses shown the prominent relationship of mutation co-occurrence relationship including ANKRD12 and RNF43, RBM39 and CUL2, besides THOC2 and C2CD5 (Fig. S1a). Through observing distinct CNV alteration of 59 m6A-related exosome genes, it is obviously found that copy number amplification occupied with a majority of alterations, whereas CNV deletion happened in LYSMD3, PGGT1B and SRFBP1 (Fig. 1c). Through the analysis of somatic mutation and CNV, it suggested that the chosen 59 genes, which were delivered by exosomes, probably derived from tumors. Subsequently, Fig. 1d exposed that the mutated site of m6A-related exosome gene on colon cancer patients’ chromosomes. Then, we enabled to tell CC patients and normal samples apart according to different expression of 59 m6A-related exosome genes (Fig. 1e). To assure whether expression of m6A-related exosome genes was affected by the aforementioned genetic variations in CC patients, we found that the changes of CNV could exert prodigious influence on perturbations of m6A-related exosome gene expression through studying mRNA expression of these 59 genes between normal and CC samples. In addition, m6A-related exosome gene with amplificated CNV in CC tissues showed a remarkedly higher expression versus normal colon tissues such as TOMM34 and RBM39 (Fig. 1c and f). No matter in the transcriptome or genomics, there was significant difference between patients and normal people in 59 m6A-related exosome genes, suggesting that the expression imbalance of m6A-related exosome gene had positive influence on the CC genesis and development.
m6A methylation modification patterns mediated by 59 m6A-related exosome gene.
We first enrolled six GEO datasets (GSE17536, GSE29621, GSE33113, GSE37892, GSE38832 and GSE39582) which contained clinical data and overall survival (OS) data into one meta-cohort. The comprehensive landscape of m6A-related exosome gene interactions for CC patients was depicted with the m6A-related exosome gene network (Fig. 2a). The relationship between those exosome related genes were shown in Fig. S1a, and HR value of the genes were shown in Fig. S1b. Then based on the expression of 59 m6A-related exosome genes, we were able to divide CC patients into three qualitative m6A-related exosome gene modification patterns using the R package of ConsensusClusterPlus. And three patterns (442 cases in pattern 1, 263 cases in pattern 2 and 361 cases in pattern 3) were distinguished by unsupervised clustering algorithm. Finally, these patterns were called as cluster 1-3, respectively (Fig. S2). The tremendous survival preponderance in cluster 1 was disclosed by prognostic analysis for three modification subtypes (Fig. 2b).
TME cell infiltration characteristics in distinct m6A-related exosome gene modification patterns.
For the reason that the biological behavior between the three m6A modification patterns was not completely understood, we conducted gene set variation analysis (GSVA) of hallmark gene sets in six GEO datasets (GSE17536, GSE29621, GSE33113, GSE37892, GSE38832 and GSE39582). As is shown in Fig. 2c, cluster 3 was markedly enriched in mismatch repair, pyrimidine metabolism and Toll-like receptor signaling pathway (Fig. 2c). However, we observed that innate immune cells such as NK cell, macrophage, MDSC and so on were conspicuously enriched in TME cell infiltration of cluster 2 (Fig. 3a). Previous studies defined that tumors are classified as three phenotypes including immune-excluded, immune-inflamed and immune-desert. And immune-excluded phenotype was characteristic of plentiful immune cells around the tumor cell nest, yet the tumor capsules showed powerful protectivity from penetration of immune cell. And it also demonstrated that poor efficiency on tumor penetration of immune cell may be caused by stromal activation [16]. Hence, we hypothesized that the anti-tumor function of immune cells in cluster 2 was restricted by stromal activation. And it is confirmed by subsequent analyses that the activation of epithelial-mesenchymal transition (EMT), transforming growth factor beta (TGFβ) and angiogenesis pathways, which is relevant to stromal activation in tumor, were remarkably increased in cluster 2 (Fig. 3b). On account of the mentioned analysis, cluster 2 which was featured with innate immune cell infiltration and stromal activation, corresponded with immune-excluded subtype. Cluster 3 which was featured with specific immune cell infiltration and immune activation, corresponded with immune-inflamed subtype; According to the comparison between cluster 1 and 3, it revealed that cluster 1 was not concentrating on antigen processing and presentation, chemokine signaling pathway and cytokine-cytokine receptor interaction which is associated with adaptive immune. For that cluster 1 which was featured with immunological ignorance, corresponded with immune-desert phenotype (Figs. 2c-d and 3a-b). Next, with the method of spearman’s correlation analysis, the mutual effect between each tumor-infiltrating immune cell type and each m6A-related exosome gene was demonstrated (Fig. S3).
m6A methylation modification patterns in GSE39582 cohort
Putting our eyes on the GSE39582 cohort is beneficial for us to make a comprehensive understanding of m6A-related exosome gene modification patterns in numerous clinical cases. Consequently, using unsupervised clustering algorithm, three patterns were distinctly classified in GSE39582 cohort (Fig. 3c and Figs. S4a-d). Also, among three different m6A-related exosome gene modification patterns, the prominent difference was shown on the Principal Component Analysis (PCA) scatter diagram. (Fig. 3d). Marisa et al. innovatively classified patients who were suffering from colon cancer into four dominant molecular subtypes including CSC (cancer stem cell), CIN (chromosome instability), KRASm (KRAS mutant) and dMMR (defective mismatch repair). And they concluded that CIN associated with up-regulation of the EMT pathways while dMMR associated with up-regulation of the immune pathways and cell proliferation. However, EMT is downregulated in KRASm subtype [17]. Consistent with the previous findings, CIN subtype patients were mostly divided into cluster 2 and 3, while dMMR subtype predominantly focused in cluster 1 (Fig. 3e). Furthermore, prognostic analysis depicted that cluster 1 had a noteworthy advantage of survival rate comparing to cluster2 and 3 (Fig. S4e). Also, we found that there was different expression of m6A regulators among three m6A methylation modification patterns (Fig. S4f).
Generation of m6A-related exosome gene signatures and functional annotation
With the purpose of discovering unknown biological functioning of m6A-related exosome gene modification patterns, we used limma package to identify 3787 m6A-related exosome genes phenotype-related DEGs (differential expression genes) (Fig. S4g). And then GO enrichment analysis for differential expression genes was exhibited by the clusterProfiler package. As a consequence, 3787 selected DEGs, which were regarded as m6A-related exosome gene signatures, were denoted as the pivotally salient indicator of three m6A-related exosome gene modification patterns. Obviously, these signatures performed significant abundance of biological processes that corresponded with the formation of exosome, m6A modification and RNA transport, which verified tumors can deliver m6A-methylated RNA by exosomes to target cells (Fig. 3f). Owing to the absence of validating to this supervision mechanism, we applied unsupervised clustering analysis which based on the selected 3787 signatures to divide patients into different genomic subtypes and obtained three newly-established distinguishing subtypes called m6A-related exosome gene cluster A-C respectively (Figs. S5a-S5d and Fig. 4a). This phenomenon ensured again that there were three distinguishing m6A-related exosome gene modification patterns existing in colon cancer. Patients with colon cancer in gene cluster A (213 patients) ranked first in the prognosis analysis. While the worst survival rate was shown in gene cluster B, with 85 patients classified. 259 patients were classified in gene cluster C, which were regarded as an intermediate prognosis (Fig. 4b). Furthermore, among three gene clusters (A-C), it was discovered that the prodigious difference in 24 m6A regulators expression was consistent with the anticipated outcome of m6A methylation modification patterns (Fig. 4c).
Characteristics of clinical and transcriptome traits in m6A-related exosome gene phenotypes.
For the sake of figuring out the function of m6A-related exosome gene phenotypes in the TME immunity moderation, we would choose some cytokines and chemokines of three groups, which were gathered from published articles online. First, TGRB1, SMAD9, TWIST1, CLDN3, TGFBR2, ACTA2, COL4A1, ZEB1 and VIM denoted as the transcripts of transforming growth factor (TGFβ)/ EMT pathway. Secondly, PD-L1, CTLA-4, IDO1, LAG3, HAVCR2, PD-1, PD-L2, CD80, CD86, TIGIT and TNFRSF9 were relevant to the transcripts of immune checkpoints. And third, TNF, IFNG, TBX2, GZMB, CD8A, PRF1, GZMA, CXCL9 and CXCL10 were to be correlated with the pathways of immune activation [18, 19]. We considered that gene cluster C was supposed to be stromal-activated group because of its increased mRNA expression of TGFβ/EMT pathway. However, transcriptional mRNAs correlating with immunity system activity were up-regulated in gene cluster A, which suggesting it probably was categorized as the immune-inflamed group (Figs. S5f-5h). The next step is to pick out a portion of known signatures in CC patients on the purpose of describing the function of m6A-related exosome signatures (Fig. S5e). As is expected, these box plots showed that gene cluster C was featured with higher stroma activity and cancer progression and metastasis such as increased EMT and WNT target, while cell cycle, DNA replication and mismatch repair were remarkably enhanced in gene cluster A.
In order to more accurately predict the patterns of m6A methylation modification in a single tumor, we should make use of an extra method to compensate for the error caused by the heterogeneity of each tumor. Hence, we developed a scoring scheme called the MREGS (m6A-related exosome gene score), which is based on the identified 3787 signature genes to calculate the score of individual CC patients. On account of the complicacy of m6A-related exosome gene modification, we used an alluvial diagram to observe the flow of data (Fig. 4d). Furthermore, by exploring the relationship between MREGS and some recognized signatures, we could have more knowledge of m6A-related exosome gene signatures. And it was found that MREGS was positively associated with EMT-, TGFβ-related pathway and angiogenesis, whereas was negatively related with DNA damage repair and mismatch repair (Fig. 4e). Also, there was prodigious difference exited in MREGS between m6A-related exosome gene clusters (cluster 1, 2 and 3) by Kruskal-Wallis test. It displayed that cluster 2 corresponded with high MREGS but cluster 1 represented lower MREGS relatively (Fig. 4f). Additionally, as the activation of fibroblasts are dependent on TGFβ secreted by immunocytes or cancer cells and IL-11 that secreted by TGFβ1-stimulated CAFs (cancer-associated fibroblasts) could enhance the survival rate and invasion ability of cancer cells[20, 21], we chose to analyze stromal activation-related and TGFβ pathways. And we found that increased stromal activation and TGFβ pathway activity in fibroblasts were dominantly corresponded with high MREGS (Fig. 4g). Moreover, dMMR subtype ranked last in MREGS, while CSC subtype was the highest (Fig. 5a). The mentioned description powerfully elucidated that low MREGS was associated with DNA damage repair and high MREGS was associated with stromal activation. Furthermore, we make an attempt to ascertain the significance of MREGS in forecasting patients’ prognosis. Patients with low MREGS demonstrated a prominent survival benefit (Fig. 5b), because their 5-year survival rate was two times higher than patients with high MREGS. Whether MREGS could act as a robust prognostic biomarker for colon cancer is the next question we were supposed to solve. So we made use of Multivariate Cox regression model analysis which contained the element of patients’ gender, age, stage, tumor location and MMR status, subtype and confirmed MREGS as a creditable and independent prognostic biomarker for evaluating patient outcomes (Figs. S6a-b). Due to adjuvant chemotherapy (ADJC) is a common treatment after colon cancer surgery, we designedly validated MREGS to predict the efficacy of ADJC in patients suffering from colon cancer. We discovered that patients with low MREGS-ADJC displayed remarkable adverse reactions compared with patients who did not received adjuvant chemotherapy. While ADJC plays a positive role on patients with high MREGS. The high MREGS-ADJC patients displayed pivotal therapeutic advantages compared with patients who did not received adjuvant chemotherapy. The other obtained results indicated that low MREGS patients invariably displayed the tremendous survival advantage regardless of ADJC treatment (Fig. 5c). Moreover, according to mutation patterns, tumors can be classified into dMMR and pMMR. DMMR (defective mismatch repair) had more mutation burden than pMMR (proficient mismatch repair)[22]. And then we discussed the correlation between MREGS and molecular subtype and discovered that pMMR subtype were distinctly related with higher MREGS. In addition, in the stage Ⅳ patients, there was a remarkable enhancement in MREGS comparing to three other groups (Fig. S6b). This is strongly consistent with the result that dMMR subtype was related to higher survival comparing with pMMR subtype. These phenomena illustrated MREGS could also indirectly assess a portion of clinical features such as MMR subtypes and clinical stage, etc.
Characteristics of m6A-related exosome gene modification in TCGA molecular subtypes and tumor somatic mutation
To comprehensively study the characteristics of m6A-related exosome gene modification patterns, we introduced TCGA project. Three phenotypes classified by TCGA project is comprised of chromosomal instability (CIN), invasive and microsatellite instability (MSI). Next, the difference of MREGS among three phenotypes was calculated. The higher MREGS was pronouncedly concentrated on CIN and had a shorter lifespan, whereas invasive phenotype is relevant with lower MREGS, which was related to better survival (Figs. 5d-e). Different stages indicated differential expression of m6A-related exosome genes in TCGA project(Fig. 5f). Additionally, we used the maftools software package to analyze the differences in the distribution of high MREGS and low MREGS somatic mutations in the TCGA-COAD cohort. Low MREGS group presented more extensive tumor mutation burden than the high MREGS group (Figs. 5g-h). Accumulated evidence demonstrated a potential connection between enhanced survival rate whose receiving PD-1/PD-L1 immunological therapy and higher somatic tumor mutation burden (TMB) was discovered. Consequently, it was indirectly elucidated that clinical reactions to some immunological treatment like anti-PD-1/PD-L1 drugs may depend on different m6A-related exosome gene modification patterns in tumors. Also, we confirmed MREGS as a reliable method to predict prognostic outcomes after immunotherapy. In both clinical trials and preclinical studies, higher TMB patients with receiving immune checkpoint inhibitor treatment has a prominent superiority in survival rate and clinical response [23].
m6A-related exosome gene modification patterns in the role of anti-PD-1/L1 immunotherapy
With the purpose of testifying the reliability of MREGS and its prognostic value, MREGS signatures, obtained from GSE39582 cohort, was applied to five other colon cancer datasets (GSE17536, GSE29621, GSE33113, GSE37892, GSE38832; Figs. S8c-g). The ROC curve was used to evaluate the accuracy of a model, and higher the AUC value, the more accurate it is. And MREGS model displayed high accuracy of predictive superiority in patients with 3-year (AUC=0.785) and 5-year (AUC=0.754) colon cancer (Figs. S8h-i). Immunological therapy exemplified by anti-PD-L1/ PD-1 drugs have strikingly made immense progress in molecular targeted cancer therapy in recent years. Therefore, we selected two immunotherapy datasets (IMvigor210, GSE78220) to validate the predictive ability of MREGS to those patients who had been treated PD-L1/ PD-1 inhibitors. In anti-PD-L1 dataset (IMvigor210), through analyzing clinical response and survival rate, we exposed that low MREGS group surpassed high MREGS group (Figs. 6a-c). While in anti-PD-1 dataset (GSE78220), there was no significant difference between patients with low MREGS and high MREGS (Figs. 6d-f). The following analysis unraveled TME stroma and TGFβ pathway in fibroblasts were notably activated in high MREGS group, which mediated tumor migration (Fig. 6h). Tumor neoantigen burden (TNB) is associated with efficacy of immunological therapy and a major element in the judgement of clinical immunotherapies. And high TNB would be expected to be characterized by extensive T cell responses and to be specifically sensitive to immunotherapy[24]. And then we discovered that low MREGS group with high neoantigens displayed significant preponderance in survival rate (Fig. 6i). As aforementioned, MREGS, quantification of m6A-related exosome gene signatures, is anticipated to be an underlying and reliable biomarker for evaluating patients’ outcomes and clinical manifestation after treating immunological therapy (Fig. 6j). All in all, calculated from m6A-related exosome gene signatures, MREGS is available for forecasting patients’ clinical response whose receiving anti-PDL1 drugs and being a referable indicator for judgement of surgeons.