In this study, we analyzed the transcriptomic profiles from public databases and uncovered 606 m6A/m5C-related lncRNAs in ESCC. By consensus clustering analysis, we classified these ESCC samples into three clusters based on lncRNA expression profiles and found that cluster1 was associated with a significant better prognosis than cluster2 and cluster 3. The survival difference reason was investigated by comparing the immune landscape. As we expected, patients in cluster 1 displayed higher immune cell proportions in CD4 memory T cells and CD8 T cells. Furthermore, most of the immune checkpoint genes were notably increased in cluster 1 than the other two subgroups, indicating that immunity plays a major role in ESCC prognosis.
LncRNA represents a diverse category of RNA molecules with intricate roles in regulating gene expression by engaging gene regulatory proteins and microRNAs. Similar to protein-cording RNAs, lncRNAs also undergo RNA methylation that contributes to their regulatory functions in the context of tumorigenesis. We employ the lasso method and survival analysis to identify ten m6A/m5C-related lncRNAs associated with ESCC prognosis, including LINC00847, TTTY15, and LINC00942. Notably, LINC00847 has been extensively studied in various cancer types. It emerges as a key player in laryngeal squamous cell carcinoma, and its overexpression has been linked to enhanced cell proliferation and cell cycle progression. Additionally, elevated LINC00847 levels are correlated with lymph node metastasis and poor differentiation, suggesting its potential as a prognostic biomarker [24]. Our results further corroborate the significance of LINC00847 in ESCC survival, consistent with previous research. Mechanistically, LINC00847 is induced by the transcription factor E2F, exerting its influence on cancer progression through the modulation of downstream targets, including miR-147a [25].
An additional integrative analysis of the single-cell RNA-seq dataset has unveiled a compelling close association between LINC00847 and prognosis in lung adenocarcinoma. LINC00847 positively correlates with the infiltration of various immune cell types, and its overexpression significantly down-regulates PDL1 expression in the in-vitro assay, thus casting it as a prospective candidate in tumor immunotherapy[26]. Furthermore, the male Y chromosome-linked lncRNAs TTTY15 assume a crucial role in carcinogenesis across diverse cancer types. Acting as an RNA sponge, TTTY15 engages several miRNAs to promote cancer progression, exemplified by its interaction with miR-29-3p in colorectal cancer [27], miR-98-5p [28] and miRNA let-7a-5p [29] in gastric cancer. In prostate cancer, TTTY15 exhibits prominent upregulation in most tumor samples, exerting a pro-carcinogenic influence by sponging miRNA let-7, subsequently elevating CDK6 and FN1 expression [30]. Interestingly, TTTY15 assumes a contrasting suppressive role in NSCLC, wherein its overexpression inhibited cancer proliferation and metastasis through the modulation of TBX4 [31].
Moreover, epigenetic modification of lncRNAs in regulating tumorigenesis and development has been reported. In breast cancer, it has been documented that LINC00942 directly engages methyltransferase METTL14, thereby facilitating METTL14-mediated RNA methylation processes downstream [32]. Moreover, LINC00942 has been implicated in promoting chemoresistance in gastric cancer by impeding the degradation of oncoprotein MSI2. This, in turn, enhances the stability of c-myc mRNA, a phenomenon reliant on m6A modification [33]. Another lncRNA of interest, EGFR-AS1, arising from the reverse strand of lncRNA EGFR, has garnered attention for its overexpression in diverse cancer types. Elevated levels of EGFR-AS1 have been closely associated with unfavorable clinical features, encompassing pathological stage, lymph node metastasis, and overall survival [34]. Functionally, EGFR-AS1 serves as an oncogene, fostering cell proliferation, chemotherapy resistance, invasion, and stemness through intricate interactions with downstream miRNAs and signaling pathways. EGFR-AS1 notably stabilizes EGFR mRNA, consequently activating the PI3K/AKT pathway to promote proliferation and metastasis in renal cancer cells [35].
Furthermore, EGFR activation has been linked to the up-regulation of PDL1 via the p-ERK1/2/p-c-Jun pathway, thereby inducing immune evasion in EGFR-driven cancer [36]. In squamous cell carcinoma, EGFR-AS1 emerges as a mediator of EGFR addiction, influencing treatment responses. Notably, EGFR-AS1 knockdown has been shown to reverse resistance to tyrosine kinase inhibitors [37]. In ESCC, EGFR-AS1 has been implicated in up-regulating ROCK1 expression by sponging miR-145, thus promoting cancer cell invasion and migration [38]. Additionally, lncMIF-AS1 has demonstrated its significance in positively regulating NDUFA4 expression in gastric cancer cells. This regulation is achieved through the sequestration of miR‐212‐5p, which attenuates NDUFA4 mRNA degradation. Upregulation of NDUFA4 activates the oxidative phosphorylation pathway, ultimately promoting proliferation and inhibiting apoptosis in gastric cancer cells [39]. It is noteworthy that the biological roles of the remaining five lncRNAs remain unclear, thus providing new directions for future research endeavors. Collectively, these findings illuminate the indispensable role of m6A/m5C-lncRNAs in cancer development. Moreover, they posit these lncRNAs as promising candidates for novel prognostic biomarkers, holding potential implications for the prognosis of patients afflicted with ESCC.
In addition, we developed a ten m6A/m5C lncRNAs-based RiskScore model, which had a promising ability in survival prediction for both TCGA cohorts and independent validation datasets. Patients stratified into high and low-RiskScore subgroups manifest conspicuous distinctions in key clinical characteristics, encompassing survival status, gender, tumor stage, and grade. Additionally, the RiskScore model unveiled significant associations with immune cell infiltration, notably affecting CD4 T cells and Tregs proportion. To further improve its clinical utility, we constructed a comprehensive nomogram model incorporating clinical variables and RiskScore. This model performed excellent in predicting the survival probability of ESCC.
Immunotherapy has emerged as a pivotal therapeutic avenue in the management of gastrointestinal cancers due to its potential to yield rapid and substantial therapeutic benefits for afflicted patients. However, it remains a huge challenge to screen the dominant populations likely to mount a robust response to immune checkpoint inhibitors (ICIs). Here, our findings unveil a strong correlation between low-RiskScores and favorable responses to immunotherapy, signifying the potential use of this system in patient stratification for enhanced therapeutic outcomes.
Moreover, emerging evidence highlights that tumor-infiltrating immune cells were correlated with prognosis and immunity. The composition changes and functional activation of immune cell populations within the TME exert a profound impact on patient survival and their responsiveness to immunotherapy [40]. For instance, augmented infiltration of CD4 + lymphocytes has consistently demonstrated a significant association with prolonged survival in ESCC [41]. In this study we also characterized increased CD4 + T cell infiltration in low-RiskScore group, which displays a better prognosis. This observation aligns seamlessly with prior research, further reinforcing the notion that increased CD4 + T cell infiltration might be indicative of a more favorable prognosis.
Interestingly, we found the immune checkpoint genes were upregulated in the low-RiskScore group. Among these genes, CD226 deficiency restrains CD8 + T cell function, consequently curtailing the efficacy of cancer immunotherapy [42]. Aberrant expression of CD70 was linked to tumor progression and immunosuppression in the tumor microenvironment, and it can facilitate immune evasion through interacting with receptor CD27 [43]. TNFRSF9 is an activation marker for tumor-infiltrating Tregs, and inhibition of TNFRSF9 boosts anti-cancer treatments via reducing the immune suppressive function of Tregs [44]. These compelling findings collectively imply a potential mechanism whereby m6A or m5C methylation on lncRNAs may exert regulatory control over immune checkpoint gene expression. Such regulation contributes to a positive response to immunotherapy in the context of ESCC.
Certain limitations should be mentioned in our context. Firstly, our results of m6A/m5C-associated lncRNAs signature were not validated in a separate patient cohort, and the survival predictive value of the RiskScore model requires more external datasets validation for clinical application. Secondly, the potential molecular mechanism of these m6A/m5C-related lncRNAs remained unclear in ESCC, and more experimental research is needed.
In summary, our finding demonstrated a ten m6A/m5C-lncRNA signature implicated in ESCC progression and established a lncRNA signature-based RiskScore model for prognosis prediction. Moreover, we assessed the immune landscape and immune checkpoint gene expression for low-RiskScore patients, which might contribute to a beneficial therapeutic response from ICI. The RiskScore system might be a useful tool to determine the m6A/m5C-lncRNA signature application in clinical practice, thus promoting treatment decisions for selecting the patient subgroup that more cline benefits from ICI therapy.