A risk score model based on TGF-β pathway-related genes predicts survival, tumor microenvironment and immunotherapy for liver hepatocellular carcinoma

It is widely acknowledged that LIHC treatment is challenging for its high possibility of drug resistance. The development of clinically validated agents against LIHC has been significantly influenced by the complex interactions of liver tumors with their immune microenvironment and a lack of understanding of the heterogeneous mechanisms of LIHC tumorigenesis and progression [28]. Several studies have shown that dysregulated signals in the TGF-β pathway have important function in immune regulation in the LIHC microenvironment [8, 11]. Therefore, TGF-β pathway-targeted drugs, including drugs targeting TGF-β ligands, TGF-β receptors, and downstream mediators of TGF-β, have been explored and clinically tested. And all of those drugs can lead to a variety of synergistic downstream effects and may improve the clinical outcome of LIHC [29, 30]. At present, effective biomarkers should be discovered to help determine the response of tumor cells for LIHC patients [31].

This work developed a risk score model based on 8 TGF-β pathway-related genes through progressively screening 54 TGF-β pathway-related genes, which can score and group LIHC samples in independent datasets. Chen et al. found that about 40% of all LIHC samples showed at least one gene mutation in the TGF-β pathway [32]. In the high- and low-risk groups, TP53 was both identified to have the highest mutation frequency, which has been identified as a common molecular event in human liver cancer [33]. Previous studies have suggested that with the continuous acquisition of genomic mutations, tumor cells show a series of mutations in different signal pathways, resulting in changes in TGF- β response [34]. This also explained the function of TGF-β in the late stage of tumor was quite different from that in the early stage. In addition, GSEA showed that two risk groups had differential enriched pathways such as metabolism-related pathways like fatty acid metabolism, and drug metabolism were more enriched in low-risk group, which may be resulted from their differential mutation patterns in TP53 and metabolism-related genes.

A number of evidences show that TGF-β can modulate cellular responses that regulate the tumor microenvironment, which may also contribute to LIHC progression and drive immune escape of cancer cells [31]. On the comparison of tumor microenvironment and immune infiltration between two risk groups, we found that high-risk group had relatively higher enrichment of helper follicular T cells, Tregs and M0 macrophages. The previous study speculated that increased number of these immunosuppressive cells endowed high-risk group a strong immunosuppressive environment, leading to an unfavorable prognosis [34]. Importantly, two risk groups showed significantly different response to immunotherapy, where TIDE score of low-risk group was noticeably lower and responsive proportion was significantly higher, suggesting that low-risk group may be more responsive to anti-PD-L1 treatment. However, high-risk group was more sensitive to chemotherapeutic drugs or targeted drugs including cisplatin, imatinib, sorafenib and salubrinal and pyrimethamine. These observations suggested that the prognostic model had a potential in guiding immunotherapy or targeted therapy for LIHC patients.

Eight prognostic genes (CDKN1C, HDAC1, SERPINE1, BMP2, ENG, FKBP1A, NOG, and BCAR3) involved in TGF-β pathway were included in our prognostic model. We found that some of them were also identified as prognostic biomarkers for cancers by the previous studies. Cyclin-dependent kinase inhibitor 1C (CDKN1C, also known as p57(KIP2)), a tumor suppressor, could regulate tumor cell differentiation, invasion, and angiogenesis, which is also validated as a prognostic biomarker in various cancer types, including in LIHC [35, 36]. In a 7-gene hypoxia signature developed by Bai et al., CDKN1C has also been identified as a prognostic gene for predicting LIHC prognosis [37]. Histone deacetylase 1 (HDAC1) is a critical enzyme for epigenetic modification, whose overexpression is strongly correlated with tumor cell proliferation and growth in many cancers [38]. High expression of HDAC1 is significantly associated with elevated cancer-specific mortality in LIHC [39]. Plasminogen activator inhibitor 1 (SERPINE1, also known as PAI-1), is considered as a prognostic biomarker for gastric cancer, gliomas, and colorectal cancer [40,41,42], hepatocellular carcinoma [43]. SERPINE1 was also in an 8-gene prognostic by Lin et al [44] High expression of bone morphogenetic protein 2 (BMP2) could promote liver cancer cell growth through activating myeloid-derived suppressor cells [45]. Other four prognostic genes were less reported in LIHC research.

The 8-gene prognostic model manifested favorable performance in different datasets, except in GSE10143 dataset with an unsatisfied AUC. Nevertheless, our model still outperformed other present prognostic models for LIHC in the same datasets (TCGA-LIHC and ICGC). 1-year, 3-year and 5-year AUC were 0.76, 0.71 and 0.70 in TCGA-LIHC dataset, respectively. 1-year, 3-year and 4-year AUC were 0.76, 0.75 and 0.64 in ICGC dataset, respectively. We included some studies containing at least one prognostic gene as our model. Sun et al. established a 2-gene prognostic model (CANX and HDAC1) for LIHC based on immune-related and autophagy-related genes using TCGA-LIHC and ICGC datasets [46]. The AUC of the 2-gene prognostic model for 1-year, 3-year and 5-year was 0.696, 0.639 and 0.642 in TCGA training dataset, 0.728, 0.685 and 0.612 in TCGA test dataset, 0.757, 0.669 and 0.644 in ICGC dataset, respectively. Lin et al. constructed an 8-gene prognostic model (SLC7A1, RIPK2, NOD2, ADORA2B, MEP1A, ITGA5, P2RX4, and SERPINE1) based on inflammatory response-related genes for LIHC also utilizing TCGA-LIHC and ICGC datasets [44]. The AUC of Lin et al’s model for predicting 3-year OS was 0.614 and 0.710 in TCGA-LIHC and ICGC datasets, respectively [44]. Compared with other prognostic models, our model was validated in more datasets, while they only validated their models in ICGC dataset.

This study had some limitations. Firstly, all the data were retrospective data, and experiments were not designed to verify them from other aspects. Secondly, our analysis was only based on TGF-β pathway-related genes, and the results did not represent all LIHC-related gene profiles. Thirdly, algorithms for characterizing tumor microenvironment, such as ESTIMATE and CIBERSORT, are not always accurate due to the atypical or unclear tumor microenvironment varying by tumor types. There was a possibility that an overlap of some gene signatures may exist between stromal cells and tumor cells because of the influence of epithelial-to-mesenchymal transition (EMT). In the future, the scope of research should be further expanded and experimental studies should be carried out to analyze the risk model based on TGF-β pathway-related genes on LIHC pathological behavior.

留言 (0)

沒有登入
gif