To date, inflammation has become a hotspot for all types of neoplastic diseases as well as preneoplastic diseases, the link between inflammation and tumorigenesis has been well established in the last two decades, and inflammatory bowel inflammation is an important risk factor for colon carcinogenesis[23]. The accumulation of inflammatory genetic alterations and loss of normal cellular regulatory processes are not only associated with cancer growth and progression but also lead to the expression of tumor-specific and tumor-associated antigens, which may activate antitumor immunity [24]. In the tumor microenvironment, antigen-specific adaptive immune responses are impeded, as soluble cytokines prevent T-cell infiltration, and cell surface signals are modulated to suppress lymphocyte-mediated killing activity[25]. Currently, MSI-H/dMMR is the only recognized biomarker that can be used to guide immunotherapy for colon cancer[26]. A small proportion of patients with microsatellite-stabilized (MSS) or proficient MMR (pMMR) mutations can benefit from immunotherapy, whereas a large proportion of MSI-H/dMMR patients exhibit intrinsic or acquired resistance to immunotherapy [27]. Therefore, the identification of new biomarkers or prognostic markers is crucial, and powerful biomarkers are needed for the stratification of colorectal cancer patients and assessment of the tumor immune microenvironment. Currently, studies have demonstrated the predictive ability of inflammation-related genes in colorectal cancer, and in this study, we screened genes in a novel way, identified new inflammation-related genes, and confirmed the value of the genes for prognostic prediction in patients with colorectal cancer through our validation.
In this study, we first classified patients into two different molecular subtypes based on IRG expression by an unsupervised consistent clustering method. Patients with IRG subtype A had poorer overall survival than those with subtype B, and there were also significant differences in pathways enriched in different subtypes and different immune cell infiltrates. We reclassified COAD patients according to the DEGs between the two subtypes. These results initially demonstrated the prognostic value of IRGs in COAD. In addition, we screened 13 genes with prognostic value using univariate Cox regression, LASSO regression, and multivariate Cox regression and then constructed risk-prognostic models. There were significant differences in survival between the high- and low-risk groups in the different cohorts, and the area under the curve (AUC) of the receiver operating characteristic (ROC) curve (AUC) curve (AUC) was also high. More importantly, the C-index, which evaluates the performance of the model, is superior to all other clinical characteristics. For better clinical application, we constructed a nomogram, and the ROC curves were able to predict the survival time of patients at 1, 3, and 5 years very well. In addition, there were significant differences in clinicopathologic correlation, TME, immune checkpoint analysis, TMB, and drug sensitivity between the high-risk and low-risk groups. Therefore, our constructed prognostic profile of inflammatory genes can assist in the clinical diagnosis of COAD patients, as well as the development of personalized and precise treatments for patients.
We screened 13 genes associated with inflammation and the prognosis of colon cancer patients. The TIMP1, GDF15, UCN, KRT4, POU4F1, NXPH1, SIX2, NPC1L1, KLK12, IGFL1, and FOXD1 genes have been studied in tumors in a correlative manner, and the ASPG and CYP4F8 genes were screened in other predictive models. For example, tissue inhibitor of metalloproteinases 1 (TIMP1) has been identified in a variety of genetic prognostic studies and was the first natural collagenase inhibitor to be discovered [28]. High TIMP1 expression is positively associated with poor prognosis in lung, brain, prostate, breast, colon and several other cancers [29]. TIMP1 causes tumor proliferation and metastasis through the FAK-PI3K/AKT and MAPK pathways in colon cancer [30]. ASPG, also known as 60 kDa lysophospholipase, is a protein with asparaginase, lysophospholipase, transacylase, and platelet-activating factor (PAF) acetylhydrolase activities and is an enzyme with antitumor activity [31]. In our model, ASPG is the gene with the largest coefficient with protective properties. To verify the impact of the database on different patients in our country, the results of our qPCR validation were consistent with the results in the database. FOXD1 plays a role in a wide range of tumors and can be of prognostic value in predicting the prognosis of patients with colorectal cancer through the ERK 1/2 pathway and promoting tumor progression[32, 33]. In conclusion, a portion of the genes for constructing predictive models are closely related to tumor development, and we also screened genes that are still lacking in tumor research; therefore, the molecular mechanisms and phenotypes of these biomarkers in cells need to be further investigated.
The role of the tumor microenvironment in oncology research has been clearly defined by researchers as a highly structured ecosystem containing cancer cells surrounded by multiple nonmalignant cell types. The TME includes a rich diversity of immune cells, cancer-associated fibroblasts (CAFs), endothelial cells (ECs), pericytes, and other cell types that vary by tissue[34]. In our analysis, the immune function of macrophages was upregulated in the high-risk group. Recent studies have shown that CXCL8 polarizes tumor-associated macrophages (TAMs) to the M2 phenotype, leading to increased M2 macrophage infiltration in CRC and reduced CD8 + T-cell infiltration, resulting in an immunosuppressive microenvironment [35]. In the low-risk group, the immune function of NK cells was upregulated, and NK cells also play a crucial role in bowel cancer. It is widely accepted that low levels of NK cell infiltration and/or impaired NK cell function are associated with poor overall patient survival and recurrence of CRC after treatment [36]. We also evaluated immune checkpoints that were differentially expressed between the high- and low-risk groups, and the risk scores showed significant positive correlations with important PD1, PDL1, PDL2, and CTLA4 checkpoints. Cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1), two inhibitory checkpoints commonly found on activated T cells, have been found to be the most reliable targets for the treatment of cancer. [37, 38]. The inflammatory cytokine interleukin 17 (IL-17) can upregulate PD-L1 protein expression in HCT116 CRC cells through activation of NF-κB and ERK1/2 signaling [39]. Smad7 is a negative regulator of TGF-β signaling, and its deficiency induces CD103 + PDL2/1 + DCs and Tregs to attenuate DSS-induced colitis [40]. Using single-cell sequencing, a recent study analyzed immune and stromal cell dynamics in neoadjuvant PD-1-blocked d-MMR/MSI-H CRC patients and showed that in patients with immunotherapy-pathologically complete remission (pCR) tumors, the proportions of CD8 + Trm-mitotic cells, CD4 + Tregs, proinflammatory IL1B + Mono cells, and CCL2 + fibroblasts after treatment were consistently decreased, whereas the proportions of CD8 + Tem cells, CD4 + Th cells, CD20 + B cells, and HLA-DRA + endothelial cells increased, providing yet another new insight into the mechanisms of ICI therapy[41]. When the level of inflammation in a patient's body is elevated, the response to ICIs is also decreased, and MSI-H CRC patients with inflammatory diseases are at increased risk of developing resistance to ICIs through neutrophil-associated T-cell depletion and hence ICI resistance [42]. In addition, in drug sensitivity analysis, patients at different risk levels have different sensitivities to drugs, and the treatment of cancer patients often requires a combination of multiple drugs; our risk scores can also guide the choice of drugs in the clinic. Our study suggested that higher expression of immune checkpoint molecules predicts that high-risk patients may benefit from these immunotherapies in the future.
Our study has several limitations: we performed external validation using only one database, GEO, and in the future, we will use several different databases for joint validation. Second, the TCGA database we analyzed contains Western case data and therefore lacks representative prospective data. We validated the expression level of only one model gene in vitro, and further validation of the mechanism of action of other genes in colorectal cancer is needed.