CRC is one of the common lethal cancers in the world, and the treatment is mainly based on surgery, with chemotherapy and radiotherapy as adjuvant modalities[27]. As most patients developed recurrence or metastasis during the follow-up treatment, it directly decreased the survival rate of patients[28, 29]. Therefore, it is a continuous goal to explore new therapeutic means for the treatment of CRC. With the progress of oncology research, immunotherapy has become a novel member of antitumor therapy, and meanwhile the importance of the immune microenvironment in tumor progression was gradually unveiled. Notably, the tumor immune microenvironment (TIM) played a double-edged role in determining the 'life' of malignancy and was significantly associated with the survival of patients with malignant tumors[30]. Previous studies had revealed that the composition of the immune microenvironment in CRC directly affects the prognosis of CRC patients, for instance, the enrichment of lymphocytes and CD8+ T lymphocytes was beneficial to improve the survival of CRC patients. However, high levels of mast cells, tumor-infiltrating Th17 cells, cause poor prognosis in CRC patients[31, 32]. Currently, the stromal/immune scores were the most commonly used measure for the bioinformatic evaluation of immune cell infiltration in the tumor microenvironment[19]. Here, the ESTIMATE algorithm was used to calculate the stromal/immune score of CRC patients from the TCGA cohort, and we found that patients in the high immune score group had a higher survival rate compared to the low immune score group. The correlation between the immune scores and clinical characteristics of CRC patients was showed that there was a significant negative correlation between the immune score and some of the clinical characteristics (pathologic stage and pathologic M stage) of patients, and that the immune score progressively decreased with tumor progression. High immune scores, i.e., immune cell infiltration played a positive role in prolonging CRC patient survival, and this finding was consistent with previous researches on immune infiltration in CRC[31].
Macrophages reportedly played a vital role in the progression of multiple malignancies, and different cell subtypes played distinct roles in the progression of malignancy. M1 macrophages can activate the immune response by secreting pro-inflammatory factors such as IL-4 and INF-γ to inhibit the progression of malignant tumors, whereas M2 macrophages can inhibit the immune response by secreting cytokines such as IL-10 and TGF-β, to enhance the immune escape of malignant tumor cells and promote tumor progression[33–35]. In contrast to the findings of previous studies regarding the promotion of tumor progression by M2 macrophages, certain studies had shown that M2 macrophages, with CD163 as a marker, might have synergistic effects and participate in tumor suppression, and were positively correlated with good prognosis[36, 37]. Our study showed similar results, with an increased proportion of M2 macrophages infiltrating in the high immune score group. This may be attributable to the heterogeneity of malignant tumors and the low accuracy of RNA-seq based algorithms. CD8+ T cells, also known as cytotoxic T lymphocytes (CTLs), have been recognized as the main anti-tumor immune effector cells, and their high levels of infiltration were associated with a favorable prognosis in various cancers [38]. Minority studies demonstrated that CD8+ T cells had a significant positive correlation with the chemotherapeutic effectiveness and immunotherapy response rates[39, 40]. Results of our study were consistent with the extant consensus, which showed that the proportion of CD8+ T cell infiltration was second only to that of macrophages. The proportion of infiltrated CD8+ T cells was significantly higher in the high immune score group, compared to that in the low immune score group, for which the prognosis was worse. Correlation plots for the infiltration of 22 TIICs showed that CD8+ T cells were positively correlated with the infiltration of M1 macrophages (r=0.29), activated memory CD4+ T cells (r=0.49), follicular helper T cells (r=0.44), and were negatively correlated with M0 macrophages(r=-0.48). This also illustrates the complexity of the interaction of immune infiltrating cells during tumor progression.
Based on these results, we identified the DEGs between high and low immune score groups and performed functional enrichment analysis for DEGs with a positive regulatory role in the high immune score group. The results showed that DEGs were significantly associated with immune regulation in terms of the molecular functions, biological processes, and cellular components, and participated in immune-related pathways such as the PD-1 checkpoint pathway and chemokine signaling pathway. Programmed cell death 1 (PD1) and its ligand (PDL1) are critical regulatory physiological immune checkpoints. The PD-L1 present on tumor cells may inhibit T cell mediated immune responses by binding to the PD-1 receptor on the surface of T cells[41]. Antibody drugs that target PD-L1 or PD-1 to block the PD-1/PD-L1 pathway are highly effective for the enhancement of T cell anti-tumor immunotherapy[42]. The chemokine signaling pathway is an essential pro-inflammatory pathway, in which chemokines recruit leukocytes to infiltrate into cells and produce local inflammatory effects in response to an exogenous pathogen attack. Chemokine signals are conveyed by chemokine receptors (G-protein coupled receptors) expressed on immune cells. Upon the activation of these receptors, the α-and β-γ-subunits of G-proteins dissociate to activate different downstream pathways, leading to cell polarization and actin reorganization[43]. Thus, CRC patients can respond effectively to immunotherapies such as anti-PD-1 owing to the immune response generated by immune cells. Our results had demonstrated that immune scores were significantly negatively correlated with the progression of certain pathologic stages and the pathologic M stage in patients. Hence, we re-clustered DEGs by their pathologic stage and pathologic M stage in chronological order using a soft clustering analysis algorithm. Then, we analysed the clustered genes using univariate Cox regression to identify genes that were significantly associated with CRC prognosis. To avoid biased results resulting from the small sample size and homogeneity of the data platform, we downloaded the GSE39582 dataset from the GEO database as an additional dataset for the screening of variables. Upon the intersection of the candidate genes generated from the above two datasets, three immune-related genes were identified. Of these, DNASE1L3 and LGALS4 were protective factors and KCNE4 was a risk factor.
The results of candidate genes expression analysis showed that the expression levels of DNASE1L3 and LGALS4 in healthy patients were significantly higher than those in CRC patients, and were negatively correlated with tumor progression, patients with high expression levels of DNASE1L3 and LGALS4 received a favorable prognosis. However, expression levels of KCNE4 were significantly high in CRC patients, and were positively correlated with tumor progression, patients with a high expression level of KCNE4 received a worse prognosis. The expression levels of proteins associated with the three candidate genes were evaluated using the immunohistochemical specimens available in the HPA database. Moderate to high expression levels of DNASE1L3 and LGALS4 related proteins were observed in healthy colon/rectal tissues, while a low or negative expression level was observed in CRC tissues. KCNE4 related proteins were mainly moderately expressed in both healthy and tumor tissues. Since the number of samples identified via immunohistochemistry analysis of the database was limited, the differential expression of the relevant proteins could not be identified in healthy and cancer tissues. Therefore, the evaluation of varied expression levels in different tissues based on the only available sample size could result in somewhat inaccurate results, and this method was only used for a preliminary assessment of cellular localization and the expression level of the protein of interest. Subsequently, we primarily evaluated the correlation between the expression of three candidate genes and the infiltration of macrophages and CD8+ T cells. The results demonstrated that the expression of DNASE1L3 was positively correlated with the infiltration of M1 macrophages and negatively correlated with the infiltration of M0 macrophages. KCNE4 expression was positively correlated with the infiltration of M2 macrophages. It was suggested that the overexpression of DNASE1L3 might be associated with the differentiation or recruitment of M1 macrophages, while the overexpression of KCNE4 might be associated with the differentiation or recruitment of M2 macrophages. Based on the above comprehensive screening and analysis, we ultimately identified two immune related genes associated with survival in CRC patients, namely DNASE1L3 and KCNE4.
In the tumor microenvironment, macrophages constitute the highest percentage of cells, and they were involved in the progression of malignancy through direct or indirect processes, including angiogenesis, tumor cell invasion, metastasis, and regulation of the tumor microenvironment[44]. The surface molecules of macrophages performed an essential role in tumor progression. CSF1R (colony-stimulating factor 1 receptor) could assist tumor-associated macrophages to infiltrate into the tumor surroundings, of which associated ligand colony-stimulating factor 1 (CSF1) can also promote macrophage differentiation[45]. CD47 on the surface of tumor cells interacts with (signal regulatory protein alpha) SIRPA on the surface of macrophages to promote the immune escape of tumor cells[46]. Targeting molecular markers on the surface of macrophages could effectively reduce macrophage recruitment in the tumor microenvironment, differentiation, and immune escape of tumor cells[47]. As a primary member of the body's immune regulation, T cells protect the body from pathogens, tumors and other damage by recognizing specific antigens. However, the combination of tumor cells and tumor microenvironment during tumor progression can inhibit the anti-tumor effect of T cells, and even cause exhaustion of T cells[48, 49]. TIM-3 (T cell immunoglobulin and mucin-domain containing-3), also known as HAVCR2, which was produced on the surface of exhausted T cells undergoing chronic tumor antigen stimulation, had been used as a marker of T cell exhaustion, and targeted inhibition of TIM-3 may be expected to restore the anti-tumor capability of T cells[50, 51]. The information from the IMVigor210 cohort was utilized as an efficacy evaluation dataset for immunotherapy, and the analysis showed that there was a higher positive correlation between DNASE1L3 and PDCD1, compared to KCNE4, and that patients with high DNASE1L3 were able to benefit more from treatment with anti-PD-1. The survival prognosis of patients in the IMVigor210 cohort also confirmed the benefit, as the results showed that patients with high DNASE1L3 had prolonged survival. The correlation between candidate genes and immune checkpoint markers predicts, in part, the effect of immunotherapy. In the analysis of the TCGA CRC queue, there was a significant positive correlation between the expression of KCNE4 and multiple immune checkpoint markers, except for PDCD1. In the TCGA CRC cohort, DNASE1L3 was unsatisfactory in immunotherapy prediction, which may be related to tumor heterogeneity and limited samples. Nevertheless, DNASE1L3 correlated higher with PDCD1, CSF1R, and TIGIT compared to other immune checkpoint markers. Furthermore, the correlation of DNASE1L3 with PDCD1 was higher than that of KCNE4, which was generally similar to the results of the IMVigor210 cohort, and these findings suggested the potential predictive value of DNASE1L3 in predicting anti-PD-1 therapy in CRC patients.
DNASE1L3 is a member of the deoxyribonuclease 1 family, which has the capacity to degrade chromosomal DNA strands [14, 52]. DNASE1L3 is mainly secreted by bone marrow cells, and functional nuclear localization signals facilitate its transportation into the nucleus, where it participates in the fragmentation of nuclear endosomal DNA during apoptosis and necrosis[53, 54]. Previous studies have shown that DNASE1L3 was closely associated with the development of autoimmune diseases, and the absence or mutation of DNASE1L3 resulted in an anti-DNA response in the body. This in turn resulted in autoimmune diseases such as systemic lupus erythematosus and systemic sclerosis[55, 56]. In recent years, the prevalence of bioinformatics technology has resulted in the identification of the role of DNASE1L3 in cancer progression. For instance, DNASE1L3 was involved in signal transduction in breast cancer patients, and could be an independent prognostic factor for patient survival after the radical resection of liver cancer [57, 58]. The role of DNASE1L3 in colorectal cancer has been examined to a lesser extent. In our study, DNASE1L3 was found to be significantly associated with M1 macrophage infiltration in the CRC tumor microenvironment and significantly correlated with CRC progression and survival, hence, it might represent a new therapeutic target for CRC treatment.
KCNE4 is the fourth and largest member of the KCNE Kv channel regulatory subunit family [59, 60]. A previous study has shown that it was mainly expressed in abundant amounts in the human brain [61]. KCNE4 acts as an ancillary peptide of Kv1.3 and negatively regulates K+ channels on the surface of immune cells, mainly by controlling the abundance and localization of K+ channel proteins on the cell membrane surface, inhibiting outward K+ currents, and accelerating channel inactivation[15]. It plays an essential role in the proliferation, activation, and apoptosis of leukocytes and other physiological processes of the immune system, as well as in angiogenesis [62, 63]. The role of KCNE4 in malignant tumors has rarely been studied. Using bioinformatics techniques, we determined that KCNE4 was significantly expressed in CRC patients and was positively correlated with tumor progression. In addition, KCNE4 expression was also significantly correlated with patient prognosis and positively correlated with M2 macrophage infiltration in the tumor immune microenvironment. KCNE4 might play an important role in the tumor microenvironments of CRC patients and could represent a novel target molecule.
Although we have assessed several noteworthy immune-related factors that might be useful for CRC immunotherapy treatment, certain limitations were associated with our study. First, the heterogeneity of the immune microenvironment in malignant tumors inevitably leads to biased results. Second, despite combining the data from the TCGA and GEO databases for the screening of genes, the databases for validating the results were established based on data from the TCGA database with relatively small sample size. In addition, the immune scores of most malignant tumor tissues and the proportions of infiltrating immune cells were calculated using the ESTIMATE and CIBERSOTR algorithms, but because these algorithms are not completely stable, the results need to be confirmed via additional experiments.