Accurately predicting cancer prognosis is of significant value to patients, oncologists, and researchers. Nevertheless, it continues to be a challenge because of the heterogeneity of tumors. With the rise of high-throughput data comes the expectation that prognostic prediction may be improved by combining information from clinical and molecular biomarkers. We systematically used several available online databases, pursuing to discover promising biomarkers to predict the prognosis of GIC patients. We first comprehensively analyzed unfavorable prognostic genes among COAD/READ, LIHC, PAAD, and STAD and determined NPC2 and ITGAV as two probable prognostic markers that shared maximum overlap in four GICs. NPC2 is a protein-coding gene with a lipid recognition domain that has been demonstrated to be associated with lipoprotein metabolism and innate immune system pathways (32). High NPC2 protein expression has been detected in several cancers, such as breast, colon, and thyroid cancer (49, 50). Emerging pieces of evidence indicate that the ITGAV gene leads to the development and progression of various malignancies, such as colon carcinoma, pancreatic adenocarcinoma, esophageal adenocarcinoma, gastric cancer, hepatocellular carcinoma (51–55). Multiple mechanisms have been discovered so far to explain how ITAGV promotes tumor growth, such as epithelial-mesenchymal transition (EMT), proliferation, migration, and chemoresistance (53, 56). However, it was not fully understood how these genes were altered or what role they played in the GICs. Therefore, we used bioinformatics tools to delve further into the functions of these genes in GICs. We conduct an in-depth investigation of how NPC2 and ITGAV are expressed in human cancers and adjacent normal tissues. Furthermore, we show that NPC2 expression correlates with LIHC pathological staging, and ITGAV expression correlates with LIHC and PAAD pathological staging. In addition, we discovered that in PAAD and COAD, methylation levels of NPC2 and ITGAV are significantly elevated. These results were supplemented by our identification of linkages between NPC2 and ITGAV and the immune signature in the TIME.
In this study, our data suggest that the mRNA expression level of NPC2 was higher in several tumors, particularly in LIHC, PAAD, and STAD. Besides, ITGAV expression was higher in ESCA, PAAD, and STAD. Next, we, on the one hand, found that, in GICs, NPC2 expression levels were significantly associated with LIHC prognosis (lower OS) and pathological stage. On the other hand, ITGAV was correlated with LIHC and PAAD pathological stage significantly but not with other GIC stages. Moreover, we provide evidence that ITGAV expression levels were associated with worse OS of LIHC and OS, PFS of STAD patients. These results suggest that NPC2 and ITGAV may be useful as novel pathological staging markers for LIHC, PAAD, and STAD; nevertheless, it would be noteworthy to elucidate further the mechanism by which NPC2 and ITGAV are only linked with LIHC, PAAD, and STAD prognosis or pathological staging.
Cancer refers to a group of illnesses characterized by the unrestrained proliferation of aberrant cells, which is supposed to be predominantly the result of genetic mutations. Some of these mutations are dubbed "drivers" because of their tendency to spur carcinogenesis and provide certain selective advantages for precancerous cells over their neighboring cells. Moreover, it has become apparent that cancer genomes also include several "passenger" mutations. Mutational patterns in driver and passenger genes in cancer genomes shed light on the varied cancer phenotypes (57). It is interesting to note that our data suggests a mutation rate of 0.5% for NPC2 and 4% for ITGAV in GICs.
Currently, DNA methylation analysis is an emerging method for improving the accuracy of pathological diagnosis and prognosis of several malignancies (58, 59). We provide preliminary evidence that, according to the UALCAN database, NPC2 and ITGAV DNA methylation levels are reduced in PAAD and COAD, respectively. Based on the UALCAN database, we report preliminary information that promotor methylation levels for NPC2 and ITGAV are increased in PAAD and COAD, respectively. It has been shown that suppressing tumor suppressor genes by hypermethylation of DNA regions in the promoter region may modulate the pathophysiology of GICs. Cancer-related genes frequently regulate the DNA repair, cell cycle, apoptosis, and tumor-specific signaling pathways; However, promoter hypermethylation can cause genomic instability (60). A growing body of research demonstrates a significant relationship between NPC2 and ITGAV genetic aberrations and carcinogenesis. According to earlier research, abnormal NPC2 expression is significantly linked to a less favorable prognosis for glioblastoma (61). Using a panel of monoclonal antibodies against NPC2, Liao et al. (50) identified NPC2 overexpression in breast, colon, and lung cancers. In addition, Taguchi et al. (62) confirmed that human and mouse lung adenocarcinoma cells abundantly secreted NPC2. It has been demonstrated that ITGAV expression increases in a variety of epithelial tumors despite being practically undetectable in normal tissue, which highlights the possible role of integrin expression during cancer development (52). Integrins participate in cell surface adhesion and signaling and conclusively play crucial roles in cancer development. For illustration, ITGAV overexpression stimulated the synergistic effect of integrin and selectin, which promoted adhesion between pancreas cancer cells and peritoneal mesothelial cells, ultimately resulting in the development of PAAD (51). The association between ITGAV expression and poor prognosis in individuals with LIHC was uncovered by Kang et al. (63). As reported by Kemper et al., high levels of ITGAV expression have been found as a risk factor for PAAD. (51). One point that should be noted is that there are no reports about the association between cancers and methylation of these genes, indicating the novelty of our study. Overall, the fact that NPC2 and ITGAV expression makes it possible to identify numerous cancerous tissues of the GI tract from their noncancerous counterparts highlights the possibility of the aforementioned gene expressions in screening GICs.
Moreover, we carried out a related-TF prediction for NPC2 and ITGAV and Identified thirty-four common TFs between these two genes. Most of these TFs have significant roles in cancer development. For instance, the transcription factor Yin Yang 1 (YY1) participates in various biological functions, such as cell proliferation (64), invasion, migration (65), and EMT (66). Therefore, YY1 is critical for tumor progression, and increasing evidence suggests a close association between YY1 and cancer. Recently, Sato et al. (67) declared that YY1 suppressed the expression of ITGAV, and this transcriptional regulation may lead to the suppression of CRC cell migration and invasion.
We explored the PPI network of NPC2 and ITGAV and found 20 genes associated with NPC2 and ITGAV (10 genes for each). These NPC2 or ITGAV-related genes all play a role in various cancers. For example, Chen et al. (68) corroborated that low expression of NPC1L1 and NPC2 in HCC tissues might prompt poor clinical outcomes in HCC patients after resection. NPC1L1 and NPC2 combination was an independent prognostic factor for OS and time to recurrence (TTR) in HCC patients (68). Furthermore, there is clinical evidence that ITGB1 expression has been significantly associated with poor prognosis in CRC patients (69). In conclusion, our discovery of NPC2 and ITGAV-related genes provides new insights for the diagnosis and treatment of GICs.
Tumor development is profoundly affected by TIME, which consists of tumor cells and infiltrating immune cells. Immune cells were identified as the key component in controlling cancer development among the many TME factors (70). Therefore, we looked into the relationship between NPC2 and ITGAV and immune cell infiltration in GICs and discovered a positive correlation between these genes and the infiltration of several immune cells, including CD8 + T cells, CD4 + T cells, macrophages, neutrophils, and dendritic cells. These data suggest that NPC2 and ITGAV may play a significant role in the infiltration of immune cells and, ultimately, the TIME of the GICs. An impressive finding of this study was that the expression of NPC2 and ITGAV were significantly correlated with the abundance of macrophage infiltration in all kinds of aforementioned GICs. According to reports, macrophages upregulate PD-L1 expression via tumor growth factor (TGF-β)—induced EMT, which plays a crucial role in tumor immunosuppression and immune evasion (71). Taking into consideration that the expression of NPC2 and ITGAV causes an increase in macrophage infiltration, this finding may provide more evidence for the association between NPC2 and ITGAV expression and a dismal prognosis for GIC patients. In light of the above results, we next evaluated the association between NPC2 and ITGAV with immunomodulators and immunosubtypes. The expression of NPC2 and ITGAV was significantly related to the majority of immune modulators, especially CD274 (PD-L1), CD96, CSF1R, HAVCR2, and PDCDCDLG2. With the discovery of tumor-related immune inhibitors, immune checkpoint inhibitors (ICIs) have been extensively used in immunotherapy with remarkable results. Meanwhile, PD-1/PD-L1 inhibitors have been authorized as a treatment for various cancers, including COAD, LIHC, PAAD, and STAD (72–75). Further, we determined that ITGAV expression was linked to COAD and STAD immune subtypes, whereas NPC2 expression was linked to LIHC and STAD immuno-subtypes. Therefore, it is plausible to hypothesize that NPC2 and ITGAV expression may govern the infiltration level of tumor immune cells and immunological responses, and accordingly alter the prognosis of tumor patients.
With the use of the DGIdb database, ten candidate drugs targeting ITGAV were identified. Among them, for Abituzumab (NCT03688230, NCT01008475, and NCT00848510), Volociximab (NCT00401570), and Etaracizumab (NCT00027729 and NCT00284817) clinical trials on GICs’ patients have been conducted or are being conducted. In one of these clinical trials, exploratory analyses indicated that abituzumab-based therapy might be effective in patients with mCRC with high ITGAV (particularly αvβ6 member) expression who have otherwise poor prognosis (76). There are a number of limitations in our study that need further examination. Before anything else, it's important to note that the samples and clinical records in online databases were gathered retrospectively, and as a result, they skewed toward instances with fresh-frozen specimens of good quality and big tumor sizes in late-stage patients. Second, there may be uneven prognostic power and overfitting due to the small sample size of some kinds of cancer. Finally, NPC2 and ITGAV's molecular pathways in cancer are complex and need to be validated in fully independent research before being implemented in clinical practice.
Taken as a whole, our investigation into NPC2 and ITGAV in GICs revealed detailed information on gene expression, genetic alteration, clinical prognosis, and immune signature. These results will offer a solid basis for future molecular experiments of NPC2 and ITGAV in tumorigenesis and a rationale for developing innovative prognostic and therapeutic approaches for GICs patients.