1. Introduction
Gastric cancer (GC) is a complex and multifactorial disease caused by genetic, epigenetic, and environmental influences [
1]. Although a worldwide decline in incidence and mortality rates has been observed in recent decades, GC still constitutes a significant burden and significantly impacts populations in developing countries. Current statistics reveal that GC is the third most common malignancy and the fourth leading cause of cancer-related mortality worldwide, accounting for more than 768,000 deaths in 2020 [
2,
3]. The National Cancer Institute (INCA) estimates for Brazil 13,340 new cases of GC in men and 8,140 in women in the three years from 2023 to 2025. These values correspond to an estimated risk of 12.63 new cases per 100,000 men and 7.36 per 100,000 women [
4].
Most gastric cancers are adenocarcinomas. According to Lauren’s classification, these represent more than 95% of all gastric malignancies and can be subdivided into intestinal and diffuse types. This classification is based on the histology of the tumor [
5]. The intestinal type consists of differentiated cancer with a tendency to form glands. It progresses mainly through successive changes in the normal gastric mucosa, leading to acute and chronic gastritis, atrophic gastritis, intestinal metaplasia, dysplasia and a gastric tumor [
5].
The standard treatment for gastric cancer is based on the triad: surgery, chemotherapy and radiotherapy. Surgical resection is considered the primary method of treatment at an early stage and the only potentially curative approach in treating gastric cancer. However, recurrence is frequently observed in many patients, even after resection. Faced with this problem, a study of 206 patients with gastric cancer revealed that patients in stages II and III had better survival rates with adjuvant chemotherapy than surgery alone [
6]. Over two decades ago, Phase II clinical studies have shown that neoadjuvant chemotherapy can increase tumor resection success rate by 72 to 87% [
7,
8].
Various chemotherapeutic drugs and therapeutic schemes are approved for the pharmacological treatment of gastric cancer. Some of these therapeutic schemes are well described in the literature: the FLOT scheme (5-fluorouracil, Leucovorin, Oxaliplatin and Taxane), the ECF scheme (epirubicin, cisplatin and fluorouracil), FOLFOX (5-fluorouracil, leucovorin and oxaliplatin), infusional cisplatin (CF) and single-drug regimen with irinotecan [
9,
10].
Nowadays, two problems related to drugs for GC need to be solved: 1) the high toxicity of drugs to patients; and 2) the resistance of GC cells to these drugs. At that point, bioinformatics was raised as a powerful tool to identify new targets or new molecules for a target. Over the last few years, bioinformatics has supported researchers worldwide to rapidly find molecules and targets that could be applied to cancer. For example, Miller et al. [
11] performed a structure-based virtual screening (SBVS) to find new molecules against the proteosome of pancreatic cancer cells. In the study, the authors analyzed 380,000 compounds against one proteasome subunit. After SBVS analysis, out of 288 compounds tested in vitro, 1 was selected for experiments analysis [
11]. This case is an example of how bioinformatics could rapidly help drug discovery.
Another study by Rohr et al. [
12] developed a pipeline employing R package scripts to analyze data from transcriptional analysis from micro-array from pre- and post-malignant colorectal cancer. Using the pipeline developed was possible to analyze a merged dataset containing 231 normal, 132 adenomas, and 342 colon cancer tissue samples across twelve independent studies analyzed by micro-array deposited in the Gene Expression Omnibus (GEO) [
12]. The analysis was done to understand the genes involved in the disease severity, progression, and new targets for treating colorectal cancer. Both studies employed bioinformatics to find new targets and molecules that could be useful in cancer treatment, corroborating bioinformatics applications in clinics.
The present study aimed to identify new potential targets involved in GC establishment in samples collected from different populations worldwide using bioinformatics tools. First, an R code script was employed to analyze the micro-array metadata from healthy patients and with GC downloaded from GEO searching for new genes involved in GC establishment. Compounds can interact with the proteins encoded by overexpressed genes (AJUBA, CD80), hypoexpressed (FBXL13 and CCDC69) and the main gene involved with the Hallmark E2F targets (NOLC1) in gastric cancer cell lines from the 601 samples analyzed, by blocking or modulating the site (orthosteric or allosteric) of these targets.
4. Discussion
Despite advances in science and the various therapeutic options currently available for controlling gastric cancer, the prognosis of patients affected by it remains unfavorable. In this context, chemotherapy resistance and the serious adverse effects of conventional pharmacological treatments represent a major obstacle to the successful treatment of gastric cancer.
Drug resistance is a phenomenon that results from a variety of pharmacokinetic and molecular changes. It refers to the ability of microorganisms or cancer cells to resist the effects of a normally effective drug against them. Although many types of cancer are initially susceptible to chemotherapy, with prolonged use, the development of resistance to chemotherapy drugs is observed through different mechanisms, such as inactivation or reduction of drug activation; changing drug targets; drug efflux; DNA damage, repair and metabolic changes; inhibition of cell death; epithelial-mesenchymal transition and metastasis; heterogeneity of cancer cells; and epigenetic modifications, with the possibility of combining any of these mechanisms [
33].
As a new type of approach, targeted therapies play an important role in the treatment of gastric cancer by interfering with gene expression or target proteins that play or regulate critical roles in tumor growth or progression, providing greater selectivity, efficacy and reduction of toxicity to the new generation of chemotherapy drugs for the treatment of cancer [
34]. Genomic and genetic studies provide valuable information about genomic changes in tumor samples compared to normal tissue samples. These studies provide important tools for understanding key information about tumor initiation, progression, and metastasis [
35]. In the present study, evaluating the transcriptional profile in gastric tumor samples is a strong strategy for identifying new molecular targets.
In searching for drug targets and discovering new drug candidates, computer-aided drug design (CADD) methodologies stand out as a powerful and promising technology for faster, cheaper and more effective drug design in drug research [
36]. Furthermore, they provide an absolute starting point for drug target discovery. Faced with the high cost and time required for research and development of drugs in the oncology area, computational models represent an efficient alternative since they are capable of predicting physical-chemical, pharmacokinetic, pharmacodynamic and toxicological parameters from a given molecular structure, as well as optimizing the in vitro test step [
36].
The results obtained so far in obtaining new drugs through in silico studies are notorious. For example, there is the development of the drug captopril, the first angiotensin-converting enzyme (ACE) inhibitor and one of the first successful drugs using computational tools to optimize drug planning in the 1980s [
37]. Following this study, structure-based drug development exhibited a significant impact on drug design with an increasing number of applications and a rapid growth of computational tools for drug discovery, including anticancer therapies, was observed.
Structure-based virtual screening (SBVS) is a robust technique that allows rapid identification of biologically active compounds, providing an efficient and cost-effective alternative to high-throughput experimental screenings. This technique allows the prediction of the best mode of interaction between two molecules to form a stable complex. It uses scoring functions to estimate the strength of non-covalent interaction between a ligand and molecular target. Different successful examples of SBVS application are reported in the literature, evidencing its versatility, high performance and great utility in drug discovery programs. In this study, the SBVS technique enabled the identification of synthetic molecules with great potential to inhibit target proteins relevant to carcinogenesis and the establishment of gastric cancer. The ligands with the best classification, considering the minimum binding energy to the target, had their ADMET properties evaluated and underwent a molecular docking validation protocol.
Analyzing ADMET properties is an important step in drug design development. It refers to the processes of absorption (A), distribution (D), metabolism (M), excretion (E) and toxicity (T) still in the early stages of the drug discovery process. This step drastically reduces the fraction of failure related to pharmacokinetics in the clinical phases and the toxic effects associated with drugs [
28].
Faced with the need to develop new therapeutic options to improve the effectiveness of gastric cancer treatment and patient survival statistics, targeted therapies stand out as instruments with great potential for therapeutic success. In this context, the present study points to the AJUBA, FBXL13, CCDC69, CD80 and NOLC1 proteins as potential candidates for targeted therapy in the treatment of GC.
The AJUBA protein has been implicated in the development of several human cancers. It is known that this protein participates in the assembly of countless protein complexes and is involved in several cellular biological processes, such as the repression of gene transcription, cell-cell adhesion, mitosis, differentiation, proliferation and cell migration [
38]. Previous studies have shown that the AJUBA protein promotes colorectal cancer cell growth by suppressing the JAK1/STAT1/IFIT2 network and activating N-cadherin expression through interaction with Twist in colorectal cancer cells [
39,
40]. However, its expression pattern and biological significance in gastric cancer are still not fully elucidated.
In the present study, analysis of the micro-array metadata revealed that AJUBA gene expression was higher in gastric cancer samples than in normal tissues. By comparing the relative expression level by qRT-PCR of this gene in samples of gastric tumor cell lines (ACP-02, ACP-03, AGP-01 and AGS) and normal gastric cells (MNP-01), the results confirmed a significant increase in AJUBA gene expression level in AGP-01 and AGS cell lines. Data from the survival analysis obtained using the Kaplan-Meier plotter database revealed that the increase in AJUBA expression is closely associated with a reduction in the overall survival rates of patients affected by GC.
Dommann et al. [
41] developed a study that analyzed the transcriptome of SW480 human colon cancer cell lines by RNA sequencing and confirmed the sequencing data with biological assays. In this analysis, it was possible to conclude that cells devoid of
AJUBA were less proliferative, more sensitive to irradiation, migrated less and were less efficient in forming colonies. Furthermore, loss of
AJUBA expression decreased tumor burden in a murine model of colorectal metastasis to the liver [
41].
From these data, it is assumed that the inhibition of the AJUBA protein in patients with gastric cancer can reproduce the phenomena observed in the work of Dommann et al. [
41]. Thus, compounds that inhibit this target may represent a new therapeutic alternative for GC.
The FBXL13 protein (F-box and leucine-rich repeat protein 13) is a member of the F-box family and substrate recognition component of the SCF-like ubiquitin ligase E3 complex (SKP1-CUL1-F-box), acting as a protein-ubiquitin ligase.
A study by Fung et al. [
42] characterized FBXL13 as a microtubule activity regulator and highlighted its role in promoting cell motility with potential tumor-promoting implications. Among the results shown, it was verified that FBXL13 interacts with the centrosome proteins Centrin-3, Centrin-152 and CEP192, the latter being a key factor in the initiation process of centrosome duplication and the control of centrosome microtubule nucleation [
42]. Data from this study indicate that the ubiquitylation of CEP192 by FBXL13 is relevant to regulate the formation of centrosome microtubule arrays and the migration of cancer cells [
42]. Being observed that the lack of centrosome microtubule arrays induced by
FBXL13 overexpression facilitates cell migration. Meanwhile,
FBXL13 depletion induces the accumulation of CEP192, promoting the formation of microtubule arrays in centrosomes, which, in turn, hinders cell migration [
42]. Given this, it is understood that the increased expression of the
FBXL13 gene may be decisive for the aberrant centrosome organization of cancer cells by promoting changes in the nucleation capacity of microtubules and, consequently, allowing tumor cells to present an increased capacity for proliferation and invasion.
Expression data from the Cancer Cell Line Encyclopedia reveal amplification of
FBXL13 in many cancer cell lines, many of which originate from breast cancers, glioblastomas, and lung cancer [
43]. Furthermore, an online repository of cancer patient cohorts showed that
FBXL13 is frequently amplified in solid tumors such as non-epithelial prostate cancer (20%), breast cancer (20%), esophageal cancer (10%) and head and neck cancer (10%) (cBioPortal) [
43].
In a CRISPR/Cas9 screening performed by Hart et al. [
44], it was seen that the
FBXL13 knockout reduced proliferation in a patient-derived glioblastoma cell line, with centrosome microtubule dysregulation being implicated as the cause of oncogenic aberrations [
45]. In microtubule-dependent processes, it has been shown that dysregulation of centrosome microtubules causes oncogenic aberrations. Among the signs of malignant transformation, cell-cell adhesion loss and cell polarization were observed [
46].
In an assay carried out in the present study by detection by real-time PCR (qRT-PCR), the expression of the FBXL13 gene in AGP-01 cells (intestinal-type metastatic tumor lineage) was notably higher compared to normal gastric cells (MNP-01). The described results reinforce the need to search for compounds capable of interacting with the protein encoded by the FBXL13 gene as a therapeutic alternative, as well as to understand more deeply the function of the F-box protein family in tumors, which may provide new insights into the role of centrosomes in cancer motility, invasion, and metastasis.
The CCDC69 protein, a coil domain-containing protein (CCDC) family member, regulates cell cycle progression and mediates apoptosis following DNA damage in eukaryotic cells. Recent studies have shown that CCDC proteins are closely related to various tumors, such as lung cancer and pancreatic ductal adenocarcinoma. Results of studies developed by Pal et al. [
47] indicate that CCDC69 acts as a framework to regulate the recruitment of midzone components and the assembly of central spindles, with the formation of the central spindle being essential for cytoplasmic division (cytokinesis) in animal cells [
47]. RNA interference (RNAi)-mediated knockdown of CCDC69 protein led to the formation of aberrant central spindles and disrupted the localization of midzone components such as aurora B kinase, cytokinesis protein regulator 1 (PRC1), MgcRacGAP/HsCYK -4 and polo-like kinase 1 (Plk1) in the central spindle [
47].
Until now, expression characteristics and mechanisms of the CCDC69 gene in patients with gastric cancer have not been well described in the literature. The results of our study demonstrated, in the analysis of the micro-array metadata, that gene expression was lower in samples of gastric cancer tumors than in normal tissues. Real-time PCR detection revealed a significant increase in the expression level of the CCDC69 gene in ACP-02 and ACP-03 cells. Survival analysis data obtained using the Kaplan-Meier plotter database revealed that increased expression of CCDC69 is associated with reduced overall survival rates of patients affected by GC.
Similar results were observed in a survey by Yi et al. [
48], in which CCDC69 was identified as a potential biomarker to predict the prognosis of breast cancer (BC). This study evaluated the relationship between
CCDC69 expression levels in breast cancer (BC) samples and their tumor clinical characteristics. For this purpose, RNA-seq information on CB samples from the TCGA database was used. The results showed that
CCDC69 expression was significantly lower in cancer samples than in normal tissues. Furthermore, low expression of
CCDC69 was associated with poor overall survival based on the Affymetrix microarray in the Kaplan-Meier plotter database [
48].
CD80 plays an important role in T cell activation, exerting a dual effect on tumor immunity: it binds to CD28 to provide a costimulatory signal for T cell activation, and it binds to CTLA-4, resulting in an immunosuppressive effect. It is understood that the binding of CD80 with CTLA-4, a receptor that acts as an important negative regulator of T-cell responses, is favorable to carcinogenesis. Because, commonly, cancer cells use the immunosuppressive function of regulatory T cells to avoid immunological attacks [
49,
50,
51].
Based on this understanding, a monoclonal antibody against CTLA-4 (ipilimumab) was approved by the FDA (Food and Drug Administration) for the treatment of melanoma [
52]. Ipilimumab blocks the co-inhibitory signal induced by CTLA-4 binding with CD80 to enable CTL-mediated antitumor immunity [
51].
Considering the positive results of blockade of the co-inhibitory signal induced by the binding of CTLA-4 with CD80 for patients with melanoma, it is believed that it is important to obtain a greater understanding of expression data and the function of CD80 in gastric cancer, as well as to characterize the existing molecular interaction between CTLA-4 and CD80 proteins.
The analysis of the protein interactions network developed in the present study using the STRING v11.5 tool made it possible to evaluate the protein-protein interactions network. Among the results of our study, it was revealed through a molecular docking assay that the molecular binding between the CTLA-4 and CD80 proteins is altered after the anchoring of the MCULE-9178344200-0-1 compound to the CD80 protein (
Figure 6). Alteration of the molecular binding between the two proteins may result in the functional change of this interaction and, consequently, promote the blockade of the co-inhibitory signal induced by the binding of CTLA-4 with CD80.
Concerning CD80 expression and function data in gastric cancer, the results of our study revealed an increase in the expression level of the CD80 gene in samples of gastric cancer tumors to normal tissues in the micro-array metadata analysis, as well as in real-time PCR detection, which revealed a significant increase in the expression level of the CD80 gene in the ACP-03 cell line. Survival analysis data obtained using the Kaplan-Meier plotter database revealed that increased CD80 expression is associated with reduced overall survival rates of patients affected by GC. These results show the protein and the CD80 gene as potential successful therapeutic targets for the control of GC.
On the other hand, in a study by Feng et al. [
51], the determination of
CD80 mRNA levels in gastric adenocarcinoma tissues and adjacent normal tissues by RT-qPCR was performed. As a result, it was seen that
CD80 is down-regulated in gastric cancer tissues in 15 out of 20 patients compared to normal gastric tissue. Specifically, 70% of gastric tumor tissues demonstrated reduced
CD80 expression [
51]. Thus, it is important to consider that the
CD80 gene is differentially expressed in gastric adenocarcinoma cell lines.
The NOLC1 protein (nucleolar and coiled-body phosphoprotein 1) is responsible for various cellular life activities, including ribosome biosynthesis, DNA replication, transcription regulation, RNA processing, cell cycle regulation, apoptosis, and cell regeneration [
53]. Our results demonstrated that gene expression was higher in samples of gastric cancer tumors than in normal tissues (
Figure 2), which was confirmed by RT-qPCR for the GC cell lines ACP-03 and AGS (
Figure 4). Data obtained using the Kaplan-Meier plotter database revealed that patients affected by GC with overexpression of
NOLC1 have shorter overall survival than those with low expression of
NOLC1 (
Figure 3). Additionally, the gene enrichment analysis suggests the participation of the
NOLC1 gene as an important regulator for the development and progression of gastric tumors.
In a study carried out by Kong et al. [
54], the role of
NOLC1 in esophageal cancer (ESCA) was determined, as well as its gene expression in ESCA tissues and cell lines evaluated by qRT-PCR, immunohistochemistry or western blot. Among the results, overexpression of
NOLC1 was observed in ESCA tissues and ESCA cell lines (EC9706, Eca109, TE-13, Kyse170, T.TN) compared to adjacent normal tissues and normal esophageal cell lines.
NOLC1 overexpression was markedly associated with larger tumor size, lymph node metastases, and advanced TNM stage.
The results of the correlation between
NOLC1 gene expression and overall survival by Kaplan Meier plotter for patients with esophageal cancer coincided with the data obtained for patients with gastric cancer, evaluated in our study. So
NOLC1 overexpression was also associated with reduced overall survival rates for patients with ESCA.
NOLC1 knockdown, in turn, inhibited proliferation, migration, invasion and cyclin B1 expression and promoted apoptosis and cleaved-caspase-3 expression of two ESCA cell lines [
54]. The in-depth study of data on
NOLC1 gene expression and its role in carcinogenesis shows that the protein encoded by this gene is a promising therapeutic target.
Cell life is a result of the interaction and coordinated activity of many proteins working at the same time. Different proteins from different pathways work together to provide a meaningful biological process essential to cell development [
54,
55,
56]. In cancer, protein-protein interaction is essential to form complexes allowing uncontrolled cellular division, development, growth, and tumor promotion. Recently, cancer, neurodegenerative diseases and even infections have been attributed as a result of aberrant protein-protein interactions [
56]. In cancer, aberrant proteins interact with other proteins, establishing the initial stages of cancer [
55,
56]. Based on that, protein-protein interaction analysis has become an alternative target for developing new anticancer compounds [
55]. Based on that, a protein-protein interaction analysis of the targets used in this study was performed (
Figure 6). The protein-protein interaction analysis revealed that all targets involved in this work are pivotal in their pathways.
For example, the AJUBA proteins revealed an interaction with the proteins CTNNB1 and β-catenin (
Figure 6A). The CTNNB1 regulates cellular adhesion and gene transcription during the mitotic fuse establishment [
57]. The β-catenin is a component of the centrosome during the interphase [
57]. Bahmanyar et al. [
57] suggested that aberrant interactions between AJUBA protein and CTNNB1 and β-catenin led to uncontrolled cellular division and, thus, cancer development. Our results revealed that the molecule MCULE-2386589557-0-6 interacts with the AJUBA leading to a misplaced interaction between AJUBA and CTNNB1, interfering in this pathway and probably preventing their role in cancer establishment.