Next Article in Journal
Oral Exposure to Titanium Dioxide E171 and Zinc Oxide Nanoparticles Induces Multi-Organ Damage in Rats: Role of Ceramide
Previous Article in Journal
Virophages, Satellite Viruses, Virophage Replication and Its Effects and Virophage Defence Mechanisms for Giant Virus Hosts and Giant Virus Defence Systems against Virophages
Previous Article in Special Issue
Non-Coding RNA as Biomarkers and Their Role in the Pathogenesis of Gastric Cancer—A Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bioinformatics Analysis and Validation of Potential Markers Associated with Prediction and Prognosis of Gastric Cancer

by
Tasuku Matsuoka
1,2 and
Masakazu Yashiro
1,2,*
1
Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan
2
Institute of Medical Genetics, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(11), 5880; https://doi.org/10.3390/ijms25115880
Submission received: 18 April 2024 / Revised: 23 May 2024 / Accepted: 25 May 2024 / Published: 28 May 2024

Abstract

:
Gastric cancer (GC) is one of the most common cancers worldwide. Most patients are diagnosed at the progressive stage of the disease, and current anticancer drug advancements are still lacking. Therefore, it is crucial to find relevant biomarkers with the accurate prediction of prognoses and good predictive accuracy to select appropriate patients with GC. Recent advances in molecular profiling technologies, including genomics, epigenomics, transcriptomics, proteomics, and metabolomics, have enabled the approach of GC biology at multiple levels of omics interaction networks. Systemic biological analyses, such as computational inference of “big data” and advanced bioinformatic approaches, are emerging to identify the key molecular biomarkers of GC, which would benefit targeted therapies. This review summarizes the current status of how bioinformatics analysis contributes to biomarker discovery for prognosis and prediction of therapeutic efficacy in GC based on a search of the medical literature. We highlight emerging individual multi-omics datasets, such as genomics, epigenomics, transcriptomics, proteomics, and metabolomics, for validating putative markers. Finally, we discuss the current challenges and future perspectives to integrate multi-omics analysis for improving biomarker implementation. The practical integration of bioinformatics analysis and multi-omics datasets under complementary computational analysis is having a great impact on the search for predictive and prognostic biomarkers and may lead to an important revolution in treatment.

1. Introduction

Gastric cancer (GC) is a common cancer type, with more than one million new cases each year [1]. Although recent developments in endoscopy, imaging, surgical procedures, and the use of anti-cancer drugs have somewhat prolonged the overall outcome of GC patients, the 5-year survival of patients with advanced GC remains <30%, mainly due to the lack of efficient therapies [2]. So far, comprehensive molecular analysis has identified many molecular alterations and dysregulated pathways in GC. Mutations in the TP53 and CDH1/RHOA genes are well-known [3]. Concerning copy number alterations, the amplification of genes involved in tyrosine kinase receptor pathways, such as FGFR2, HER2, EGFR, MET, and WNT, has been noted, as well. Other important mutations in GC involve the ARID1A, PIK3CA, CCNE1, CLDN18-ARHGAP26, and BRCA2 genes [4]. Despite these advances in knowledge only one oncogenic driver-based therapy (Her2-directed treatment with trastuzumab) has shown acceptable survival benefit [5]. Other therapeutic options, such as cytotoxic agents or anti-angiogenic therapy, are only transiently useful except checkpoint inhibitors for patients with microsatellite unstable GCs, which are infrequent at the advanced stage. Therefore, it is crucial to find relevant biomarkers with good predictive accuracy to select those patients with GC who are likely to benefit from targeted therapies.
The development of innovative computational approaches for integrating data from various cohorts would be useful for resolving these problems. Rapid technological revolutions in genome-wide sequencing, such as next-generation sequencing (NGS), have recently provided researchers with enormous expression datasets [6]. The advancement of omics data due to breakthroughs in high-throughput technologies has generated the concept of “big data” in cancer, the analysis of which necessitates large computational resources and bioinformatic algorithms [7]. With the interpretation of bioinformatics technology, a comprehensive understanding of genetic, epigenetic, proteomic, transcriptomic, and metabolic processes has been developed as a framework with which researchers can decipher the various mechanisms responsible for the heterogeneity of GC [8]. “Omics” is a series of approaches that aim to characterize and quantify the pools of biomolecules within organisms related to structures, functions, and dynamics. However, individual profiling using a single-omics approach only offers a limited landscape of the complex molecular regulatory axes in malignancies. Thus, integration analysis of multi-omics (multi-layer) data is a crucial step toward a greater understanding of the signatures of different mechanisms of carcinogenesis and, indeed, the mechanisms themselves. The integration of bioinformatic analysis with multi-omics platforms is emerging as a promising tool for elucidating accurate prognosis, prediction of effective therapies, and biomarkers to increase the response rate to targeted treatments in patients with GC [9] (Figure 1).
In this review, we summarize an overview of how bioinformatic analysis contributes to biomarker discovery for prognosis and prediction of therapeutic efficacy in GC. Emerging single-omics datasets, such as genomics, epigenomics, transcriptomics, proteomics, and metabolomics for validating putative markers, are systematically reviewed. Finally, we discuss the current challenges and future perspectives to integrate multi-omics analysis to improve biomarker implementation at the clinical level.

2. Genomics-Based Validation of Predictive and Prognostic Markers of GC

Recently, high-throughput technology combined with bioinformatics algorithms has been broadly used in the detection of genetic alteration derived in oncogenesis. To diagnose differentially expressed genes (DEGs) associated with GC, databases such as The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) are frequently used [10]. Meanwhile, the former conventional studies have only focused on the distinctive DEGs and disregarded the network complex with a high degree of interaction between the DEGs. Protein–protein interaction (PPI) networks and weighted gene co-expression network analysis (WGCNA) based on the RNA sequencing and microarray data have been revealed to establish potent organized biological strategies for mining the functional gene modules and detecting novel prognostic biomarkers [11,12]. These tools enable a better understanding of the mechanisms underlying the progression of GC and are utilized to identify novel targets for the prediction of effective therapy and prognosis.
While numerous single-gene biomarkers have been described, multi-gene signatures reveal more usefulness and significance for the prognosis of patients with cancer. Current topics of GC predictive and prognostic biomarkers based on a variety of genetic features based on bioinformatics analysis were summarized in Table 1. A recent study integrating data in multiple public databases detected a six-metastasis-related gene (PCOLCE2, TMEM132C, UPK1B, SLITRK2, PM20D1, and FLJ35024) set that predicts prognosis in patients with GC through TGCA database analysis. They also evaluated the expression of the six distant-metastasis gens in the GC tissue sample, and the reliability of the prognostic value was shown [13]. Based on 375 TCGA GC samples, 50 DEGs between GC and paired normal tissues were identified [14]. The association of gene expression with immune cells was examined based on the TIMER database. Ten of the fifty genes (RXRG, AGT, BCHE, UBE2QL1, PLCXD3, ADCYAP1R1, NRCAM, MAMDC2, CDH19, and GAMT) were linked with the outcome and macrophage infiltration in GC. AGT was correlated to GC carcinogenesis and cooperated with BRD9, GOLPH3, NOM1, KLHL25, and PSMD11 [14]. Interestingly, systematic analysis summarized 39 prognostic gene signatures for GC patients to detect the most overlapped gene. Three genes (COL1A1, COL1A2, EGFR) were identified three times, suggesting that they have superior potential in GC prognosis and may provide a theoretic foundation for improving treatment strategies [15]. Another study has identified DEGs by using WGCNA and detected five hub genes (CEMIP, CST1, COL8A1, PMEPA1, and MSLN) in GC and has constructed a prognostic signature with a good predictive performance by integrating multiple GEO datasets and WGCNA algorithms. Among them, CEMIP repressed the growth and migration and impaired the chemoresistance of GC cells to 5-FU, suggesting that CEMIP may be applied to precision medicine for GC patients [16]. Similarly, an integrated bioinformatics analysis was performed to identify the common hub genes derived from the PPI and WGCNA network based on the common DEGs from the TCGA-STAD and three GEO datasets (GSE65801, GSE54129, and GSE118916). Among the six common hub genes, TRYOBP and C1QB were negatively associated with the overall survival (OS) of the GC patients. Both genes were also correlated with the CD8+ T cells, CD4+ T cells, and macrophages using an immune analysis based on the TCG-STAD [17]. A recent study using a prognostic risk model by multivariate Cox regression analysis in GEO datasets GSE15459 investigated potential prognostic genes [18]. A total of eight genes significantly related to survival were identified, and decreased survival was significantly associated with AGT, SERPINH1, and MMP7, indicating that the abnormal regulation of these genes in cancer may impact the OS of GC. Moreover, the exosome level of AGT and MMP7 were associated with the serum level, and these proteins stimulate the migration of GC cell lines, suggesting that exosomal AGT and MMP7 may be useful biomarkers for GC prognosis and included in GC progression. Notably, a study using DAVID analysis detected clusters of nine genes (CTHRC1, BGN, FAP, THBS2, COL12A1, COL5A2, SULF1, SPP1, and COL10A1) which were associated with alterations in the extracellular matrix (ECM), cell adhesion, and collagen catabolic process. The high expression of BGN, COL5A1, COL10A1, COL12A1, CTHRC1, SULF, and THBS2 are negatively correlated with OS and may serve as useful prognostic biomarkers for GC. Among them, a superior hazard ratio (HR) for poor OS was shown in BGN, THBS2, and CTHRC1 compared with other hub genes [19].
Overall, genetic correlations with GC based on integrated bioinformatic analysis have been identified. The main purpose is to progress our knowledge about GC-associated biological pathways, predicting GC prognosis and treatment outcomes. Moreover, further understanding of the genetic loci associated with GC can allow us to identify more precise predictive/prognostic biomarkers.

3. Epigenomics-Based Validation of Predictive and Prognostic Markers of GC

Epigenetic regulations are non-heritable changes induced by covalent tags or other modifications of the genome [20]. Similar to genomics, an increasing number of evidence suggests that epigenetic modifications, including DNA methylation of CpG islands, post-translational modifications of histones, and nucleosome positioning, are also comprised in the GC initiation, progression, and chemo-resistance [21]. DNA methylation alterations are essential to all phases of cancer genomics, and hypomethylation has been revealed to be pivotal in cancer progression [22]. Recently, DNA methylation has been revealed to play a significant predictive role in the prognosis of patients with GC.
The nomogram has been broadly utilized as a predictive method in oncology for a decade [23]. It requires an integrated model and is useful for clinicians to exploit in prognosis prediction. A previous study using a risk score and OS nomogram for predicting the prognosis of GC patients identified six DNA methylation-driven DEGs (PODN, MYO1A, NPY, MICU3, TUBB6, and RHOJ) as prognostic genes using a least absolute shrinkage and selection operator (LASSO) logistic regression model [24]. They verified that hypermethylated five genes (PODN, NPY, MICU3, TUBB6, and RHOJ) may have a protective role. In contrast, MYO1A was hypomethylated and may be a typical tumor suppressor gene. These six genes may contribute to a cost-effective and accurate prediction of the outcome of GC in clinical use. Another recent study integrated publicly available GC DNA methylation datasets obtained from TCGA and GEO databases to construct a prognostic model by matching differential methylation sites (DMSs) with OS [25]. The prognostic model identified an 11-DMS (CEP290, CCDC69, UBXN8, KDM4A, AKR1B1, RASSF2, KDELR3, CHRNB2, EGR1, ARMC9, and RPN1) signature. Among these DMGs, five DNA methylation driver genes (KDM4A, AKR1B1, RASSF2, CHRNB2, and EGR1) are found to be closely correlated to the progression of GC. Afterward, the prognostic risk score was constructed, and values were verified. Patients in the high-risk group revealed a worse prognosis, which was positively correlated with the VIM gene and negatively correlated with the CDH1 gene. Of note, CHRNB2 expression was significantly suppressed only in the TP53 mutation group of GC patients [25]. Also, a study identifying methylated-differentially expressed genes (MDEGs) on a genome-wide scale systematically explored Multi-Omics cohorts from the TGCA and GEO to construct a new MDEGs-based signature [26]. The eight-MDEGs signature (TREM2, MICAL2, INHBA, PCSK5, NRP1, YAP1, RAI14, and MATN3) was identified and validated the prognosis performance. The multivariate Cox regression analysis presented that the eight-MDEGs signature risk score showed statistical significance as an independent variable in the TCGA-GC and GSE 84437. These MDEGs significantly divide GC patients into high- and low-risk groups in OS.
The DNA methylation patterns may have clinical implications as prognostic markers of GC patients, which provide information useful for detecting therapeutic targets. DNA methylation investigated as potential prognostic GC markers are listed in Table 2.

4. Transcriptmics-Based Validation of Predictive and Prognostic Markers of GC

A transcriptomic investigation analyzes an organism’s whole RNA content. Transcription is the method of copying a segment of DNA into RNA and reveals an outline of the activity of the cells [27]. Recently, transcriptomics has remarkably progressed in molecular genetics [28]. Transcriptomes denote all RNA molecules created in a cell from the genome under certain pathological situations [29,30]. Recently, the expression profile of G-protein-coupled receptor 27 (GPR27) in GC was investigated based on TCGA and GEO database [31]. GPR27 mRNA levels were lower in GC tissues than in normal gastric specimens. Missense mutation of GPR27 was found in 4% of GC patients through cBioPortal, and GPR27 mutation was associated with poor prognosis and GPR27 cg03024619 showed the most prognostic significance. Interestingly, GPR27 had an apparent interaction with the activation of T cells and tumor mutation burden.
Although bulk RNA-seq analyzes only the mean transcript expression in a cell population, single-cell RNA sequencing (scRNA-seq) evaluates the transcriptomic condition of specific populations of single cells [32]. A study using an immune-targeted single-cell profiling strategy examined the role of tumor immune microenvironment (TIME) in the progression of gastric signet ring cell carcinoma (GSRCC) [33]. TIME heterogeneity between GSRCC and non-GSRCC was compared by scRNA-seq. They discovered that the GSRCC TIME may be dormant, where Treg-FOXP3 and CD8-Tex were hard to mobilize and impaired B cells’ accurate activities. They also indicated the potential role of CXCL13 in forming the TIME within GSRCC, suggesting that the diminution of CXCL13-mediated regulatory effect in CD8-Tex would be associated with the poor prognosis of GSRCC which is validated by the cytometry by time of flight (CyTOF) results. In another study, RNA-seq-based transcriptomics and TMT liquid chromatography-mass spectrometry (LC-MS)/MS-based proteomics study were conducted in which 14 differentiating genes were identified. These 14 genes were significantly enriched in metabolic pathways, suggesting that metabolic genes have a pivotal role in GC [34]. They also found that the gene set was negatively associated with Treg, monocytes, Th1, DC, and macrophages and positively correlated to NKT, CD4+, and MAIT cells. Besides, some metabolic genes such as ADH1B, PHGDH, BCAT2, NE3, PCCB, and CS were positively associated with PD-1.
Non-coding RNAs (ncRNAs), which comprise more than 90% of the RNA transcripts of the human genome, are deficient in protein-coding activity. However, accumulating evidence has found that ncRNAs contribute to the initiation and progression of GC and can act as important biomarkers in prognosis [35,36]. Hence, detecting differentially expressed ncRNAs may be associated with the prediction of survival in GC. A recent study using the gene expression profiles of the GEO database of GC identified 110 consistent differentially expressed miRNA (DEMs) between GC and paired normal tissues, which were processed with the limma package in R [37]. Subsequently, to validate the relationship between DEMs and the prognosis of GC patients. 361 specimens from TGCA were collected. Three DEMs (miR-145-3p, miR-125b-5p, and miR-99a-5p) were associated with poorer prognosis by using a log-rank test and Kaplan-Meier curve. The multivariate analysis showed that the T stage and the three-DEM signature were all independent factors in predicting the prognosis of patients with GC [37]. Another study examining intestinal GC molecular networks’ gene-microRNA relationship has shown that three regulatory circuits are identified in a complex interaction network [38]. Besides, enrichment analysis revealed that KRAS, PI3K, and p53 were associated with these regulatory circuit networks. Noteworthy, some of the hub components operated together and contributed to prognosis. The upregulation of hsa-mir-200b and the downregulation of its target CFL2 were closely correlated with worse survival.
Growing evidence has shown that long non-coding RNAs (lncRNAs), with a length of more than 200 nucleotides, can manage cancer development and the alterations of variable genes [39]. Aberrant expression of several lncRNAs is involved in the pathogenesis, suggesting that novel lncRNAs may serve as the new clinical biomarkers. A study using a variety of machine learning algorithms and constructed immune-related lncRNA prognostic model (ILPM) detected 18 prognostic immune-related lncRNAs (SNHG5, LINC01270, CHKB. AS1, NUTM2A.AS1, MIR181A2HG, CCNT2.AS1, DLG3.AS1, LINC01134, NIFK.AS1, RP11.443B7.1, LSAMP.AS1, HMGN3.AS1, LPP.AS2, RP11.710C12.1, RP11.155O18.6, CASC15, RP11.449P15.2, and FLJ16779) [40]. All patients were classified into high- and low-risk groups depending on the median risk score. Patients belonging to the high-risk group showed poor outcomes, lower immune cell infiltration, and a higher metabolism. Notably, 86 DEGs were mainly enriched in immune-related pathways between high- and low-risk groups, and 18 drugs in the Genomics of Drug Sensitivity in Cancer were sensitive to the high-risk patients. LncRNAs also have been shown to promote the evasion of immune surveillance by cancer through various mechanisms. The competitive endogenous RNAs (ceRNA) hypothesis declares that as ceRNAs, some RNAs can mediate the expression of downstream mRNA by linking distributed miRNAs [41]. The integrative analysis of GEO and TCGA data reported that three lncRNAs (UCA1, HOTTIP, and HMGA1P4) may be involved in the development, and their prospective activities may be related to the prognosis of GC [42]. CeRNA network analysis found an interaction between hsa-miR-508-3p and HMGA1P4. Enrichment analysis showed that genes are enriched in the cell cycle and cellular senescence. Similarly, seven glycolysis-related lncRNAs (AL353804.1, AC010719.1, TNFRSF10A-AS1, AC005586.1, AL355574.1, AC009948.1, and AL161785.1) were identified based on the TCGA-STAD databases using LASSO and Cox regression analyses. These lncRNAs were enriched in the cell adhesion molecules, leukocyte transendothelial migrations, calcium signaling pathway, and JAK-STAT signaling pathway [43]. Studies using transcriptome in the prediction/prognosis of GC in the past 5 years are collected in Table 3.

5. Proteomics-Based Validation of Predictive and Prognostic Markers of GC

Proteins are vigorous regulatory elements of cells that sustain cellular processes. The normal activities of proteins may be disordered due to stress, which results in abnormal cell activity. Proteomics analysis can reflect the alterations in the final regulatory products of the cells. Recent technologies allow to prediction of protein expression change, which may mediate the cellular biological processes [46]. Proteomics study ordinarily examines the construction and functions of proteins, and activities during carcinogenesis. Proteomics biomarkers are generally protein components derived from the body’s system between normal and pathological conditions. The developments in knowledge and technologies of high-throughput mass spectroscopy (MS) provoke the identification of effective biomarkers for GC [47]. In this section, we aim to provide recent developments in finding biomarker candidates and the prospective application of MS-based biomarkers for detecting highly specific and sensitive prognostic biomarkers with their outlooks in clinical practice (Table 4).
A recent study established a prognostic risk model [48]. Three proteins (COLLAGEN VI, CD20, TIGAR) were identified and may be utilized as an independent factor that is closely associated with GC prognosis. Enrichment analyses showed that DNA metabolic and growth regulation pathways were enriched. Likewise, a study characterized by RPPA-based functional proteomic data identified the functional proteome signatures (MYH11, CD20, CHK1_pS345, AR, PR, HER3, MYH11, and SMAD1), suggesting that these candidate protein markers can serve as potential prognostic biomarkers in GC [49]. Another proteomic analysis attempted to characterize molecular changes in the extracellular matrix (ECM) surrounding GC. ECM components of tumor tissues revealed no difference from normal mucosa, whereas their levels were generally displayed as increased ECM proteins (LOX, LAMB3, ADAMTSL1, and FBN2) and decreased basement membrane components (COL4A5, COL4A6, COL28A1, NID1, and LAMA5) that were closely related to tumor angiogenesis, invasion, and metastasis, which were closely related to OS of GC patients [50]. Integrating proteomics with nano-liquid LC-MS identified a total of 429 differentially expressed proteins [51]. Enriched analysis revealed that several pathways were activated and correlated with chaperone function, apoptosis, and immune response. Generally involved pathways are the overexpression of heat shock protein (HSP) families, which are commonly expressed in response to stress, the antiapoptotic pathway, the BH3 interacting domain, and the BLC-protein family. Trefoil factor 1 and 2, chromogranin A, REGIA, and the mitogen factor gastrin 7 were downregulated. In addition, among the 20 hub proteins detected, IST1, VAT1, ALYREF, and HEXB were closely related to worse survival [51]. Interestingly, a recent study applied a deep learning-based proteomic subtyping workflow that inserted the autoencoder framework. Patients with a good prognosis showed effective responses to chemotherapy, whereas patients with a poor prognosis resisted. Enrichment analysis showed patients with good revealed outcomes of the pathways that were related to DNA replication, cell cycle, and programmed cell death. Meanwhile, patients with poor outcomes are enriched in ECM-related pathways [52].
Table 4. Proteomics-based predictive/prognostic GC biomarkers using bioinformatics analysis for the past 5 years.
Table 4. Proteomics-based predictive/prognostic GC biomarkers using bioinformatics analysis for the past 5 years.
Number of PatientsDatabasesAnalytical PlatformStatistical AnalysisProposed Proteomics-Based BiomarkersCommentsRefs.
18GEO
EGA TCGA
Nano-LC-MS
PLGS ExpressionE tool algorithm
MetaCoreTM
KEGG Mapper
PPI analyses
Kaplan–Meier MethodHSP, CHGA, TFF 1 and 2, REGIA, MAPK1IST1, VAT1, ALYREF, and HEXB were closely related with the worse survival.[51]
48TCGA
TIMER
GSCA
TMT-MSKaplan–Meier Plotter databaseBCAT2, ALDH6A1, MCEE, PCCB, BCKDHB, DBT, AUHThe expression of BCAT2 was suppressed in cancerous samples from GC patients and was negatively correlated with survival.[33]
352TCGA
TCPA
RPPA
dplyr, ggplot2, and ggrepel software packages, https://ggrepel.slowkow.com.
GO and KEGG analyses
Kaplan–Meier survival curve
Cox regression analysis
COLLAGEN VI, CD20, TIGARTri-protein was detected as an independent prognostic factor.
[48]
N.A.TCGA
TCPA
RPPA
MIFS algorithms
PCA, PLS-DA, t-SN, and heatmap analysis
Kaplan–Meier survival analysis
Cox regression analysis
MYH11, CD20, CHK1_pS345, AR, PR, HER3, MYH11, SMAD1 RPPA-based functional proteomic data identified the functional proteome signatures. [49]
367TCGA-STADNano LC-MS/MS
Proteome Discoverer 2.4.0.305 software
InterPro Protein Domains and Features KEGG pathways analysis
mRNA-Seq
Student’s t-test or One-way ANOVA
Kolmogorov–Smirnov test
Brown–Forsythe test
LOX, LTBP2, and COL1A2 provide disease progression and patient poor prognosis. ECM constituents that characterize the matrisome of GC contexts were identified.[50]
Abbreviations: GC, gastric cancer; TCGA, The Cancer Genome Atlas; GEO, Gene Expression Omnibus; EGA, European genome-phenome archive; MS, mass spectrometry; KEGG, The Kyoto Encyclopedia of Genes and Genomes; PPI, protein–protein interaction; HSP, heat shock protein; TFF, trefoil factor; MAPK, mitogen-activated protein kinase; CHGA, Chromogranin A; TMT-MS, tag-mass spectrometry; TCPA, The Cancer Proteome Atlas; RPPA, reverse-phase protein array; PCA, principal component analysis; PLS-DA, partial least squares discriminant analysis; N.A., not applicable.

6. Metabolomics-Based Validation of Predictive and Prognostic Markers of GC

Metabolomics is a comparatively novel field of study that measures all the metabolites within a biological sample (metabolome) and the purposes of quantifying metabolites in a metabolome [53]. Metabolomics can amplify small alterations in the genome, transcriptome, and proteome, which indicate the termination of gene activities and changes in individuals [54]. Thus, metabolomics is thought to be the last direction of omics research. Unlike normal cells, cancerous cells have been shown to present an extremely marked metabolic phenotype, which is depicted by amplified energy synthesis, elevated antioxidant regeneration, and enhanced macromolecule production. Commonly, cancer metabolism has a significant role in the tumorigenesis, progression, as well as drug resistance of cancer [55]. Compared with other “omics” involving genomics, transcriptomics, and proteomics, metabolomics could provide direct and general information on metabolites and result in response to both genetic alterations and pathological stimulation [56]. Over the past few years, with the advancement of metabolomics technology, previous studies have examined the application of metabolomics to identify novel biomarkers and uncover prediction biomarkers for prognosis in GC [57] (Table 5). Aberrant metabolites in metabolism pathways, such as carbohydrate, lipid, amino acid, and nucleotide metabolisms, may be useful for developing potential biomarkers for GC prognosis and prediction for GC therapy. In addition, metabolomic analysis is cost-effective compared with a genome, a transcriptome, or a proteome analysis.
A study screening GC survival-associated metabolites identified 24 metabolites by using Kaplan–Meier survival analysis [58]. Eight metabolites (ornithine, phenyl acetyl-L-glutamine, porphobilinogen, linoleic acid, DL-dipalmitoyl phosphatidylcholine, 5′-mdthylthioadenosine, inosine triphosphate, and paraxanthine) were indicated as an independent prognostic factor of GC by the forest plot of the Cox regression analysis. Enrichment analysis found that purine metabolism and linoleic acid metabolism were found to be the most significant metabolic pathways related to the survival of patients with GC. Another metabolomics study proposed to detect differential levels of metabolites in the pleural effusion of GC patients. They divided the patients into two groups depending on the existence of peritoneal metastasis, serosal invasion, and CEA mRNA [59]. Seventeen differential metabolites (sulfite, TG (54:2), TG (53:4), G3P, α-aminobutyric acid, α-CEHC, dodecanol, glutamyl alanine, 3-methyl alanine, 3-hydroxysteroid, CL (63:4), PE-NMe (40:5), retinol, MG (21:0/0:0/0:0), tetradecanoic acid, tridecanoic acid, octacosanoic acid and myristate glycine) were found with a weight of 100% using SVM. These metabolites had potential predictive ability and independent risk factors for peritoneal metastasis of GC patients.
Lipid metabolism deregulation is one of the most familiar metabolic alterations in cancer [60]. Interestingly, a study examining differential serum metabolites showed significant alterations in the abundance of several metabolites among patients before and after surgery [61]. Particularly, lysophosphatidic acids, triglycerides, lysine, and sphingosine-1-phosphate revealed increased levels in the postoperative groups compared with the preoperative and recurrence groups. Meanwhile, phosphatidylcholine, oxidized ceramide, and phosphatidylglycerol displayed decreased levels. These results may suggest that these differential metabolites may be useful biomarkers for validating the prognosis and monitoring the recurrence of GC. A more recent study that employed LC–MS-based targeted metabolomics to illustrate the global metabolomic profiling of large cohorts analyzed plasma samples from GC patients utilizing machine learning [62]. Machine learning-derived 28-metabolite prognostic model showed successful predictive activity with an AUROC of 0.832 and a concordance index of 0.83. Among them, 11 of the 28 metabolites (symmetric dimethylarginine/asymmetric dimethylarginine, neopterin, thymine, glucuronate, hydroxyproline, 14:0 Carnitine, indole acrylate, 8:0 Carnitine, acetylalanine, 2-aminoadipate, and GlcNAc6p) significantly discriminated the prognosis of GC patients. Patients who are distinguished by the 28-PM model from the high-risk group are more likely to reveal worse survival compared with the low-risk group and provide an advantage from intensive monitoring, intervention, and clinical trial. A study by Song and colleagues attempted to detect metabolic features for gastrointestinal cancer prognostic assessment [63]. MS-based platforms analyzed plasma samples, and 133 metabolites were significantly altered in the GC samples. Pathway enrichment analysis displayed that they are principally involved in steroid hormone biosynthesis, cysteine and methionine metabolism, arginine and proline metabolism, galactose metabolism, amino acids, and purine metabolism pathways. Ubiquinone and other terpenoid–quinone biosyntheses, phenylalanine, tyrosine, and tryptophan biosynthesis, histidine metabolism, starch and sucrose metabolism, fructose and mannose metabolism, and inositol phosphate metabolism were associated with GC patients. Interestingly, metabolic patterns changed before- and after surgical procedures in GC. The levels of PE 38:4, threonic acid, l-lysyl-l-glutamine, and temorine were relatively high after GC surgery.
Genome-wide association studies (GWAS) can identify hundreds of quantitative trait loci vigorously related to plasma/serum levels of metabolites. Studies integrating metabolomics and genetics analyses, known as metabolite GWAS (mGWAS), enable the examination of correlations between a variety of gene alterations and metabolites and a valuable understanding of the genetic control of metabolite levels [64]. A recent study investigated the genetic variation mechanism of GC prognosis-related metabolites using mGWAS. It detected 32 genetic variant loci that were potentially relevant to GC survival-related metabolites, corresponding to 7 genes, including VENTX, PCDH7, JAKMIP1, MIR202HG, MIR378D1, LINC02472, and LINC02310 [65]. In addition, gene oncology (GO) enrichment analysis found that these genes are principally associated with cell signaling molecular transmission, organism growth and development, nervous system regulation, and immune escape-related pathways, suggesting that these functional genes regulate the metabolites via related pathways and thereby affect GC survival.
Table 5. Metabolomics-based predictive/prognostic GC biomarkers using bioinformatics analysis for the past 5 years.
Table 5. Metabolomics-based predictive/prognostic GC biomarkers using bioinformatics analysis for the past 5 years.
Number of PatientsAnalytical PlatformStatistical AnalysisProposed Proteomics-Based BiomarkersCommentsRefs.
218Agilent MassHunter qualitative analysis software (version B.01.00, Agilent Technologies, Santa Clara, CA, USA)
MetaboAnalyst
Kaplan–Meier analysis
log-rank tests
Multivariate Cox regression analyses
Lasso regression
omithine, phenylacetyl-L-glutamine, porphobilinogen, linoleic acid, DL-dipalmitoyl phosphatidylcholine, 5′-methylthioadenosine, inosine triphosphate, paraxanthineThese metabolites were indicated as an independent prognostic factor.[58]
65LC-MS
Total ion current spectra
PCA
t-test, Support vector machine sulfite, TG (54:2), TG (53:4), G3P, α-aminobutyric acid, α-CEHC, dodecanol, glutamyl alanine, 3-methyl alanine, 3-hydroxysteroid, CL (63:4), PE-NMe (40:5), retinol, MG (21:0/0:0/0:0), tetradecanoic acid, tridecanoic acid, octacosanoic acid and myristate glycineThese metabolites had independent risk factors for peritoneal metastasis of GC patients.[59]
31LC-MSt-testlysophosphatidic acids, triglycerides, lysine, and sphingosine-1-phosphate, phosphatidylcholine, oxidized ceramide, phosphatidylglycerolThese differential metabolites may be useful biomarkers for validating the prognosis and monitoring the recurrence of GC. [61]
181LC-MS
Random survival forest method
LC-MS analysis
Wilcoxon rank-sum test
KEGG pathway enrichment analysis
Kaplan–Meier curves
LASSO regression algorithm
symmetric dimethylarginine/asymmetric dimethylarginine, neopterin, thymine, glucuronate, hydroxyproline, 14:0 Carnitine, indole acrylate, 8:0 Carnitine, acetylalanine, 2-aminoadipate, and GlcNAc6pEleven metabolites significantly discriminated the prognosis of GC patients.[62]
37LC-MS
GC-MS
Waters ACQUITY UPLC
ChromaTOF
TargetSearch
Normalization Autoencoder
t-test
PE 38:4, threonic acid, l-lysyl-l-glutamine, temorine These metabolites differed before and after surgery.[63]
218mGWAS
GO enrichment analysis
Cytoscape software, https://cytoscape.org/
Multifactorial Cox regression analysisVENTX, PCDH7, JAKMIP1, MIR202HG, MIR378D1, LINC02472, and LINC02310These functional genes regulate the metabolites via related pathways and thereby affect GC survival.[65]
Abbreviations: GC, gastric cancer; LASSO, least absolute shrinkage and selection operator; LC-MS, liquid chromatography-mass spectrometry; PCA, principal component analysis; GC-MS, gas chromatography-mass spectrometry; KEGG, The Kyoto encyclopedia of genes and genomes; UPLC, ultra-performance liquid chromatography; mGWAS, metabolite genome-wide association studies; N.A. = not applicable.
These newly detected metabolites may be significant prognostic markers or predict therapeutic targets for patients with GC.

7. Pharmacogenomics-Based Validation of Predictive Markers of GC

The prediction of treatment efficiency, such as chemosensitivity, is a major challenge in the management of GC. To select efficient drugs, recent clinical trials have focused on multi-omics profiling of patients with cancer by analyzing genome-wide data, including data from DNA sequencing, gene expression profiling, and copy number profiling [66]. Pharmacogenomics proposed to understand how genetic variants affect drug response and toxicity. The capacity to predict the efficacy of a patient with cancer on a particular treatment regimen is the elaborate goal of precision oncology [67].
The large-scale pharmacogenetic study using a drug sensitivity dataset based on patient-derived GC specimens identified the lineage-specific drug sensitivities, including the effect of epidermal growth factor receptor inhibitors for signet-ring cells, vascular endothelial growth factor receptor inhibitors for diffuse-type tumors, PARP inhibitors for tumors with BRCA2 mutation, WNT inhibitors for ALK-mutant tumors, and AKT blockade for PIK3CA-E542K mutation [68]. A study proposed to build a predictive model to guess the susceptibility to platinum of GC patients to excuse patients from unnecessary exposure to toxicity [69]. Twenty-three genes that are markedly relevant to the survival of patients with GC were detected by using univariate Cox regression analysis. Subsequently, three potential genes were selected to establish the predictive model consisting of ND6, BRMS1, and SRXN1. This model can accurately predict the outcome of GC patients after platinum-based treatment. Based on in silico data mining, the treatment benefit of up to 15% of GC samples from the TCGA dataset can be predicted by currently approved targeted therapy for GC (trastuzumab, pembrolizumab, and ramucirumab). In contrast, 50% of the patients would be predicted to benefit from FDA-approved alternative targeted therapies. A major GC target identified in this analysis was the high frequency of PIK3CA mutation variants. Other drugs identified here for therapeutic options are sorafenib (16% of TCGA GC patients), cobozantinib, and regorafenib [70]. Another study has described that six lactylation-related prognostic gene models in GC tissues were established using the GSEA, TCGA, and GEO databases [71]. Lactylation score was achieved by infiltrated immune cells and genetic instability amounts, and it was shown that lactylation score was intensely associated with GC prognosis. GC tissues with a high lactate fraction have higher immune dysfunction and reduced response to immune checkpoint inhibitors, indicating higher risks of immune evasion and dysfunction. These findings suggested that the lactate score may be useful for predicting prognosis and immune escape of GC. A recent study investigated the indicators for the efficacy of first-line chemotherapeutic agents, such as XELOX (capecitabine and oxaliplatin), DOS (docetaxel, oxaliplatin, and S-1), and targeted therapies, such as anti-HER2-based therapy by employing comprehensive proteomic technology implemented with machine learning statistics of 206 tumor samples before treatment [72]. The proteomic subtypes were established and correlated with therapy subcohorts. HER2 subcohort showed distinct distribution differences among proteomic subtypes. The expression of one of the ECM proteins, THSD4, was closely relevant to worse survival and associated with chemosensitivity (involving 5-FU, oxaliplatin, and docetaxel) in GC cell lines. Besides, highly expressed extracellular matrix proteins (THSD4, SRPX2, TGFBI, THBS1, and LAMB2) can serve as indicators to predict drug resistance. They also found PI3K-AKT signaling pathway had potential relevance to the ECM proteins (COL4A1, COL6A5, FN1, GP1BA, ITGA4, THBS3, and THBS4), and downregulation of apoptosis correlated proteins (BCL2, BCL2L1, CASP3, CASP7, and CDKN2A), suggesting the synergistic efficacy of PI3K-AKT inhibitor combined with anti-HER2 targeted agents. Interestingly, increased expression of MSI-sig CTSE enhances the effectiveness of DOS therapy but not of XELOX therapy. Meanwhile, TKTL1 is negatively associated with the sensitivity of patients to DOS therapy.
Previous studies have indicated that metabolomics analysis in GC may offer a useful approach to chemosensitivity prediction [73,74,75]. Recently, a study examined the role of metabolite profiles in sensitivity to neoadjuvant chemotherapy in GC [76]. They collected serum samples from GC patients with diverse sensitivities to chemotherapy. LC-MS analysis found that four serum metabolites (Deoxyribose 1-phosphate, S-lactoylglutathione, lysoPC (16:0), and O-arachidonoyl ethanolamine) revealed potent divergences in patients with chemo-sensitive and chemo-resistant tumors with a sensitivity and specificity of 82.5%. Besides, deoxyribose 1-phosphate and S-lactoylglutathione were independently correlated with chemosensitivity by multivariate regression analysis. The approved biomarkers and candidate biomarkers for therapeutic prediction for targeting GC using bioinformatical analysis are summarized in Table 6.
The results obtained by network analysis, combined with multi-omics data via bioinformatics algorithms, strongly suggest that the selection of therapies for patients with GC should consider these multi-omics profiles for effective individualized treatment strategies.

8. Discussion and Future Directions

This review summarizes single omics and potential multi-omics applications for predictive and prognostic biomarkers in GC. Predictive biomarkers can be used broadly to predict the prognosis and therapeutic effects and identify individuals at high risk. In this review, we focus on the role of predictive biomarkers in predicting therapeutic strategies. For a more specific patient stratification along with prognosis and therapeutic responses, comprehensive profiling using integrated bioinformatics analysis enables the classification of GC cases by molecular subtypes. As mentioned above, numerous studies have analyzed the genetic, epigenetic, transcriptomic, proteomic, or metabolomic signatures of GC and found multiple molecular alterations associated with GC. The fusion of this evidence has prompted TCGA and the Asian Cancer Research Group (ACRG) to propose several molecular classifications [77,78]. However, rational application of these classifications in clinical settings is still not widespread, mainly due to their intricacy. Beyond these studies, many researchers have sought other surrogate molecular classifications of GC to detect biomarkers and validate therapeutic prediction based on various analyses, including epigenome, proteome, metabolome, immunity, and tumor mutational burden [56,72,79,80,81,82,83,84,85,86,87]. However, no studies can be said to have succeeded in identifying alternative markers that allow more efficient categories than those of TCGA and ACRG. The application of novel molecular techniques beyond NGS, such as third-generation and single-cell sequencing, and the classification of patients in clinical trials according to molecular alterations may eventually lead to a consensus molecular classification [88].
Reproducibility, clinical feasibility, and validation in randomized clinical trials would make biomarkers more clinically relevant. To translate research results into the clinical use of a biomarker for cancer, findings must be validated by extensive clinical trials across diverse institutions. Currently, there are several bioinformatics-based clinical trials for patients with GC: three trials are ongoing (NCT06035250, NCT05319392, NCT05985577), and one trial is completed (NCT05620537). In addition, a recently completed clinical trial examined nutritional status assessments, such as Nutrition Risk Screening 2002 score and Patient-generated Subjective Global Assessment in patients with GC after radical surgery; its analysis featured the LASSO regression model as well as Cox regression (ClinicalTrials.gov identifiers: NCT05620537). A multicentered prospective cohort study is evaluating the efficacy of neoadjuvant chemotherapy and prognosis in patients with GC using a deep-learning-based predictive model integrating patients’ clinical data, biopsy pathology images, and CT imaging data (NCT06035250). Patients with GC are currently being recruited for a clinical trial to search the transcriptome profiling of GC tumor samples and adjacent normal tissue to examine DEGs based on TCGA and GEO. DEGs between cancerous and normal tissue were detected and annotated using bioinformatic analysis to gain insight into the TME (NCT05319392). Similarly, a clinical trial characterizing the prognostic proteomic landscape of GSRCC using LC-MS/MS and clinical outcome data is currently underway (NCT05985577).
Although it has recently become easier to acquire multi-omics information that can involve several layers at once, it is often challenging to fully use the gathering of large amounts of omics data available, and integrating the omics into a significant model is difficult due to the number of datasets. Thus, various integration strategies have been developed. Recent advances in proteomics through the maturation of several MS techniques have enabled the introduction of proteogenomic approaches that can integrate genomic data with proteomics and information on post-translational modifications [89]. For instance, there has been deep attention to the comprehensive characterization of the divergent mechanisms of diffuse-GC and intestinal-GC [90]. For instance, integrative proteogenomic analysis showed that the ARID1A mutation conferred contradictory prognostic influences in the two different entities. Meanwhile, diverse effects on corresponding proteomes were demonstrated, implying that diffuse-GC and intestinal-GC should be characterized based on multilevel data, including proteogenomics. More recently, comprehensive proteomic profiling using LC-MS has been applied to peritoneal carcinomatosis cells from GCs and compared with transcriptomic profiles [91]. Unsupervised hierarchical clustering analysis revealed three subgroups based on the extent of enrichment in cancer cells: Cluster A (tumor-cell enriched), Cluster B (tumor-immune mixed), and Cluster C (immune-cell enriched), which may be correlated with prognosis and drug response. When mutational profiles were integrated into proteomic data, many somatic mutations in driver genes and a higher tumor mutation burden were detected in Cluster A, while no somatic mutations were found in Cluster C. Pathway enrichment analysis displayed several enriched biological pathways (glycolysis, oxidative phosphorylation, TP53, MYC, mTORC1, TGF-β, neutrophil degranulation, neutrophil activation, neutrophil-mediated immunity, and cytokine production pathways) and several druggable targets (cancer-testis antigens, kinases, and receptors). Proteogenomic analysis revealed several genes with conflicting protein and mRNAs expression in peritoneal carcinomatosis cells, such as CTNNA2, LTF, CTAGE1, LAIR1, and HAVCR2, related to oncogenic, metabolic, and immune-related pathways, suggesting that combining proteomic and transcriptional analysis might result in a better understanding of the complex mechanisms affecting protein expression in peritoneal carcinomatosis from GC [91]. Potentially actionable therapeutic molecular targets will be detected using proteogenomic approaches to study more about the molecular composition of patients and their tumors. It will then be possible to study the correlation between molecular discoveries and cancer treatment outcomes and to facilitate novel clinical trials with prognostic and predictive biomarkers.
Despite substantial advances in the detection of carcinogenesis, the association of cancer with genetic vulnerability to microbial infections remains a challenge. The microbiome has been associated with regulating various physiological processes in human diseases, including cancer, and this connection is attracting increasing attention [92]. Among all tumors, GC is arguably the most intensely affected by gut bacteria owing to their spatial proximity, and their associations with the gut microbiome have been intensively investigated [93,94]. As a standout example, Helicobacter pylori infection is one of GC’s most important risk factors [95]. A recent study characterized the mucosal microbiome composition in paired cancerous and adjacent normal tissues and found significant correlations with the prognosis of GC patients [96]. From 128 samples, 5450 different phylotypes belonging to 19 phyla and 296 genera were detected using a bioinformatics approach. The GC patients with enrichment of Fusobacterium nucleatum and the genus Prevotella I in their cancerous tissues showed worse survival using PERMANOVA and ANOSIM analyses. Bacterial network analysis revealed that F. nucleatum and Prevotella closely interacted with each other and with gastric bacterial communities in patients with GC, indicating that the gastric microbial environment may be responsible for the GC phenotype.
As described above, there are several obstacles to the extensive implementation of omics data in clinical practice. One problem is the necessity for large phenotypically characterized datasets. High accuracy and reproducibility are required for measurement technology in multi-omics studies. It is difficult to obtain the high-grade datasets needed for omics profiling in clinical practice due to the challenge of acquiring appropriate tissue samples, strict requirements for sample preparation, and high costs. Data mining may overcome these technical challenges. Omics data can be retrieved from multiple databases or collected with technologies for disease. These are often incompatible, which makes it difficult to compare the data obtained among different research groups. Normalization of patient characteristics with GC, such as age, race, and stage, from different databases, is very important to integrate multi-omics data. When it comes to the clinical utility of the presented biomarkers, they still need to be validated through comprehensive cohort studies and large-scale clinical trials. Further data standardization and the proceeding of dominant public databases for omics data will be required in the future. Each type of omics data is distinctive to a single “layer” of biological phenomena, and single-layer omics studies have offered a useful understanding of disease mechanisms. However, the interactions of the individual layers are complex and difficult to analyze. Recent studies have shown that integrative multi-omics analysis facilitates the identification of the networks that regulate transitions from health to cancer development and evaluation in greater depth of clinically related subgroups for potential biomarkers. Meanwhile, multi-omics data formats and storage methods are diverse. The majority of the multi-omics integrative analysis methods require specific formats for data. Thus, each omics dataset requires preprocessing, which comprises systematic normalization, filtering of data, and elimination of bunch effects (Figure 1). The data filtering step has a crucial role in reducing the noise and the volume of inputs.
Along with the introduction of high-throughput technologies and the availability of multi-omics data derived from a number of samples, a large set of multi-omics data integrative tools and methods have been reported for the discovery of biomarkers. These methods can be divided into categories, such as Bayesian integration (e.g., Pathway Recognition Algorithm using Data Integration on Genomic Models (PARADIGM) [97], iClusterPlus [98], Patient-specific data fusion (PSDF) [99]), network and pathway integration (e.g., Similarity network fusion (SNF) [100], PaintOmics [101], MultiGSEA [102], multi-omics gene-set analysis (MOGSA) [103], Cytoscape [104]), fusion-based integration (e.g., Protein fusion analysis (PFS) [105]), similarity-based integration (e.g., Perturbation clustering for data integration and disease subtyping(PINSPlus) [106], Neighborhood-based multi-omics clustering (NEMO) [107]), and other multivariate approaches (e.g., mixOMICS [108], moCluster [109], Integrative nonnegative matrix factorization (NMF) [110]) [111,112]. Considering such a broad spectrum of tools/methods to integrate multi-omics datasets, a detailed comparison and benchmarking of clustering methods utilizing identical datasets may be beneficial. In addition, there is a varied array of platforms that facilitate visualization and clarification of multi-omics data, involving GENEASE [113], CGDV [114], and SLIDE [115], which provide ease of visualization and interpretation of large biological data sets. Since the methods are widely available, the development of an identical framework can efficiently process and analyze multi-omics data with easy and unified analysis. Further comprehensive reviews of statistical and algorithmic aspects of integrative multi-omics data are accessible elsewhere [111,112]. Competitive endogenous RNAs (ceRNA) is a novel regulatory mechanism that has been found to be involved in a variety of progression processes in cancer [116]. Integrative multi-omics strategies using Cytoscape were applied to examine the relationship between the prognosis and biomarkers in the STAD-linked ceRNA network by Kaplan–Meier survival analysis [45]. Fifteen genes were associated with the outcome of GC patients, involving AC011374.1, AC018781.1, ADAMTS9-AS1, AL139002.1, AL391152.1, HOTTIP, FLRT1, NKX2-1-AS1, ADAMTS9-AS2, LINC00326, VCAN-AS1, SERPINE1, POU6F2-AS2, IGF2-AS, and miR-145. Among them, in the ceRNA network, the lncRNAs VCAN-AS1, AL139002.1, LINC00326, AC018781.1, and C15orf54 acted as hub nodes. In particular, this study also found an association between TIME, (including monocytes, and neutrophils) and lncRNA-based ceRNA regulatory network [45]. In recent years, with the rapid progress of artificial intelligence, deep learning has been continuously developing. Interestingly, Chen and colleagues demonstrated a deep learning-based model on GC to find multi-omics features that were associated with the distinctive survival of GC patients [117]. They preprocessed the data and acquired 16,699 genes from RNA-seq, 390 miRNAs from miRNA-seq, and 18,992 genes from DNA methylation data as input features. These three types of omics features were stacked together using an autoencoder deep learning architecture. By the silhouette index and the Calinski–Harabasz criterion, two subgroups were identified from k-means clustering, and a significant difference in prognosis was shown between the two subgroups. Notably, implementation of the model based on the autoencoder was superior to those of two alternative familiar integrative multi-omics tools, principal component analysis and similarity network fusion. Membership in one subgroup was deemed to be a risk factor using the univariate and multivariate Cox-PH model, and the model based on the autoencoder-integrating multi-omics analysis showed more suitable performance as a prognostic marker than other clinicopathological features [117]. Thus, proper omics data creation and management and the selection of data integration methods under the complementary computational analysis, could be essential for providing the effective application of multi-omics datasets in clinical translation.
Collectively, the practical integration of bioinformatics analysis and multi-omics datasets is having a great impact on the search for predictive and prognostic biomarkers and may lead to an important revolution in treatment.

9. Materials and Methods

A non-systematic review was performed based on an electronic search through the medical literature using PubMed and Google Scholar. The keywords “gastric cancer”, “bioinformatics analysis”, “multi-omics”, “artificial intelligence”, “machine learning”, “prognosis”, “prediction”, and “biomarker” were used. Review articles and guidelines (from 2018 to 2023) investigating the current status of bioinformatic approaches in understanding GC’s genetic, epigenetic, transcriptomic, proteomic, and metabolic processes were included in this review. When more than one guideline concerning the same subject was available, the most recently updated guideline was selected. Only full articles published in English within the last ten years were considered for further review. Close attention was also given to “clinical study” and “review” articles dealing with the topic. The exclusion criteria were duplicate articles and studies lacking diagnostic outcomes. Case reports, correspondence, letters, and non-human research were excluded.

10. Conclusions

While not fully realized, the era of comprehensive molecular analysis, including not only genomics but also epigenomics, transcriptomics, proteomics, and metabolomics data, has arrived. In this review, we have summarized the use of these omics layers in the detection of predictive and prognostic biomarkers in GC. It is expected that the use of bioinformatics analysis to identify potential biomarkers leads to better therapeutic strategies. However, further elucidation is needed to apply these biomarker candidates in a clinical setting. The practical, integrative analyses of multi-platform datasets via machine learning could assist in validating novel and specific biomarkers associated with prognosis and prediction and may provide opportunities for clinical translation in the future.

Author Contributions

Conceptualization, T.M. and M.Y.; writing—original draft preparation, T.M. and M.Y.; writing—review and editing, T.M. and M.Y.; supervision, M.Y.; funding acquisition, M.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by KAKENHI (Grant-in-Aid for Scientific Research), No. 21H03008.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
  2. Ajani, J.A.; Lee, J.; Sano, T.; Janjigian, Y.Y.; Fan, D.; Song, S. Gastric adenocarcinoma. Nat. Rev. Dis. Primers 2017, 3, 17036. [Google Scholar] [CrossRef] [PubMed]
  3. Katona, B.W.; Rustgi, A.K. Gastric Cancer Genomics: Advances and Future Directions. Cell. Mol. Gastroenterol. Hepatol. 2017, 3, 211–217. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, K.; Yuen, S.T.; Xu, J.; Lee, S.P.; Yan, H.H.; Shi, S.T.; Siu, H.C.; Deng, S.; Chu, K.M.; Law, S.; et al. Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat. Genet. 2014, 46, 573–582. [Google Scholar] [CrossRef] [PubMed]
  5. Bang, Y.J.; Van Cutsem, E.; Feyereislova, A.; Chung, H.C.; Shen, L.; Sawaki, A.; Lordick, F.; Ohtsu, A.; Omuro, Y.; Satoh, T.; et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet 2010, 376, 687–697. [Google Scholar] [CrossRef] [PubMed]
  6. Hu, T.; Chitnis, N.; Monos, D.; Dinh, A. Next-generation sequencing technologies: An overview. Hum. Immunol. 2021, 82, 801–811. [Google Scholar] [CrossRef]
  7. Jiang, P.; Sinha, S.; Aldape, K.; Hannenhalli, S.; Sahinalp, C.; Ruppin, E. Big data in basic and translational cancer research. Nat. Rev. Cancer 2022, 22, 625–639. [Google Scholar] [CrossRef] [PubMed]
  8. Phillips, K.A. Assessing the Value of Next-Generation Sequencing Technologies: An Introduction. Value Health 2018, 21, 1031–1032. [Google Scholar] [CrossRef] [PubMed]
  9. Nicora, G.; Vitali, F.; Dagliati, A.; Geifman, N.; Bellazzi, R. Integrated Multi-Omics Analyses in Oncology: A Review of Machine Learning Methods and Tools. Front. Oncol. 2020, 10, 1030. [Google Scholar] [CrossRef]
  10. Li, C.; Sun, Y.D.; Yu, G.Y.; Cui, J.R.; Lou, Z.; Zhang, H.; Huang, Y.; Bai, C.G.; Deng, L.L.; Liu, P.; et al. Integrated Omics of Metastatic Colorectal Cancer. Cancer Cell 2020, 38, 734–747.e9. [Google Scholar] [CrossRef]
  11. Langfelder, P.; Horvath, S. WGCNA: An R package for weighted correlation network analysis. BMC Bioinform. 2008, 9, 559. [Google Scholar] [CrossRef] [PubMed]
  12. Szklarczyk, D.; Franceschini, A.; Wyder, S.; Forslund, K.; Heller, D.; Huerta-Cepas, J.; Simonovic, M.; Roth, A.; Santos, A.; Tsafou, K.P.; et al. STRING v10: Protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 2015, 43, D447–D452. [Google Scholar] [CrossRef] [PubMed]
  13. Hu, B.L.; Xie, M.Z.; Li, K.Z.; Li, J.L.; Gui, Y.C.; Xu, J.W. Genome-wide analysis to identify a novel distant metastasis-related gene signature predicting survival in patients with gastric cancer. Biomed. Pharmacother. 2019, 117, 109159. [Google Scholar] [CrossRef] [PubMed]
  14. Gao, W.; Yang, M. Identification by Bioinformatics Analysis of Potential Key Genes Related to the Progression and Prognosis of Gastric Cancer. Front. Oncol. 2022, 12, 881015. [Google Scholar] [CrossRef]
  15. Li, X.; Duan, Y.; Hao, Y. Identification of super enhancer-associated key genes for prognosis of germinal center B-cell type diffuse large B-cell lymphoma by integrated analysis. BMC Med. Genom. 2021, 14, 69. [Google Scholar] [CrossRef]
  16. Guo, H.; Yang, J.; Liu, S.; Qin, T.; Zhao, Q.; Hou, X.; Ren, L. Prognostic marker identification based on weighted gene co-expression network analysis and associated in vitro confirmation in gastric cancer. Bioengineered 2021, 12, 4666–4680. [Google Scholar] [CrossRef] [PubMed]
  17. Jiang, J.; Ding, Y.; Wu, M.; Lyu, X.; Wang, H.; Chen, Y.; Wang, H.; Teng, L. Identification of TYROBP and C1QB as Two Novel Key Genes With Prognostic Value in Gastric Cancer by Network Analysis. Front. Oncol. 2020, 10, 1765. [Google Scholar] [CrossRef] [PubMed]
  18. Liu, L.; Pang, H.; He, Q.; Pan, B.; Sun, X.; Shan, J.; Wu, L.; Wu, K.; Yao, X.; Guo, Y. A novel strategy to identify candidate diagnostic and prognostic biomarkers for gastric cancer. Cancer Cell Int. 2021, 21, 335. [Google Scholar] [CrossRef] [PubMed]
  19. Chivu-Economescu, M.; Necula, L.G.; Matei, L.; Dragu, D.; Bleotu, C.; Sorop, A.; Herlea, V.; Dima, S.; Popescu, I.; Diaconu, C.C. Collagen Family and Other Matrix Remodeling Proteins Identified by Bioinformatics Analysis as Hub Genes Involved in Gastric Cancer Progression and Prognosis. Int. J. Mol. Sci. 2022, 23, 3214. [Google Scholar] [CrossRef]
  20. Ilango, S.; Paital, B.; Jayachandran, P.; Padma, P.R.; Nirmaladevi, R. Epigenetic alterations in cancer. Front. Biosci. (Landmark Ed.) 2020, 25, 1058–1109. [Google Scholar] [CrossRef]
  21. Bernstein, B.E.; Stamatoyannopoulos, J.A.; Costello, J.F.; Ren, B.; Milosavljevic, A.; Meissner, A.; Kellis, M.; Marra, M.A.; Beaudet, A.L.; Ecker, J.R.; et al. The NIH Roadmap Epigenomics Mapping Consortium. Nat. Biotechnol. 2010, 28, 1045–1048. [Google Scholar] [CrossRef] [PubMed]
  22. Weisenberger, D.J. Characterizing DNA methylation alterations from The Cancer Genome Atlas. J. Clin. Investig. 2014, 124, 17–23. [Google Scholar] [CrossRef] [PubMed]
  23. Balachandran, V.P.; Gonen, M.; Smith, J.J.; DeMatteo, R.P. Nomograms in oncology: More than meets the eye. Lancet Oncol. 2015, 16, e173–e180. [Google Scholar] [CrossRef] [PubMed]
  24. Bai, Y.; Wei, C.; Zhong, Y.; Zhang, Y.; Long, J.; Huang, S.; Xie, F.; Tian, Y.; Wang, X.; Zhao, H. Development and Validation of a Prognostic Nomogram for Gastric Cancer Based on DNA Methylation-Driven Differentially Expressed Genes. Int. J. Biol. Sci. 2020, 16, 1153–1165. [Google Scholar] [CrossRef] [PubMed]
  25. Liu, D.; Li, L.; Wang, L.; Wang, C.; Hu, X.; Jiang, Q.; Wang, X.; Xue, G.; Liu, Y.; Xue, D. Recognition of DNA Methylation Molecular Features for Diagnosis and Prognosis in Gastric Cancer. Front. Genet. 2021, 12, 758926. [Google Scholar] [CrossRef] [PubMed]
  26. Li, Y.; Sun, R.; Zhang, Y.; Yuan, Y.; Miao, Y. A methylation-based mRNA signature predicts survival in patients with gastric cancer. Cancer Cell Int. 2020, 20, 284. [Google Scholar] [CrossRef] [PubMed]
  27. Mantione, K.J.; Kream, R.M.; Kuzelova, H.; Ptacek, R.; Raboch, J.; Samuel, J.M.; Stefano, G.B. Comparing bioinformatic gene expression profiling methods: Microarray and RNA-Seq. Med. Sci. Monit. Basic. Res. 2014, 20, 138–142. [Google Scholar] [CrossRef]
  28. Lowe, R.; Shirley, N.; Bleackley, M.; Dolan, S.; Shafee, T. Transcriptomics technologies. PLoS Comput. Biol. 2017, 13, e1005457. [Google Scholar] [CrossRef] [PubMed]
  29. Wang, Z.; Gerstein, M.; Snyder, M. RNA-Seq: A revolutionary tool for transcriptomics. Nat. Rev. Genet. 2009, 10, 57–63. [Google Scholar] [CrossRef]
  30. Jacquier, A. The complex eukaryotic transcriptome: Unexpected pervasive transcription and novel small RNAs. Nat. Rev. Genet. 2009, 10, 833–844. [Google Scholar] [CrossRef]
  31. Pan, J.; Gao, Y. Prognostic significance and immune characteristics of GPR27 in gastric cancer. Aging 2023, 15, 9144–9166. [Google Scholar] [CrossRef] [PubMed]
  32. Kashima, Y.; Sakamoto, Y.; Kaneko, K.; Seki, M.; Suzuki, Y.; Suzuki, A. Single-cell sequencing techniques from individual to multiomics analyses. Exp. Mol. Med. 2020, 52, 1419–1427. [Google Scholar] [CrossRef] [PubMed]
  33. Chen, J.; Liu, K.; Luo, Y.; Kang, M.; Wang, J.; Chen, G.; Qi, J.; Wu, W.; Wang, B.; Han, Y.; et al. Single-Cell Profiling of Tumor Immune Microenvironment Reveals Immune Irresponsiveness in Gastric Signet-Ring Cell Carcinoma. Gastroenterology 2023, 165, 88–103. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, Y.; Liu, W.; Feng, W.; Wang, X.; Lei, T.; Chen, Z.; Song, W. Identification of 14 Differentially-Expressed Metabolism-Related Genes as Potential Targets of Gastric Cancer by Integrated Proteomics and Transcriptomics. Front. Cell Dev. Biol. 2022, 10, 816249. [Google Scholar] [CrossRef] [PubMed]
  35. Jiang, C.; Chen, X.; Alattar, M.; Wei, J.; Liu, H. MicroRNAs in tumorigenesis, metastasis, diagnosis and prognosis of gastric cancer. Cancer Gene Ther. 2015, 22, 291–301. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, Y.; Yang, F.; Yang, Q. The regulatory roles and potential prognosis implications of long non-coding RNAs in gastric cancer. Histol. Histopathol. 2020, 35, 433–442. [Google Scholar] [CrossRef] [PubMed]
  37. Zhang, C.; Zhang, C.D.; Ma, M.H.; Dai, D.Q. Three-microRNA signature identified by bioinformatics analysis predicts prognosis of gastric cancer patients. World J. Gastroenterol. 2018, 24, 1206–1215. [Google Scholar] [CrossRef] [PubMed]
  38. Dos Santos, E.C.; Rohan, P.; Binato, R.; Abdelhay, E. Integrated Network Analysis of microRNAs, mRNAs, and Proteins Reveals the Regulatory Interaction between hsa-mir-200b and CFL2 Associated with Advanced Stage and Poor Prognosis in Patients with Intestinal Gastric Cancer. Cancers 2023, 15, 5374. [Google Scholar] [CrossRef]
  39. Lin, C.; Yang, L. Long Noncoding RNA in Cancer: Wiring Signaling Circuitry. Trends Cell Biol. 2018, 28, 287–301. [Google Scholar] [CrossRef]
  40. Li, G.; Huo, D.; Guo, N.; Li, Y.; Ma, H.; Liu, L.; Xie, H.; Zhang, D.; Qu, B.; Chen, X. Integrating multiple machine learning algorithms for prognostic prediction of gastric cancer based on immune-related lncRNAs. Front. Genet. 2023, 14, 1106724. [Google Scholar] [CrossRef]
  41. Qi, X.; Chen, X.; Zhao, Y.; Chen, J.; Niu, B.; Shen, B. Prognostic Roles of ceRNA Network-Based Signatures in Gastrointestinal Cancers. Front. Oncol. 2022, 12, 921194. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, X.; Zhang, W.; Jiang, Y.; Liu, K.; Ran, L.; Song, F. Identification of functional lncRNAs in gastric cancer by integrative analysis of GEO and TCGA data. J. Cell. Biochem. 2019, 120, 17898–17911. [Google Scholar] [CrossRef] [PubMed]
  43. Liao, T.; Lu, Y.; Li, W.; Wang, K.; Zhang, Y.; Luo, Z.; Ju, Y.; Ouyang, M. Construction and validation of a glycolysis-related lncRNA signature for prognosis prediction in Stomach Adenocarcinoma. Front. Genet. 2022, 13, 794621. [Google Scholar] [CrossRef] [PubMed]
  44. Dai, J.; Li, Z.X.; Zhang, Y.; Ma, J.L.; Zhou, T.; You, W.C.; Li, W.Q.; Pan, K.F. Whole Genome Messenger RNA Profiling Identifies a Novel Signature to Predict Gastric Cancer Survival. Clin. Transl. Gastroenterol. 2019, 10, e00004. [Google Scholar] [CrossRef] [PubMed]
  45. Xu, J.; Hu, S.; Chen, Q.; Shu, L.; Wang, P.; Wang, J. Integrated bioinformatics analysis of noncoding RNAs with tumor immune microenvironment in gastric cancer. Sci. Rep. 2023, 13, 15006. [Google Scholar] [CrossRef] [PubMed]
  46. Hedl, T.J.; San Gil, R.; Cheng, F.; Rayner, S.L.; Davidson, J.M.; De Luca, A.; Villalva, M.D.; Ecroyd, H.; Walker, A.K.; Lee, A. Proteomics Approaches for Biomarker and Drug Target Discovery in ALS and FTD. Front. Neurosci. 2019, 13, 548. [Google Scholar] [CrossRef] [PubMed]
  47. Li, D.; Chan, D.W. Proteomic cancer biomarkers from discovery to approval: It’s worth the effort. Expert. Rev. Proteom. 2014, 11, 135–136. [Google Scholar] [CrossRef] [PubMed]
  48. Zheng, G.L.; Zhang, G.J.; Zhao, Y.; Zheng, Z.C. Screening Protein Prognostic Biomarkers for Stomach Adenocarcinoma Based on The Cancer Proteome Atlas. Front. Oncol. 2022, 12, 901182. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, Y.; Gao, X.; Wang, J. Functional Proteomic Profiling Analysis in Four Major Types of Gastrointestinal Cancers. Biomolecules 2023, 13, 701. [Google Scholar] [CrossRef]
  50. Moreira, A.M.; Ferreira, R.M.; Carneiro, P.; Figueiredo, J.; Osorio, H.; Barbosa, J.; Preto, J.; Pinto-do, O.P.; Carneiro, F.; Seruca, R. Proteomic Identification of a Gastric Tumor ECM Signature Associated With Cancer Progression. Front. Mol. Biosci. 2022, 9, 818552. [Google Scholar] [CrossRef]
  51. Santos, E.C.; Binato, R.; Fernandes, P.V.; Ferreira, M.A.; Abdelhay, E. The protein-protein interaction network of intestinal gastric cancer patients reveals hub proteins with potential prognostic value. Cancer Biomark. 2022, 33, 83–96. [Google Scholar] [CrossRef] [PubMed]
  52. Zhao, X.; Xia, X.; Wang, X.; Bai, M.; Zhan, D.; Shu, K. Deep Learning-Based Protein Features Predict Overall Survival and Chemotherapy Benefit in Gastric Cancer. Front. Oncol. 2022, 12, 847706. [Google Scholar] [CrossRef] [PubMed]
  53. Clish, C.B. Metabolomics: An emerging but powerful tool for precision medicine. Cold Spring Harb. Mol. Case Stud. 2015, 1, a000588. [Google Scholar] [CrossRef]
  54. Kaushik, A.K.; DeBerardinis, R.J. Applications of metabolomics to study cancer metabolism. Biochim. Biophys. Acta Rev. Cancer 2018, 1870, 2–14. [Google Scholar] [CrossRef]
  55. Vernieri, C.; Casola, S.; Foiani, M.; Pietrantonio, F.; de Braud, F.; Longo, V. Targeting Cancer Metabolism: Dietary and Pharmacologic Interventions. Cancer Discov. 2016, 6, 1315–1333. [Google Scholar] [CrossRef] [PubMed]
  56. Aftabi, Y.; Soleymani, J.; Jouyban, A. Efficacy of Analytical Technologies in Metabolomics Studies of the Gastrointestinal Cancers. Crit. Rev. Anal. Chem. 2022, 52, 1593–1605. [Google Scholar] [CrossRef]
  57. Lei, C.; Gong, D.; Zhuang, B.; Zhang, Z. Alterations in the gastric microbiota and metabolites in gastric cancer: An update review. Front. Oncol. 2022, 12, 960281. [Google Scholar] [CrossRef]
  58. Cao, K.; Lyu, Y.; Chen, J.; He, C.; Lyu, X.; Zhang, Y.; Chen, L.; Jiang, Y.; Xiang, J.; Liu, B.; et al. Prognostic Implication of Plasma Metabolites in Gastric Cancer. Int. J. Mol. Sci. 2023, 24, 12774. [Google Scholar] [CrossRef]
  59. Pan, G.; Ma, Y.; Suo, J.; Li, W.; Zhang, Y.; Qin, S.; Jiao, Y.; Zhang, S.; Li, S.; Kong, Y.; et al. Discovering Biomarkers in Peritoneal Metastasis of Gastric Cancer by Metabolomics. OncoTargets Ther. 2020, 13, 7199–7211. [Google Scholar] [CrossRef]
  60. Sun, H.; Huang, X.; Wang, Z.; Zhang, G.; Mei, Y.; Wang, Y.; Nie, Z.; Wang, S. Triglyceride-to-high density lipoprotein cholesterol ratio predicts clinical outcomes in patients with gastric cancer. J. Cancer 2019, 10, 6829–6836. [Google Scholar] [CrossRef]
  61. Qu, T.; Zhang, S.; Yang, S.; Li, S.; Wang, D. Utilizing serum metabolomics for assessing postoperative efficacy and monitoring recurrence in gastric cancer patients. BMC Cancer 2024, 24, 27. [Google Scholar] [CrossRef] [PubMed]
  62. Chen, Y.; Wang, B.; Zhao, Y.; Shao, X.; Wang, M.; Ma, F.; Yang, L.; Nie, M.; Jin, P.; Yao, K.; et al. Metabolomic machine learning predictor for diagnosis and prognosis of gastric cancer. Nat. Commun. 2024, 15, 1657. [Google Scholar] [CrossRef] [PubMed]
  63. Song, G.; Wang, L.; Tang, J.; Li, H.; Pang, S.; Li, Y.; Liu, L.; Hu, J. Circulating metabolites as potential biomarkers for the early detection and prognosis surveillance of gastrointestinal cancers. Metabolomics 2023, 19, 36. [Google Scholar] [CrossRef] [PubMed]
  64. Chang, L.; Zhou, G.; Ou, H.; Xia, J. mGWAS-Explorer: Linking SNPs, Genes, Metabolites, and Diseases for Functional Insights. Metabolites 2022, 12, 526. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, Y.; Lyu, Y.; Chen, L.; Cao, K.; Chen, J.; He, C.; Lyu, X.; Jiang, Y.; Xiang, J.; Liu, B.; et al. Exploring the Prognosis-Related Genetic Variation in Gastric Cancer Based on mGWAS. Int. J. Mol. Sci. 2023, 24, 15259. [Google Scholar] [CrossRef] [PubMed]
  66. Horak, P.; Heining, C.; Kreutzfeldt, S.; Hutter, B.; Mock, A.; Hullein, J.; Frohlich, M.; Uhrig, S.; Jahn, A.; Rump, A.; et al. Comprehensive Genomic and Transcriptomic Analysis for Guiding Therapeutic Decisions in Patients with Rare Cancers. Cancer Discov. 2021, 11, 2780–2795. [Google Scholar] [CrossRef] [PubMed]
  67. Morganti, S.; Tarantino, P.; Ferraro, E.; D’Amico, P.; Duso, B.A.; Curigliano, G. Next Generation Sequencing (NGS): A Revolutionary Technology in Pharmacogenomics and Personalized Medicine in Cancer. Adv. Exp. Med. Biol. 2019, 1168, 9–30. [Google Scholar] [CrossRef] [PubMed]
  68. Sa, J.K.; Hong, J.Y.; Lee, I.K.; Kim, J.S.; Sim, M.H.; Kim, H.J.; An, J.Y.; Sohn, T.S.; Lee, J.H.; Bae, J.M.; et al. Comprehensive pharmacogenomic characterization of gastric cancer. Genome Med. 2020, 12, 17. [Google Scholar] [CrossRef]
  69. Pan, J.; Xiang, Z.; Dai, Q.; Wang, Z.; Liu, B.; Li, C. Prediction of platinum-resistance patients of gastric cancer using bioinformatics. J. Cell. Biochem. 2019, 120, 13478–13486. [Google Scholar] [CrossRef]
  70. Hescheler, D.A.; Plum, P.S.; Zander, T.; Quaas, A.; Korenkov, M.; Gassa, A.; Michel, M.; Bruns, C.J.; Alakus, H. Identification of targeted therapy options for gastric adenocarcinoma by comprehensive analysis of genomic data. Gastric Cancer 2020, 23, 627–638. [Google Scholar] [CrossRef]
  71. Yang, H.; Zou, X.; Yang, S.; Zhang, A.; Li, N.; Ma, Z. Identification of lactylation related model to predict prognostic, tumor infiltrating immunocytes and response of immunotherapy in gastric cancer. Front. Immunol. 2023, 14, 1149989. [Google Scholar] [CrossRef] [PubMed]
  72. Li, Y.; Xu, C.; Wang, B.; Xu, F.; Ma, F.; Qu, Y.; Jiang, D.; Li, K.; Feng, J.; Tian, S.; et al. Proteomic characterization of gastric cancer response to chemotherapy and targeted therapy reveals new therapeutic strategies. Nat. Commun. 2022, 13, 5723. [Google Scholar] [CrossRef] [PubMed]
  73. Wang, X.; Yan, S.K.; Dai, W.X.; Liu, X.R.; Zhang, W.D.; Wang, J.J. A metabonomic approach to chemosensitivity prediction of cisplatin plus 5-fluorouracil in a human xenograft model of gastric cancer. Int. J. Cancer 2010, 127, 2841–2850. [Google Scholar] [CrossRef] [PubMed]
  74. Sasada, S.; Miyata, Y.; Tsutani, Y.; Tsuyama, N.; Masujima, T.; Hihara, J.; Okada, M. Metabolomic analysis of dynamic response and drug resistance of gastric cancer cells to 5-fluorouracil. Oncol. Rep. 2013, 29, 925–931. [Google Scholar] [CrossRef] [PubMed]
  75. Kim, K.B.; Yang, J.Y.; Kwack, S.J.; Kim, H.S.; Ryu, D.H.; Kim, Y.J.; Bae, J.Y.; Lim, D.S.; Choi, S.M.; Kwon, M.J.; et al. Potential metabolomic biomarkers for evaluation of adriamycin efficacy using a urinary 1H-NMR spectroscopy. J. Appl. Toxicol. 2013, 33, 1251–1259. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, D.; Li, W.; Yin, L.; Du, Y.; Zhang, S.; Suo, J. Association of serum levels of deoxyribose 1-phosphate and S-lactoylglutathione with neoadjuvant chemotherapy sensitivity in patients with gastric cancer: A metabolomics study. Oncol. Lett. 2020, 19, 2231–2242. [Google Scholar] [CrossRef] [PubMed]
  77. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar] [CrossRef]
  78. Cristescu, R.; Lee, J.; Nebozhyn, M.; Kim, K.M.; Ting, J.C.; Wong, S.S.; Liu, J.; Yue, Y.G.; Wang, J.; Yu, K.; et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat. Med. 2015, 21, 449–456. [Google Scholar] [CrossRef]
  79. Wang, H.; Ding, Y.; Chen, Y.; Jiang, J.; Chen, Y.; Lu, J.; Kong, M.; Mo, F.; Huang, Y.; Zhao, W.; et al. A novel genomic classification system of gastric cancer via integrating multidimensional genomic characteristics. Gastric Cancer 2021, 24, 1227–1241. [Google Scholar] [CrossRef]
  80. Furukawa, K.; Hatakeyama, K.; Terashima, M.; Nagashima, T.; Urakami, K.; Ohshima, K.; Notsu, A.; Sugino, T.; Yagi, T.; Fujiya, K.; et al. Molecular classification of gastric cancer predicts survival in patients undergoing radical gastrectomy based on project HOPE. Gastric Cancer 2022, 25, 138–148. [Google Scholar] [CrossRef]
  81. Lin, Y.; Pan, X.; Zhao, L.; Yang, C.; Zhang, Z.; Wang, B.; Gao, Z.; Jiang, K.; Ye, Y.; Wang, S.; et al. Immune cell infiltration signatures identified molecular subtypes and underlying mechanisms in gastric cancer. NPJ Genom. Med. 2021, 6, 83. [Google Scholar] [CrossRef] [PubMed]
  82. Li, L.; Ma, J. Molecular characterization of metabolic subtypes of gastric cancer based on metabolism-related lncRNA. Sci. Rep. 2021, 11, 21491. [Google Scholar] [CrossRef]
  83. Wu, D.; Feng, M.; Shen, H.; Shen, X.; Hu, J.; Liu, J.; Yang, Y.; Li, Y.; Yang, M.; Wang, W.; et al. Prediction of Two Molecular Subtypes of Gastric Cancer Based on Immune Signature. Front. Genet. 2021, 12, 793494. [Google Scholar] [CrossRef] [PubMed]
  84. Cheong, J.H.; Wang, S.C.; Park, S.; Porembka, M.R.; Christie, A.L.; Kim, H.; Kim, H.S.; Zhu, H.; Hyung, W.J.; Noh, S.H.; et al. Development and validation of a prognostic and predictive 32-gene signature for gastric cancer. Nat. Commun. 2022, 13, 774. [Google Scholar] [CrossRef] [PubMed]
  85. Ye, Y.; Yang, W.; Ruan, X.; Xu, L.; Cheng, W.; Zhao, M.; Wang, X.; Chen, X.; Cai, D.; Li, G.; et al. Metabolism-associated molecular classification of gastric adenocarcinoma. Front. Oncol. 2022, 12, 1024985. [Google Scholar] [CrossRef] [PubMed]
  86. Zhou, Q.; Yuan, Y.; Lu, H.; Li, X.; Liu, Z.; Gan, J.; Yue, Z.; Wu, J.; Sheng, J.; Xin, L. Cancer functional states-based molecular subtypes of gastric cancer. J. Transl. Med. 2023, 21, 80. [Google Scholar] [CrossRef] [PubMed]
  87. Weng, S.; Li, M.; Deng, J.; Xu, H.; Ren, Y.; Zhou, Z.; Wang, L.; Zhang, Y.; Xing, Z.; Li, L.; et al. Epigenetically regulated gene expression profiles decipher four molecular subtypes with prognostic and therapeutic implications in gastric cancer. Clin. Epigenetics 2023, 15, 64. [Google Scholar] [CrossRef] [PubMed]
  88. Diaz Del Arco, C.; Fernandez Acenero, M.J.; Ortega Medina, L. Molecular Classifications in Gastric Cancer: A Call for Interdisciplinary Collaboration. Int. J. Mol. Sci. 2024, 25, 2649. [Google Scholar] [CrossRef] [PubMed]
  89. Mani, D.R.; Krug, K.; Zhang, B.; Satpathy, S.; Clauser, K.R.; Ding, L.; Ellis, M.; Gillette, M.A.; Carr, S.A. Cancer proteogenomics: Current impact and future prospects. Nat. Rev. Cancer 2022, 22, 298–313. [Google Scholar] [CrossRef]
  90. Shi, W.; Wang, Y.; Xu, C.; Li, Y.; Ge, S.; Bai, B.; Zhang, K.; Wang, Y.; Zheng, N.; Wang, J.; et al. Multilevel proteomic analyses reveal molecular diversity between diffuse-type and intestinal-type gastric cancer. Nat. Commun. 2023, 14, 835. [Google Scholar] [CrossRef]
  91. Zhao, S.; Wang, R.; Song, S.; Hao, D.; Han, G.; Song, X.; Zhang, J.; Pizzi, M.P.; Shanbhag, N.; Futreal, A.; et al. Proteogenomic landscape of gastric adenocarcinoma peritoneal metastases. iScience 2023, 26, 106913. [Google Scholar] [CrossRef] [PubMed]
  92. Cullin, N.; Azevedo Antunes, C.; Straussman, R.; Stein-Thoeringer, C.K.; Elinav, E. Microbiome and cancer. Cancer Cell 2021, 39, 1317–1341. [Google Scholar] [CrossRef] [PubMed]
  93. Zitvogel, L.; Galluzzi, L.; Viaud, S.; Vetizou, M.; Daillere, R.; Merad, M.; Kroemer, G. Cancer and the gut microbiota: An unexpected link. Sci. Transl. Med. 2015, 7, 271ps1. [Google Scholar] [CrossRef] [PubMed]
  94. Janney, A.; Powrie, F.; Mann, E.H. Host-microbiota maladaptation in colorectal cancer. Nature 2020, 585, 509–517. [Google Scholar] [CrossRef] [PubMed]
  95. Polk, D.B.; Peek, R.M., Jr. Helicobacter pylori: Gastric cancer and beyond. Nat. Rev. Cancer 2010, 10, 403–414. [Google Scholar] [CrossRef] [PubMed]
  96. Lehr, K.; Nikitina, D.; Vilchez-Vargas, R.; Steponaitiene, R.; Thon, C.; Skieceviciene, J.; Schanze, D.; Zenker, M.; Malfertheiner, P.; Kupcinskas, J.; et al. Microbial composition of tumorous and adjacent gastric tissue is associated with prognosis of gastric cancer. Sci. Rep. 2023, 13, 4640. [Google Scholar] [CrossRef] [PubMed]
  97. Vaske, C.J.; Benz, S.C.; Sanborn, J.Z.; Earl, D.; Szeto, C.; Zhu, J.; Haussler, D.; Stuart, J.M. Inference of patient-specific pathway activities from multi-dimensional cancer genomics data using PARADIGM. Bioinformatics 2010, 26, i237–i245. [Google Scholar] [CrossRef] [PubMed]
  98. Mo, Q.; Wang, S.; Seshan, V.E.; Olshen, A.B.; Schultz, N.; Sander, C.; Powers, R.S.; Ladanyi, M.; Shen, R. Pattern discovery and cancer gene identification in integrated cancer genomic data. Proc. Natl. Acad. Sci. USA 2013, 110, 4245–4250. [Google Scholar] [CrossRef] [PubMed]
  99. Yuan, Y.; Savage, R.S.; Markowetz, F. Patient-specific data fusion defines prognostic cancer subtypes. PLOS Comput. Biol. 2011, 7, e1002227. [Google Scholar] [CrossRef]
  100. Wang, B.; Mezlini, A.M.; Demir, F.; Fiume, M.; Tu, Z.; Brudno, M.; Haibe-Kains, B.; Goldenberg, A. Similarity network fusion for aggregating data types on a genomic scale. Nat. Methods 2014, 11, 333–337. [Google Scholar] [CrossRef]
  101. Hernandez-de-Diego, R.; Tarazona, S.; Martinez-Mira, C.; Balzano-Nogueira, L.; Furio-Tari, P.; Pappas, G.J., Jr.; Conesa, A. PaintOmics 3: A web resource for the pathway analysis and visualization of multi-omics data. Nucleic Acids Res. 2018, 46, W503–W509. [Google Scholar] [CrossRef] [PubMed]
  102. Canzler, S.; Hackermuller, J. multiGSEA: A GSEA-based pathway enrichment analysis for multi-omics data. BMC Bioinform. 2020, 21, 561. [Google Scholar] [CrossRef] [PubMed]
  103. Meng, C.; Basunia, A.; Peters, B.; Gholami, A.M.; Kuster, B.; Culhane, A.C. MOGSA: Integrative Single Sample Gene-set Analysis of Multiple Omics Data. Mol. Cell. Proteom. 2019, 18 (Suppl. S1), S153–S168. [Google Scholar] [CrossRef] [PubMed]
  104. Franz, M.; Lopes, C.T.; Fong, D.; Kucera, M.; Cheung, M.; Siper, M.C.; Huck, G.; Dong, Y.; Sumer, O.; Bader, G.D. Cytoscape.js 2023 update: A graph theory library for visualization and analysis. Bioinformatics 2023, 39, btad031. [Google Scholar] [CrossRef] [PubMed]
  105. Shi, Q.; Zhang, C.; Peng, M.; Yu, X.; Zeng, T.; Liu, J.; Chen, L. Pattern fusion analysis by adaptive alignment of multiple heterogeneous omics data. Bioinformatics 2017, 33, 2706–2714. [Google Scholar] [CrossRef] [PubMed]
  106. Nguyen, H.; Shrestha, S.; Draghici, S.; Nguyen, T. PINSPlus: A tool for tumor subtype discovery in integrated genomic data. Bioinformatics 2019, 35, 2843–2846. [Google Scholar] [CrossRef] [PubMed]
  107. Rappoport, N.; Shamir, R. NEMO: Cancer subtyping by integration of partial multi-omic data. Bioinformatics 2019, 35, 3348–3356. [Google Scholar] [CrossRef] [PubMed]
  108. Welham, Z.; Dejean, S.; Le Cao, K.A. Multivariate Analysis with the R Package mixOmics. Methods Mol. Biol. 2023, 2426, 333–359. [Google Scholar] [CrossRef] [PubMed]
  109. Meng, C.; Helm, D.; Frejno, M.; Kuster, B. moCluster: Identifying Joint Patterns Across Multiple Omics Data Sets. J. Proteome Res. 2016, 15, 755–765. [Google Scholar] [CrossRef]
  110. Yang, Z.; Michailidis, G. A non-negative matrix factorization method for detecting modules in heterogeneous omics multi-modal data. Bioinformatics 2016, 32, 1–8. [Google Scholar] [CrossRef]
  111. Subramanian, I.; Verma, S.; Kumar, S.; Jere, A.; Anamika, K. Multi-omics Data Integration, Interpretation, and Its Application. Bioinform. Biol. Insights 2020, 14, 1177932219899051. [Google Scholar] [CrossRef] [PubMed]
  112. Athieniti, E.; Spyrou, G.M. A guide to multi-omics data collection and integration for translational medicine. Comput. Struct. Biotechnol. J. 2023, 21, 134–149. [Google Scholar] [CrossRef] [PubMed]
  113. Ghandikota, S.; Hershey, G.K.K.; Mersha, T.B. GENEASE: Real time bioinformatics tool for multi-omics and disease ontology exploration, analysis and visualization. Bioinformatics 2018, 34, 3160–3168. [Google Scholar] [CrossRef] [PubMed]
  114. Jha, V.; Singh, G.; Kumar, S.; Sonawane, A.; Jere, A.; Anamika, K. CGDV: A webtool for circular visualization of genomics and transcriptomics data. BMC Genom. 2017, 18, 823. [Google Scholar] [CrossRef] [PubMed]
  115. Ghosh, S.; Datta, A.; Tan, K.; Choi, H. SLIDE—A web-based tool for interactive visualization of large-scale—Omics data. Bioinformatics 2019, 35, 346–348. [Google Scholar] [CrossRef] [PubMed]
  116. Wu, X.; Sui, Z.; Zhang, H.; Wang, Y.; Yu, Z. Integrated Analysis of lncRNA-Mediated ceRNA Network in Lung Adenocarcinoma. Front. Oncol. 2020, 10, 554759. [Google Scholar] [CrossRef]
  117. Chen, S.; Zang, Y.; Xu, B.; Lu, B.; Ma, R.; Miao, P.; Chen, B. An Unsupervised Deep Learning-Based Model Using Multiomics Data to Predict Prognosis of Patients with Stomach Adenocarcinoma. Comput. Math. Methods Med. 2022, 2022, 5844846. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram of validation of biomarker by the integration of bioinformatic analysis with multi-omics platforms. Different omics technologies and their features, such as omics targeted, sequencing techniques, data processing, data analysis, and output, were displayed. The first step is the identification of high-quality samples obtained by blood, tissue samples from GC patients, or high-throughput datasets. Multi-platform omics technologies are analyzed individually to identify the DNA, RNA, proteins, and metabolites responsible for the GC progression. Omics data are extracted by the search for various omics databases, mining of the data, and obtaining the pre-processed omics data. Thereby, data are transformed and processed through normalization, quality control, and feature selection to mine interpretable information. These multidimensional data can be integrated based on statistical analysis, machine learning, and functional analysis, which lead to the discovery of putative biomarkers.
Figure 1. Schematic diagram of validation of biomarker by the integration of bioinformatic analysis with multi-omics platforms. Different omics technologies and their features, such as omics targeted, sequencing techniques, data processing, data analysis, and output, were displayed. The first step is the identification of high-quality samples obtained by blood, tissue samples from GC patients, or high-throughput datasets. Multi-platform omics technologies are analyzed individually to identify the DNA, RNA, proteins, and metabolites responsible for the GC progression. Omics data are extracted by the search for various omics databases, mining of the data, and obtaining the pre-processed omics data. Thereby, data are transformed and processed through normalization, quality control, and feature selection to mine interpretable information. These multidimensional data can be integrated based on statistical analysis, machine learning, and functional analysis, which lead to the discovery of putative biomarkers.
Ijms 25 05880 g001
Table 1. Genomics-based predictive/prognostic GC biomarkers using bioinformatics analysis for the past 5 years.
Table 1. Genomics-based predictive/prognostic GC biomarkers using bioinformatics analysis for the past 5 years.
Number of PatientsDatabasesAnalytical PlatformStatistical AnalysisGene set IdentifiedCommentsRefs.
415TCGAEdgeR and DESeq packages
GO and KEGG pathway enrichment analyses
Cox regression analysisPCOLCE2, TMEM132C, UPK1B, SLITRK2, PM20D1, FLJ3502Six distant metastasis-related genes can be an independent indicator of the prognosis of GC and as a clinically useful classification tool to facilitate individualized GC therapy.[13]
N.A.GEO
(GSE84433, GSE26942)
TCGA
Limma and pheatmap package
WGCNA
GO and KEGG pathway enrichment analyses
Kaplan–Meier PlotterCST1, CEMIP, COL8A1, PMEPA1, MSLNFive genes were identified as the hub genes by utilizing WGCNA. CEMIP plays an important role in GC progression[16]
375TGCA
TIMER
Limma package WGCNACox analysisRXRG, AGT, BCHE, UBE2QL1, PLCXD3, ADCYAP1R1, NRCAM, MAMDC2, CDH19, GAMTAGT expression is higher in GC and BRD9, GOLPH3, NOM1, KLHL25, and PSMD11 interact with AGT, which is closely related to the poor prognosis.[14]
60GEO
(GSE15459) TCGA
GO cellular component analysisCox regression analysisAGT, SERPINH1, MMP7The exosome levels of AGT and MMP7 were associated with the serum level and stimulated migration of GC cell lines.[18]
747GEO
(GSE65801, GSE54129, GSE118916, GSE15459, GSE51575, GSE65801)
TCGA-STAD
Limma and edgeR R packages
GO and KEGG pathway enrichment analyses
WGCNA
PPI network
TIMER
Kaplan–Meier survival analysis log-rank testTRYOBP, C1QBTRYOBP and C1QB were correlated with the CD8+ T cells, CD4+ T cells, and macrophages[17]
351GEO
(GSE103236,GSE13911, GSE79973)
Geo2R analysis.
DAVID, GO, and KEGG pathway enrichment analyses
PPI network
TIMER
Kaplan–Meier plotterCTHRC1, BGN, FAP, THBS2, COL12A1, COL5A2, SULF1, SPP1, COL10A1Nine genes negatively correlated with poor outcomes and an immune infiltrate based especially on immunosuppressive M2 macrophages.[19]
Abbreviations: GC, gastric cancer; TCGA, The Cancer Genome Atlas; GEO, Gene Expression Omnibus; GO, gene oncology; KEGG, The Kyoto Encyclopedia of Genes and Genomes; WGCNA, weighted correlation network analysis; TIMER, Tumor Immune Estimation Resource; PPI, protein–protein interaction; N.A., not applicable.
Table 2. Epigenomics-based predictive/prognostic GC biomarkers using bioinformatics analysis for the past 5 years.
Table 2. Epigenomics-based predictive/prognostic GC biomarkers using bioinformatics analysis for the past 5 years.
Number of PatientsDatabasesAnalytical PlatformStatistical AnalysisGene set IdentifiedCommentsRefs.
343GEO (GSE62254)
TCGA
cBioPortal
GSEA
DESeq package
DAVID
ConsensusPathDB
MethylMix analysis
GO and
KEGG pathway enrichment analyses
Kaplan–Meier analysis
LASSO
Cox regression model
PODN, MYO1A, NPY, MICU3, TUBB6, RHOJSix DNA methylation-driven DEGs identified using an OS nomogram were associated with good prognosis.[24]
192GEO
(GSE13911, GSE30601, GSE79973, GSE25869, GSE15459)
TCGA
Illumina microarray platform
Geo2R analysis.
DAVID, GO and
KEGG pathway enrichment analyses
Cox and LASSO regression analysis
Kaplan–Meier method
TREM2, MICAL2, INHBA, PCSK5, NRP1, YAP1, RAI14, MATN3The eight-MDEGs signature risk score showed statistical significance as an independent variable.[26]
443GEO
(GSE30601)
TCGA
Wilcoxon test
Pheatmappackage
cBioPortal
KEGG pathway enrichment analyses
PPI network
Cox and LASSO regression analysis
Kaplan–Meier method
CEP290, CCDC69, UBXN8, KDM4A, AKR1B1, RASSF2, CHRNB2, EGR1, ARMC9, RPN1Eleven prognostic-related DMSs were positively correlated with the VIM gene and negatively correlated with the CDH1 gene.[25]
Abbreviations: GC, gastric cancer; TCGA, The Cancer Genome Atlas; GEO, Gene Expression Omnibus; GO, gene oncology; KEGG, The Kyoto Encyclopedia of Genes and Genomes; PPI, protein-protein interaction; DMS, differential methylation site.
Table 3. Transcriptmics-based predictive/prognostic GC biomarkers using bioinformatics analysis for the past 5 years.
Table 3. Transcriptmics-based predictive/prognostic GC biomarkers using bioinformatics analysis for the past 5 years.
Number of PatientsDatabasesAnalytical PlatformStatistical AnalysisGene Set IdentifiedCommentsRefs.
32GEO (GSE212212)scRNA-seq
CellChat R package
T-test, Wilcoxon rank-sum test, and Fisher’s exact test.CXCL13GSRCC TIME may be dormant, where Treg-FOXP3 and CD8-Tex were hard to mobilize and impaired B cells’ accurate activities.[33]
48TCGA
TIMER
GSEA
RNA sequencing
GO and KEGG pathway enrichment analyses
Kaplan–Meier Plotter databaseBCAT2, ALDH1A2, MDH1, PHGDH, CKB, ADH1B, PCCB, NNT, CKM, DCXR, LIPF, ASS1, ME3, CSFourteen metabolic genes were significantly enriched and were correlated with PD-1.[34]
361GEO
(GSE883415)
TCGA
Limma package Functional Enrichment analysisKaplan–Meier method,
Log-rank test
Cox regression analysis
miR-145-3p, miR-125b-5p, and miR-99a-5pThe multivariate analysis showed that the three DEM signatures were all independent factors in predicting the prognosis.[37]
180TCGADESeq2 package
MultiMiR package
LC-MS/MS
Enrichr
Kaplan–Meier method
Log-rank test
hsa-mir-200b
CFL2
These regulatory circuit networks were associated with survival.[38]
97GEO
TCGA
GEPIA 2
TIMER
cBioPortal
GSEA
MethSurv
Kaplan–Meier plotter
GPR27GPR27 was useful for predicting prognosis and had a clear interaction with immune cells’ infiltration as well as their markers in patients with GC.[34]
441GEO (GPL6947, GSE84437)
TCGA-STAD
RNA-seq
DAVID, GO, and KEGG pathway enrichment analyses
PPI network
Cox regression modelsRIMS1, PRICKLE1, MCC, DCLK1, FLRT2, SLCO2A1, CDO1, GHR, CD109, SELP, UPK1B, CD36Thirteen mRNA-based risk score model implemented acceptably in discriminating the risk of GC prognosis.[44]
N.A.GEO (GSE57303, GSE62254)
TCGA-STAD
GDSC
ImmLnc
GSEA, GSVA
The CIBERSORT algorithm
GO and KEGG pathway enrichment analyses
Kaplan–Meier survival analysis Cox regression analysis
Immune-related lncRNA prognostic model
LASSO
SNHG5, LINC01270, CHKB. AS1, NUTM2A.AS1, MIR181A2HG, CCNT2.AS1, DLG3.AS1, LINC01134,etcAmong the 18 lncRNAs, the p-value of MIR181A2HG is the lowest, implying that it possesses good prediction activity.[40].
N.A.TGCA-STAD
Ensembl
CIBERSORT
Illumina HiSeqRNASeq platforms
EdgeR analysis
DESeq2 analysis
Cytoscape
GO and KEGG pathway enrichment analyses
PPI network
Kaplan–Meier survival analysis
Cox hazard model
LASSO
VCAN-AS1, AL139002.1, LINC00326, AC018781.1, C15orf54The five lncRNAs served as hub nodes in the ceRNA network.[45]
64GEO (GSE53137, GSE70880, GSE99417)
TCGA-STAD
Sva and limma package
miRDB, TargetScan, miRTarBase
GO and KEGG pathway enrichment analyses
PPI network
Kaplan–Meier analysisUCA1, HOTTIP, HMGA1P4Three lncRNAs may be involved in the development, and their prospective activities may be related to the prognosis of GC.[41]
337TCGA-STADGSEA-MSigDB database
GO and KEGG pathway enrichment analyses
Kaplan–Meier survival analysis
Log-rank test
Cox and Lasso regression analysis
AL353804.1, AC010719.1, TNFRSF10A-AS1, AC005586.1, AL355574.1, AC009948.1, AL161785.1These lncRNAs were enriched in the cell adhesion molecules and JAK-STAT3 signaling pathway.[43]
Abbreviations: GC, gastric cancer; TCGA, The Cancer Genome Atlas; GEO, Gene Expression Omnibus; scRNA-seq, single-cell RNA sequencing; GSRCC, gastric signet ring cell carcinoma; TIME, tumor immune microenvironment; TIMER, Tumor Immune Estimation Resource; GSEA, gene set enrichment analysis; GO, gene ontology; KEGG, The Kyoto Encyclopedia of Genes and Genomes; PD, programming death; DEM, differentially expressed miRNA; MS, mass spectrometry; LASSO, least absolute shrinkage, and selection operator; GDSC, Genomics of Drug Sensitivity in Cancer; lncRNAs, long non-coding RNAs; ceRNA, competing endogenous RNA; GSVA, Gene set variation analysis; PPI, protein-protein interaction; STAT3, signal transducer and activator of transcription 3.
Table 6. Biomarkers for therapeutic prediction of GC using bioinformatical analysis.
Table 6. Biomarkers for therapeutic prediction of GC using bioinformatical analysis.
Number of PatientsDatabasesAnalytical PlatformStatistical AnalysisGene Set IdentifiedCommentsRefs.
253GEO
(GSE662254, GSE26942)
TCGA
CCLE
Limma R package
GO pathway enrichment analyses
Cox regression analysis
Kaplan–Meier method Log-rank test
ND6, BRMS1, SRXN1Patients of the high-risk group should be treated with other chemotherapeutics to prevent unnecessary exposure to agents.[69]
393GC TCGAMyCancerGenome
CiViC
TARGET OncoKB
χ2 testPIK3CA mutation variantsDrugs identified here for therapeutic options are sorafenib, cabozantinib, regorafenib.[70]
206TCGAiBAQ algorithm
LC-MS/MS
KEGG pathway enrichment analysis
ssGSEA
PRM method
Two-sided Student’s t-test)
Kaplan–Meier survival curves
Cox analysis
THSD4, SRPX2, TGFBI, THBS1, LAMB2, COL4A1, COL6A5, FN1, GP1BA, ITGA4, THBS3, THBS4, BCL2, BCL2L1, CASP3, CASP7
CDKN2A
PI3K-AKT signaling pathway had potential relevance to the ECM proteins.[72]
804GEO
(GSE84437)
TCGA
GSEA
TCIA
SVA package
ConsensusClusterPlus package
ssGSEA algorithm
GO and KEGG enrichment pathways
PPI network
Cox analysis
Kaplan–Meier method
PLOD2, GLUT3The lactate score may be useful for predicting the prognosis and immune escape of GC.[71]
32GEO
(GSE1617297)
scRNA-seq
CellChat R package
UMAP algorithm
BCR/TCR analysis
t-test, Wilcoxon rank-sum test,CXCL13CXCL13 is an important regulator for the immune response of the GC TIME.[33]
47Human Metabolome DatabaseLC-MSt-testDeoxyribose 1-phosphate, S-lactoylglutathione, lysoPC (16:0), and O-arachidonoyl ethanolamineFour serum metabolites revealed potent divergences in patients with chemo-sensitive and chemo-resistant tumors.[76]
Abbreviations: GC, gastric cancer; TCGA, The Cancer Genome Atlas; GEO, Gene Expression Omnibus; CCLE, cancer cell line encyclopedia; TCIA, The Cancer Immunome Database; iBAQ, intensity-based absolute quantification; ssGSEA, single sample gene set enrichment analysis; PRM, parallel reaction monitoring; PI3K, phosphoinositide 3-kinase; GSEA, gene set enrichment analysis; GO, gene oncology; KEGG, The Kyoto Encyclopedia of Genes and Genomes; TIME, tumor immune microenvironment; LC-MS, liquid chromatography-mass spectrometry.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Matsuoka, T.; Yashiro, M. Bioinformatics Analysis and Validation of Potential Markers Associated with Prediction and Prognosis of Gastric Cancer. Int. J. Mol. Sci. 2024, 25, 5880. https://doi.org/10.3390/ijms25115880

AMA Style

Matsuoka T, Yashiro M. Bioinformatics Analysis and Validation of Potential Markers Associated with Prediction and Prognosis of Gastric Cancer. International Journal of Molecular Sciences. 2024; 25(11):5880. https://doi.org/10.3390/ijms25115880

Chicago/Turabian Style

Matsuoka, Tasuku, and Masakazu Yashiro. 2024. "Bioinformatics Analysis and Validation of Potential Markers Associated with Prediction and Prognosis of Gastric Cancer" International Journal of Molecular Sciences 25, no. 11: 5880. https://doi.org/10.3390/ijms25115880

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop