Open Access

Identification of putative drugs for gastric adenocarcinoma utilizing differentially expressed genes and connectivity map

  • Authors:
    • Zu‑Xuan Chen
    • Xiao‑Ping Zou
    • Huang‑Qun Yan
    • Rui Zhang
    • Jin‑Shu Pang
    • Xin‑Gan Qin
    • Rong‑Quan He
    • Jie Ma
    • Zhen‑Bo Feng
    • Gang Chen
    • Ting‑Qing Gan
  • View Affiliations

  • Published online on: December 13, 2018     https://doi.org/10.3892/mmr.2018.9758
  • Pages: 1004-1015
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric adenocarcinoma (GAC) is a challenging disease with dim prognosis even after surgery; hence, novel treatments for GAC are in urgent need. The aim of the present study was to explore new potential compounds interfering with the key pathways related to GAC progression. The differentially expressed genes (DEGs) between GAC and adjacent tissues were identified from The Cancer Genome Atlas (TCGA) and Genotype‑Tissue Expression (GTEx) database. Connectivity Map (CMap) was performed to screen candidate compounds for treating GAC. Subsequently, pathways affected by compounds were overlapped with those enriched by the DEGs to further identify compounds which had anti‑GAC potential. A total of 843 DEGs of GAC were identified. Via Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, 13 pathways were significantly enriched. Moreover, 78 compounds with markedly negative correlations with DEGs were revealed in CMap database (P<0.05 and Enrichment <0). Subpathways of cell cycle and p53 signaling pathways, and core genes of these compounds, cyclin B1 (CCNB1) and CDC6, were identified. This study further revealed seven compounds that may be effective against GAC; in particular methylbenzethonium chloride and alexidine have never yet been reported for GAC treatment. In brief, the candidate drugs identified in this study may provide new options to improve the treatment of patients with GAC. However, the biological effects of these drugs need further investigation.

Introduction

Globally, gastric cancer is the fifth leading cause of cancer and the third leading cause of death from cancer (1,2). In 2015, 679,100 new cases of gastric cancer were diagnosed in China, accounting for 15.8% of the total number of newly occurred cancer cases. In addition, gastric cancer resulted in 498,000 deaths, 17.7% of all cancer-related deaths, and the incidence of gastric cancer has been steadily increasing (3). Among these cases, gastric adenocarcinoma (GAC) accounts for 95% of all gastric cancer cases. Research indicates that, even after surgery, the outcome of GAC patients remains dim (46). Therefore, other novel treatments for GAC should be developed. The study of small-molecule drugs aiming at multiple protein pathways modulating tumor progression, invasion, and metastasis formation, has received much interest in recent years (79). The purpose of this study was to discover new, potential small-molecule drugs by using multiple online databases.

Connectivity Map (CMap) is one of the gene expression profile databases used to process the genetic data. CMap was developed by Lamb and his colleagues from Broad Institute of MIT, Whitehead Institute and Harvard Medical School, (Boston, MA, USA) (10). CMap utilizes the differential gene expression of human cells which are treated with small-molecule drugs, to construct a biological application database based on connection of small-molecule drugs, gene expression and different diseases. CMap allows scholars of drug development to take advantage of gene expression profiling data and, therefore, identify the drugs highly correlated with disease, infer the main chemical structure of most drug molecules, and summarize the mechanism of possible action of drug molecules.

To explore new drugs for GAC, based on the integrated subpathway analysis, we implemented an in silico method for the reuse of GAC drugs. First, we identified the differentially expressed genes (DEGs) between GAC and non-tumor tissues identified in The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases, and then determined the potential pathways affecting the progression of GAC. Next, CMap was used to verify the pathways of GAC affected by small-molecule treatment. Finally, small-molecule drugs that can target subpathways related to GAC were considered as potential new agents in the treatment of GAC (Fig. 1). The candidate drugs identified in our approach may provide a new direction for improving the treatment of patients with GAC.

Materials and methods

DEG analysis of GAC

Using the GEPIA online analysis website (http://gepia.cancer-pku.cn/), the expression data of mRNA of GAC in TCGA and GTEx databases were performed with the value of fold change (FC). Among these data, only the genes with logFC >2 and logFC <-2 were defined as DEGs, including upregulated and downregulated ones.

Enrichment analysis of DEGs

DEGs were performed with Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis with the WebGestalt database (http://www.webgestalt.org/). Also, pathway analysis was conducted by Gene List Analysis (http://www.pantherdb.org/) to obtain possible pathways during the development of GAC. Finally, we used the STRING database (https://string-db.org/) to analyze the protein-protein interaction (PPI) of the ultimate DEGs as previously reported (1116). In this study, GO outcomes were analyzed visibly with Cytoscape software (version 3.7.0, U.S. National Institute of General Medical Sciences (NIGMS), http://cytoscape.org/).

CMap for DEG analysis of drug molecule cures for GAC

The CMap database (https://portals.broadinstitute.org/CMap/) (build 02) contains over 7,000 gene expression profiles and 1,309 chemicals. To analyze this potential mechanism for the development of GAC, we first set up the files in query signature format for DEGs obtained from the TCGA (https://cancergenome.nih.gov/) and GTEx databases (https://gtexportal.org/home/). We then entered the CMap quick query interface to import the files of upregulated and downregulated genes and ran them with CMap analysis. In this way, we analyzed the drug molecules for the DEGs of GAC (17). The negatively related drugs (P<0.05 and Enrichment <0) for anti-GAC were then screened.

Correlation data between drug molecules and subpathways

The chip expression profiles of 1,309 drugs and the genes affected by the drugs using the CMap database were downloaded. Furthermore, we identified the subpathways that obtain significant enrichment for each small-molecule drug with the affected genes according to the method reported by a previous publication (18). Consistent with the reference, 196 small-molecular drugs and 104 subpathways were also achieved. The overlapped pathways between those from CMap and those enriched by DEGs were determined, which were identified as potential pathways related to both the treatment and pathogenesis of GAC. Finally, the drug-pathway network was constructed for GAC.

Results

Screening results of DEGs

Altogether, 843 DEGs in mRNA expression of GAC were obtained, which included 638 upregulated genes and 205 downregulated ones. The next analysis was based on this screening result.

Functional annotation, pathway enrichment and PPI network analysis

Through GO analysis, in the annotations of biological progress, the top three most significant processes were mitotic sister chromatid segregation, mitotic cell cycle and nuclear division. In the terms of cellular component, the top most significant annotations were extracellular space, chromosome, centromeric region and spindle. As for analysis of molecular function, the top three most significant functions were serine hydrolase activity, chemokine activity and serine-type peptidase activity (Table I and Fig. 2). KEGG pathway analysis indicated that DEGs were obviously centralized in 13 pathways, including cell cycle, protein digestion and absorption, Staphylococcus aureus infection, and the p53 signaling pathway (Table II and Fig. 3). From the PPI network analysis, we acquired the following hub genes: CCNB1, AURKA, CDC6, KIF11, OIP5, NCAPG, KIF23, DLGAP5 and NDC80 (nodes ≥100) (Fig. 4).

Table I.

Top 10 of the most significantly enriched GO terms.

Table I.

Top 10 of the most significantly enriched GO terms.

Pathway IDTermsGene countFDRP-value
BP
  GO:0000070Mitotic sister chromatid segregation3600
  GO:0000278Mitotic cell cycle11200
  GO:0000280Nuclear division7600
  GO:0000819Sister chromatid segregation4700
  GO:0007049Cell cycle14200
  GO:0007059Chromosome segregation5800
  GO:0007067Mitotic nuclear division6900
  GO:0008283Cell proliferation16500
  GO:0022402Cell cycle process12500
  GO:0042127Regulation of cell proliferation12700
CC
  GO:0005615Extracellular space12900
  GO:0000775Chromosome, centromeric region341.14E-132.22E-16
  GO:0005819Spindle421.52E-134.44E-16
  GO:0000793Condensed chromosome352.86E-131.11E-15
  GO:0098687Chromosomal region422.54E-121.23E-14
  GO:0000779Condensed chromosome, centromeric region252.88E-121.68E-14
  GO:0000776Kinetochore261.18E-118.04E-14
  GO:0000777Condensed chromosome kinetochore231.39E-111.20E-13
  GO:0009986Cell surface641.39E-111.21E-13
  GO:0005694Chromosome711.40E-101.36E-12
MF
  GO:0017171Serine hydrolase activity302.76E-071.77E-10
  GO:0008009Chemokine activity142.76E-073.88E-10
  GO:0008236Serine-type peptidase activity292.76E-075.77E-10
  GO:0004252Serine-type endopeptidase activity272.76E-076.04E-10
  GO:0042379Chemokine receptor binding153.08E-078.91E-10
  GO:0004175Endopeptidase activity423.08E-071.01E-09
  GO:0045236CXCR chemokine receptor binding94.33E-071.66E-09
  GO:0001664G-protein coupled receptor binding276.08E-052.66E-07
  GO:0032395MHC class II receptor activity67.59E-053.74E-07
  GO:0042802Identical protein binding821.18E-046.44E-07

[i] GO, Gene Ontology; BP, biological progress; CC, cellular component; MF, molecular function; FDR, false discovery rate.

Table II.

Significantly enriched KEGG pathway.

Table II.

Significantly enriched KEGG pathway.

Pathway IDTermsGene countFDRP-value
hsa04110Cell cycle262.83E-089.34E-11
hsa04974Protein digestion and absorption171.33E-048.80E-07
hsa05150Staphylococcus aureus infection129.58E-049.49E-06
hsa04115p53 signaling pathway131.35E-031.79E-05
hsa05140Leishmaniasis129.11E-031.50E-04
hsa05323Rheumatoid arthritis131.40E-023.07E-04
hsa04610Complement and coagulation cascades121.40E-023.24E-04
hsa05416Viral myocarditis101.56E-024.13E-04
hsa05310Asthma71.73E-025.12E-04
hsa05164Influenza A184.47E-021.63E-03
hsa04512ECM-receptor interaction114.47E-021.64E-03
hsa04060Cytokine-cytokine receptor interaction244.47E-021.77E-03
hsa04640Hematopoietic cell lineage124.86E-022.09E-03

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes; FDR, false discovery rate.

CMap analysis to achieve potential compounds for GAC

The 843 DEGs of GAC mentioned above led to 78 compounds by CMap (Table III) when P<0.05 and Enrichment <0.

Table III.

CMap compounds matched by the DEGs of gastric adenocarcinoma.

Table III.

CMap compounds matched by the DEGs of gastric adenocarcinoma.

RankCMap nameCell lineNEnrichmentP-valueSpecificityPercent non-null
1 PhenoxybenzamineMCF73−0.98400100
2VorinostatMCF77−0.84400.1262100
3Trichostatin APC355−0.70500.114996
4Trichostatin AMCF792−0.5900.188188
5Trichostatin AHL6034−0.46500.194652
6LY-294002MCF734−0.45400.162570
7ResveratrolMCF76−0.8650.000020.0082100
8AlexidinePC32−0.9960.000040100
915-Delta prostaglandin J2MCF78−0.6950.000180.041487
10MeticranePC32−0.9910.000260100
11AstemizolePC32−0.990.000260.0192100
12ThiostreptonMCF72−0.9730.001410.0283100
13ClemizolePC32−0.9730.001410100
14SulconazoleMCF72−0.9730.001570100
15MefloquinePC32−0.9710.001670.0431100
16MG-262PC32−0.9680.002230.0738100
17CloperastinePC32−0.9680.002230.0149100
18ThioridazinePC35−0.7360.00270.102100
19MethotrexateMCF73−0.8770.003790.0853100
20Valproic acidHL6014−0.4480.004030.288364
21CloperastineMCF73−0.8730.004150.0196100
22 FludroxycortidePC32−0.9540.004530.0171100
23PyrantelPC32−0.9460.006440.0144100
24ThioguanosineMCF72−0.9450.006580.0455100
25 6-Bromoindirubin-3′-oxime methylbenzethoniumPC34−0.7550.007320.0498100
26ChloridePC32−0.9390.007670.0598100
27ChlorpromazinePC34−0.7490.00790.0168100
28VorinostatHL603−0.8390.008370.1705100
29VitexinMCF72−0.9360.008610.0051100
30AcetazolamideMCF72−0.9310.009840100
31PyrviniumMCF74−0.7310.01050.1304100
325224221MCF72−0.9270.010970.1429100
33Methacholine chlorideMCF72−0.9240.011810.0278100
34CortisoneMCF72−0.9210.012620.0117100
35CarbacholMCF72−0.9190.013180.0058100
36ClotrimazoleMCF73−0.8070.014440.0556100
37DipyridamoleMCF73−0.7990.016710.04100
38AbamectinMCF72−0.9070.017460.05100
39LY-294002PC312−0.4230.018020.366966
40TroglitazonePC34−0.6960.018040.1159100
41LuteolinMCF72−0.9040.018390.0476100
42 HydroflumethiazideMCF72−0.9020.019130.0601100
43 HomochlorcyclizineMCF72−0.8980.020660.0968100
44GemfibrozilPC32−0.8960.021670.0208100
45Withaferin APC32−0.8940.022230.0917100
46TanespimycinPC312−0.4140.022390.338258
47 ProchlorperazineMCF79−0.4720.02310.189266
48CiclosporinMCF74−0.6790.023490.057675
49DisulfiramPC32−0.8910.023820.0667100
50ProcainePC32−0.890.0240.0294100
510173570-0000PC34−0.6770.024070.134975
52TretinoinMCF713−0.3950.025310.365561
53FluphenazinePC33−0.7690.025340.1026100
54LoperamideMCF73−0.7670.0260.087100
55DilazepPC32−0.8860.026120.0784100
56 TrifluoperazinePC33−0.7650.026560.1379100
57 3-AcetylcoumarinMCF73−0.7640.026920.022100
58FlunarizineMCF72−0.8840.027120.068100
59SulfaguanidinePC32−0.8780.029720.0202100
60EthaverineMCF72−0.8780.030040.0133100
61AmiodaroneMCF73−0.7540.030430.1039100
62PicotamidePC32−0.8750.031270.0162100
63FelodipineMCF75−0.5940.03180.137680
64Prestwick-1084MCF72−0.8730.032010.0545100
65MonobenzoneMCF72−0.8710.033060.0548100
66PioglitazonePC35−0.5860.035850.343660
67LevocabastineMCF72−0.8660.036260.0615100
68NoretynodrelMCF72−0.8650.036280.0822100
69 TrifluoperazineMCF79−0.4480.036550.230855
7015-Delta prostaglandin J2HL603−0.7380.036840.1429100
71EtoposideMCF72−0.8640.037120.1100
72BufexamacMCF72−0.8630.03760.0556100
730179445-0000PC34−0.6440.038530.068575
7415-Delta prostaglandin J2PC33−0.7340.038560.1507100
75MinaprinePC32−0.8580.040080.031100
76OxymetazolinePC32−0.8550.041810.0345100
77NortriptylineMCF72−0.8520.043380.0901100
78CP-690334-01MCF74−0.6330.044180.102750
79SB-203580PC32−0.850.045150.0464100
80ScriptaidPC32−0.8490.045370.1596100
81EsculetinMCF72−0.8480.046090.0671100
82FluspirileneMCF72−0.8480.04640.1748100
83SulfadoxineMCF72−0.8450.048290.0481100
84MonordenPC35−0.5620.049320.10660
85IvermectinMCF72−0.8430.049370.1404100
86NorethisteroneMCF72−0.8420.049940.0263100

[i] CMap, Connectivity Map; DEGs, differentially expressed genes. N, number of all instances of the same perturbagen made in the same cell line. A total of 78 compounds were included, among which, four compounds were administered to two different cell lines and two compounds were administered to three different cell lines. Thus, there are 86 rows in the table.

Intersection of small-molecule drug correlative pathways and KEGG pathways

According to a previous method (18), we performed subpathway analysis and obtained 104 subpathways. After integrating these 104 subpathways with 13 KEGG pathways generated by the DEGs, two pathways related to anti-GAC drug molecules were finally achieved (Table IV and Fig. 5), including cell cycle and p53 signaling pathways. These two pathways were related to 32 genes and seven CMap small-molecule drugs. The genes involved in these two KEGG pathway were CDKN2A, DBF4, CHEK1, ORC6, SFN, MAD2L1, MCM2, MCM4, MCM5, PCNA, PLK1, CCND1, BUB1, BUB1B, TTK, CDC45, CCNA2, CCNB1, PKMYT1, CCNB2, PTTG1, ESPL1, CDK1, CDC6, CDC20, CDC25C, IGFBP3, GTSE1, SERPINB5, RPRM, RRM2 and BID. The PPI analysis with the above 32 genes demonstrated two hub genes (CCNB1 and CDC6). The seven CMap small-molecule drugs were troglitazone, methylbenzethonium chloride, thiostrepton, alexidine, vorinostat, methotrexate and etoposide (Fig. 6).

Table IV.

CMap negatively correlated compounds matched by pathway.

Table IV.

CMap negatively correlated compounds matched by pathway.

Drug namePathway nameSubpathway ID
Alexidinep53 signaling pathwaypath:04115_2; path:04115_1; path:04115_7
MefloquineToll-like receptor signaling pathwaypath:04620_17; path:04620_18; path:04620_22; path:04620_9
MefloquineSteroid hormone biosynthesispath:00140_3; path:00140_19; path:00140_16; path:00140_8
AstemizoleToll-like receptor signaling pathwaypath:04620_12; path:04620_9; path:04620_18; path:04620_17
Thiostreptonp53 signaling pathwaypath:04115_1
Methotrexatep53 signaling pathwaypath:04115_7; path:04115_1; path:04115_4; path:04115_3; path:04115_2
SulconazoleMetabolism of xenobiotics by cytochrome P450path:00980_3
ResveratrolTryptophan metabolismpath:00380_5
ResveratrolToxoplasmosispath:05145_18
ThioguanosineSteroid hormone biosynthesispath:00140_7; path:00140_8
MG-262Steroid hormone biosynthesispath:00140_1; path:00140_9; path:00140_8; path:00140_6; path:00140_5
Methylbenzethonium chloridep53 signaling pathwaypath:04115_1
MonobenzoneMAPK signaling pathwaypath:04010_30
TrifluoperazineProtein processing in endoplasmic reticulumpath:04141_18: path:04141_1
5224221Steroid hormone biosynthesispath:00140_18; path:00140_27; path:00140_9; path:00140_8; path:00140_4
VitexinSteroid hormone biosynthesispath:00140_19
DisulfiramProtein processing in endoplasmic reticulumpath:04141_1
ThioridazinePathways in cancerpath:05200_29; path:05200_18; path:05200_11
Vorinostatp53 signaling pathwaypath:04115_1; path:04115_2; path:04115_4; path:04115_3
Etoposidep53 signaling pathwaypath:04115_7; path:04115_1; path:04115_3
Withaferin ASteroid hormone biosynthesispath:00140_25; path:00140_5; path:00140_10; path:00140_4
PyrviniumSteroid hormone biosynthesispath:00140_6; path:00140_16; path:00140_19; path:00140_17; path:00140_18; path:00140_4
ScriptaidSteroid hormone biosynthesispath:00140_9; path:00140_6; path:00140_17; path:00140_16; path:00140_5; path:00140_1
Trichostatin ASteroid hormone biosynthesispath:00140_10; path:00140_19; path:00140_6; path:00140_8; path:00140_9
0173570-0000Steroid hormone biosynthesispath:00140_16; path:00140_4; path:00140_17; path:00140_3; path:00140_6; path:00140_10; path:00140_18; path:00140_13; path:00140_7; path:00140_8
TroglitazoneCell cyclepath:04110_17
ProchlorperazineProtein processing in endoplasmic reticulumpath:04141_1
LY-294002Steroid hormone biosynthesispath:00140_6; path:00140_27
TanespimycinMAPK signaling pathwaypath:04010_15
MonordenSteroid hormone biosynthesispath:00140_3; path:00140_7; path:00140_18

[i] CMap, connectivity map.

Expression levels of CCNB1 and CDC6 mRNA in GAC tissues

The expression levels of CCNB1 and CDC6 mRNA in GACs were queried from GEPIA database (http://gepia.cancer-pku.cn/). The results showed that the two genes were both highly expressed in GAC tissues compared to non-cancerous gastric tissues (Fig. 7).

Verification of predicting small-molecule drugs of GAC with online literature retrieval

Using PubMed, we identified studies that investigated the effect of relevant drugs on GAC. We found 268 articles related to the effect of methotrexate on GAC, 403 articles related to etoposide, and 17 articles related to troglitazone, which is a diabetes drug that may inhibit GAC. Nine studies concerned vorinostat and three studies were related to thiostrepton. Most importantly, methylbenzethonium chloride and alexidine have never been addressed in the literature of GAC.

Discussion

In the present study, we identified DEGs of GAC and found several pathways and hub genes that may play a critical role in the pathogenesis and development of GAC. Also, through the connectivity mapping approach, some known compounds were found to share similar pathways of those generated from the DEGs of GAC, including methotrexate, etoposide, troglitazone, thiostrepton, vorinostat, methylbenzethonium chloride and alexidine. The findings from the present study suggest that methylbenzethonium chloride and alexidine could act as novel potential drugs for the treatment of GAC and warrant further investigation, as they have never been tested previously.

The CMap database reveals the connection between disease, genes and drugs, using gene expression data and the ‘similarity’ concept with a small-molecular compound or the gene expression spectrum of the drug as the core (19). CMap database provides a unique method for drug development through comparison to filter candidate compounds curing diseases, and it has been adopted by several scholars (20,21). For instance, Xiao et al used gene expression profile chip technology and the CMap database to study molecular mechanisms of Hirschsprung disease (HD) and potential drugs. They found differences in the neuronal developmental disorders of HD genes and signaling pathways, and discovered that some compounds may offset the damage of HD development (22).

In this study, the DEGs between GAC and adjacent tissues were compared with the expression profiles in CMap to identify negatively correlative compounds that are potential compounds for GAC. Among the candidate compounds determined in the present study, two compounds (alexidine and methylbenzethonium) are particularly important. Alexidine is an antimicrobial agent with high affinity for bacteria, which can be used in the root canal irrigation solution of oral treatment (23). Feng et al, using high-throughput drug screening tests, identified that alexidine is an antitumor drug that can inhibit cytokines and growth factors necessary for multiple myeloma (24). Meanwhile, methylbenzethonium chloride, a broad spectrum antibiotic, was found to be able to specifically induce apoptosis in undifferentiated embryonic stem cells of mice (25). The effect could be applied to prevent reoccurrence of the tumor after stem cell transplantation therapy. Methylbenzethonium chloride may become another novel anticancer agent (25).

The present study showed that alexidine had the lowest connectivity score (−0.996), indicating a highly negative correlation with the DEGs of GAC. The connectivity score of methylbenzethonium chloride also suggests that it has the capacity to inhibit the growth of GAC. In addition, this study predicted that both alexidine and methylbenzethonium chloride can play a vital role in inhibiting GAC by regulating the p53 signaling pathway. Previous studies have shown that the p53 signaling pathway regulates various cellular functions, including apoptosis, induction of aging, and inhibition of cell growth, migration and invasion (2628). However, the specific molecular mechanisms of alexidine and methylbenzethonium chloride for antitumor activity need to be further explored.

Five other compounds achieved in the present study have been mentioned in other studies. Troglitazone hinders BGC-823 GAC cell proliferation and promotes its apoptosis by inducing expression of the non-steroidal anti-inflammatory drug-activated gene (NAG) (29). In addition, thiostrepton was found to reverse drug resistance in GAC by inhibiting the forkhead box transcription factor 1 (FOXM1) (30). Vorinostat (31), methotrexate (32) and etoposide (33) are proven to inhibit the proliferation of GAC cells. This evidence indicates that the predictive method in this study is convincing and worth being used for drug exploration.

In this study, we used bioinformatic methods to screen differentially expressing potential genetic biomarkers based on RNA-seq data. The results of pathway enrichment analysis indicated 13 pathways which were evidently enriched with DEGs, including the cell cycle, protein digestion and absorption, Staphylococcus aureus infection and the p53 signaling pathway. In addition, these DEGs were analyzed with CMap and subpathways, and two (cell cycle and p53 signaling pathway) were found to be closely related to the treatment potential and occurrence of GAC. CCNB1 and CDC6 in these pathways were also hub genes in the PPI network.

The clinical role of these hub genes was analyzed also based on publicly available RNA-seq data, and it was found that CCNB1 was upregulated in patients with GAC. CCNB1 is a member of the cell cycle protein B family; it is a regulatory protein involved in mitosis, mostly expressed in the G2/M period, and plays a significant role in the S-to-G2/M phases (34). Therefore, overexpression of CCNB1 in GAC leads to chaos in the cell cycle, mitosis promotion and cell proliferation. Previous research has shown that silencing of CDKN3 stimulates cell cycle arrest by reducing the expression of CDK2, CDC25, CCNB1 and CCNB2 in human GAC cells, thus, inhibits the proliferation of tumor cells (35). It was found in vivo that dipalmitoyl phosphatidic acid could dramatically inhibit the growth of tumors in a mouse subcutaneous tumor model, and suppress cell proliferation and angiogenesis in triple-negative breast cancer. The suppressing effect was mediated partly due to reduction in the expression of CCNB1 (36). Therefore, CCNB1 may be an important target gene in the treatment of GAC, and the present study predicted that compounds aimed at this target gene may be reasonable and effective in treating GAC. Recent studies have shown that knockdown of CDC6 expression levels can interfere with the cell cycle and inhibit the proliferation of prostate and ovarian cancer cells (37,38). This evidence suggests that CDC6 may also be a potential biomarker for GAC therapy.

The present study comprehensively analyzed the possible mechanism of treating GAC by data mining in the public gene chip databases and bioinformatic analyses. We discovered cell cycle and p53 signaling pathways and key gene targets CCNB1 and CDC6 as potential targets of GAC treatment. We further predicted that seven known compounds may be effective in curing GAC, including methylbenzethonium chloride and alexidine, which have never been previously reported to treat GAC. However, several limitations should be admitted. Firstly, the current findings were based on in silico methods and validations are certainly needed. Secondly, CMap did not cover GAC cell lines and only provided general DEGs post treatment of existing drugs. The overlapping pathways of DEGs from TCGA and pathways from Cmap also need to be confirmed. Thirdly, the precise mechanism of the drugs we recommended remains to be investigated. Hence, further clinical, in vitro and in vivo experiments are needed to verify the definite effects and molecular mechanism of the potential drugs on GAC.

Acknowledgements

Not applicable.

Funding

The present study was supported by a fund from the Promoting Project of Basic Capacity for Young and Middle-Aged University Teachers in Guangxi, China (KY2016YB077).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

ZXC, XPZ, HQY, RZ and JSP analyzed and interpreted the data and wrote the draft of the manuscript. XGQ, RQH, JM, ZBF, GC and TQG conceived and designed the study, supervised the data mining, corrected and revised the draft. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

GAC

gastric adenocarcinoma

CMap

connectivity map

TCGA

The Cancer Genome Atlas

DEGs

differentially expressed genes

KEGG

Kyoto Encyclopedia of Genes and Genomes

FC

fold change

GO

Gene Ontology

PPI

protein-protein interaction

HD

Hirschsprung disease

NAG

non-steroidal anti-inflammatory drug-activated gene

FOXM1

forkhead box transcription factor 1

References

1 

Wippel HH, Santos MDM, Clasen MA, Kurt LU, Nogueira FCS, Carvalho CE, McCormick TM, Neto GPB, Alves LR, da Gloria da Costa Carvalho M, et al: Comparing intestinal versus diffuse gastric cancer using a PEFF-oriented proteomic pipeline. J Proteomics. 171:63–72. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Li W, Song D, Li H, Liang L, Zhao N and Liu T: Reduction in peripheral CD19+CD24hCD27+ B cell frequency predicts favourable clinical course in XELOX-treated patients with advanced gastric cancer. Cell Physiol Biochem. 41:2045–2052. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Costa NR, Gil da Costa RM and Medeiros R: A viral map of gastrointestinal cancers. Life Sci. 199:188–200. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Coburn N, Cosby R, Klein L, Knight G, Malthaner R, Mamazza J, Mercer CD and Ringash J: Staging and surgical approaches in gastric cancer: A systematic review. Cancer Treat Rev. 63:104–115. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Chen W, Zhou H, Ye L and Zhan B: Overexpression of SULT2B1b promotes angiogenesis in human gastric cancer. Cell Physiol Biochem. 38:1040–1054. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Van Cutsem E, Sagaert X, Topal B, Haustermans K and Prenen H: Gastric cancer. Lancet. 388:2654–2664. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Zheng H, Hong H, Zhang L, Cai X, Hu M, Cai Y, Zhou B, Lin J, Zhao C and Hu W: Nifuratel, a novel STAT3 inhibitor with potent activity against human gastric cancer cells. Cancer Manag Res. 9:565–572. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Pavlakis N, Sjoquist KM, Martin AJ, Tsobanis E, Yip S, Kang YK, Bang YJ, Alcindor T, O'Callaghan CJ, Burnell MJ, et al: Regorafenib for the treatment of advanced gastric cancer (INTEGRATE): A multinational placebo-controlled phase II trial. J Clin Oncol. 34:2728–2735. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Belizário JE, Sangiuliano BA, Perez-Sosa M, Neyra JM and Moreira DF: Using pharmacogenomic databases for discovering patient-target genes and small molecule candidates to cancer therapy. Front Pharmacol. 7:3122016. View Article : Google Scholar : PubMed/NCBI

11 

Liang L, Zeng JH, Wang JY, He RQ, Ma J, Chen G, Cai XY and Hu XH: Down-regulation of miR-26a-5p in hepatocellular carcinoma: A qRT-PCR and bioinformatics study. Pathol Res Pract. 213:1494–1509. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Xiong DD, Lv J, Wei KL, Feng ZB, Chen JT, Liu KC, Chen G and Luo DZ: A nine-miRNA signature as a potential diagnostic marker for breast carcinoma: An integrated study of 1,110 cases. Oncol Rep. 37:3297–3304. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Zhang Y, Huang JC, Cai KT, Yu XB, Chen YR, Pan WY, He ZL, Lv J, Feng ZB and Chen G: Long non-coding RNA HOTTIP promotes hepatocellular carcinoma tumorigenesis and development: A comprehensive investigation based on bioinformatics, qRT-PCR and meta-analysis of 393 cases. Int J Oncol. 51:1705–1721. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Zhang Y, Dang YW, Wang X, Yang X, Zhang R, Lv ZL and Chen G: Comprehensive analysis of long non-coding RNA PVT1 gene interaction regulatory network in hepatocellular carcinoma using gene microarray and bioinformatics. Am J Transl Res. 9:3904–3917. 2017.PubMed/NCBI

15 

He RQ, Li XJ, Liang L, Xie Y, Luo DZ, Ma J, Peng ZG, Hu XH and Chen G: The suppressive role of miR-542-5p in NSCLC: The evidence from clinical data and in vivo validation using a chick chorioallantoic membrane model. BMC Cancer. 17:6552017. View Article : Google Scholar : PubMed/NCBI

16 

Gao L, Li SH, Tian YX, Zhu QQ, Chen G, Pang YY and Hu XH: Role of downregulated miR-133a-3p expression in bladder cancer: A bioinformatics study. Onco Targets Ther. 10:3667–3683. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Dang YW, Lin P, Liu LM, He RQ, Zhang LJ, Peng ZG, Li XJ and Chen G: In silico analysis of the potential mechanism of telocinobufagin on breast cancer MCF-7 cells. Pathol Res Pract. 214:631–643. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Wang J, Li M, Wang Y and Liu X: Integrating subpathway analysis to identify candidate agents for hepatocellular carcinoma. Onco Targets Ther. 9:1221–1230. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Musa A, Ghoraie LS, Zhang SD, Glazko G, Yli-Harja O, Dehmer M, Haibe-Kains B and Emmert-Streib F: A review of connectivity map and computational approaches in pharmacogenomics. Brief Bioinform. 19:506–523. 2018.PubMed/NCBI

20 

Brum AM, van de Peppel J, Nguyen L, Aliev A, Schreuders-Koedam M, Gajadien T, van der Leije CS, van Kerkwijk A, Eijken M, van Leeuwen JPTM and van der Eerden BCJ: Using the connectivity map to discover compounds influencing human osteoblast differentiation. J Cell Physiol. 233:4895–4906. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Busby J, Murray L, Mills K, Zhang SD, Liberante F and Cardwell CR: A combined connectivity mapping and pharmacoepidemiology approach to identify existing medications with breast cancer causing or preventing properties. Pharmacoepidemiol Drug Saf. 27:78–86. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Xiao SJ, Zhu XC, Deng H, Zhou WP, Yang WY, Yuan LK, Zhang JY, Tian S, Xu L, Zhang L and Xia HM: Gene expression profiling coupled with connectivity map database mining reveals potential therapeutic drugs for Hirschsprung disease. J Pediatr Surg. 53:1716–1721. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Varadan P, Ganesh A, Konindala R, Nagendrababu V, Ashok R and Deivanayagam K: Comparison of the antibacterial efficacy of alexidine and chlorhexidine against enterococcus faecalis: An in vitro study. Cureus. 9:e18052017.PubMed/NCBI

24 

Feng R, Rios JA, Onishi T, Lokshin A, Gorelik E and Lentzsch S: Cell-based and cytokine-directed chemical screen to identify potential anti-multiple myeloma agents. Leuk Res. 34:917–924. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Conesa C, Doss MX, Antzelevitch C, Sachinidis A, Sancho J and Carrodeguas JA: Identification of specific pluripotent stem cell death-inducing small molecules by chemical screening. Stem Cell Rev. 8:116–127. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Huang H, Han Y, Yang X, Li M, Zhu R, Hu J, Zhang X, Wei R, Li K and Gao R: HNRNPK inhibits gastric cancer cell proliferation through p53/p21/CCND1 pathway. Oncotarget. 8:103364–103374. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Fu H, Wang C, Yang D, Wei Z, Xu J, Hu Z, Zhang Y, Wang W, Yan R and Cai Q: Curcumin regulates proliferation, autophagy, and apoptosis in gastric cancer cells by affecting PI3K and P53 signaling. J Cell Physiol. 233:4634–4642. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Sun M, Si G, Sun HS and Si FC: Inhibition of CREPT restrains gastric cancer growth by regulation of cycle arrest, migration and apoptosis via ROS-regulated p53 pathway. Biochem Biophys Res Commun. 496:1183–1190. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Wang C, Wang J and Bai P: Troglitazone induces apoptosis in gastric cancer cells through the NAG-1 pathway. Mol Med Rep. 4:93–97. 2011.PubMed/NCBI

30 

Li X, Qiu W, Liu B, Yao R, Liu S, Yao Y and Liang J: Forkhead box transcription factor 1 expression in gastric cancer: FOXM1 is a poor prognostic factor and mediates resistance to docetaxel. J Transl Med. 11:2042013. View Article : Google Scholar : PubMed/NCBI

31 

Yoo C, Ryu MH, Na YS, Ryoo BY, Lee CW, Maeng J, Kim SY, Koo DH, Park I and Kang YK: Phase I and pharmacodynamic study of vorinostat combined with capecitabine and cisplatin as first-line chemotherapy in advanced gastric cancer. Invest New Drugs. 32:271–278. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Ebrahimifar M, Hasanzadegan Roudsari M, Kazemi SM, Ebrahimi Shahmabadi H, Kanaani L, Alavi SA and Izadi Vasfi M: Enhancing effects of curcumin on cytotoxicity of paclitaxel, methotrexate and vincristine in gastric cancer cells. Asian Pac J Cancer Prev. 18:65–68. 2017.PubMed/NCBI

33 

Duo-Ji MM, Ci-Ren BS, Long ZW, Zhang XH and Luo DL: Short-term efficacy of different chemotherapy regimens in the treatment of advanced gastric cancer: A network meta-analysis. Oncotarget. 8:37896–37911. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Shi Q, Wang W, Jia Z, Chen P, Ma K and Zhou C: ISL1, a novel regulator of CCNB1, CCNB2 and c-MYC genes, promotes gastric cancer cell proliferation and tumor growth. Oncotarget. 7:36489–36500. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Li Y, Ji S, Fu LY, Jiang T, Wu D and Meng FD: Knockdown of cyclin-dependent kinase inhibitor 3 inhibits proliferation and invasion in human gastric cancer cells. Oncol Res. 25:721–731. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Zhang QQ, Chen J, Zhou DL, Duan YF, Qi CL, Li JC, He XD, Zhang M, Yang YX and Wang L: Dipalmitoylphosphatidic acid inhibits tumor growth in triple-negative breast cancer. Int J Biol Sci. 13:471–479. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Karanika S, Karantanos T, Li L, Wang J, Park S, Yang G, Zuo X, Song JH, Maity SN, Manyam GC, et al: Targeting DNA damage response in prostate cancer by inhibiting androgen receptor-CDC6-ATR-Chk1 signaling. Cell Rep. 18:1970–1981. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Deng Y, Jiang L, Wang Y, Xi Q, Zhong J, Liu J, Yang S, Liu R, Wang J, Huang M, et al: High expression of CDC6 is associated with accelerated cell proliferation and poor prognosis of epithelial ovarian cancer. Pathol Res Pract. 212:239–246. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 19 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen ZX, Zou XP, Yan HQ, Zhang R, Pang JS, Qin XG, He RQ, Ma J, Feng ZB, Chen G, Chen G, et al: Identification of putative drugs for gastric adenocarcinoma utilizing differentially expressed genes and connectivity map. Mol Med Rep 19: 1004-1015, 2019
APA
Chen, Z., Zou, X., Yan, H., Zhang, R., Pang, J., Qin, X. ... Gan, T. (2019). Identification of putative drugs for gastric adenocarcinoma utilizing differentially expressed genes and connectivity map. Molecular Medicine Reports, 19, 1004-1015. https://doi.org/10.3892/mmr.2018.9758
MLA
Chen, Z., Zou, X., Yan, H., Zhang, R., Pang, J., Qin, X., He, R., Ma, J., Feng, Z., Chen, G., Gan, T."Identification of putative drugs for gastric adenocarcinoma utilizing differentially expressed genes and connectivity map". Molecular Medicine Reports 19.2 (2019): 1004-1015.
Chicago
Chen, Z., Zou, X., Yan, H., Zhang, R., Pang, J., Qin, X., He, R., Ma, J., Feng, Z., Chen, G., Gan, T."Identification of putative drugs for gastric adenocarcinoma utilizing differentially expressed genes and connectivity map". Molecular Medicine Reports 19, no. 2 (2019): 1004-1015. https://doi.org/10.3892/mmr.2018.9758