Open Access

Evaluation of the target genes of arsenic trioxide in pancreatic cancer by bioinformatics analysis

  • Authors:
    • Cong‑Ya Zhou
    • Liu‑Yun Gong
    • Rong Liao
    • Ning‑Na Weng
    • Yao‑Yue Feng
    • Yi‑Ping Dong
    • Hong Zhu
    • Ya‑Qin Zhao
    • Yuan‑Yuan Zhang
    • Qing Zhu
    • Su‑Xia Han
  • View Affiliations

  • Published online on: September 19, 2019     https://doi.org/10.3892/ol.2019.10889
  • Pages: 5163-5172
  • Copyright: © Zhou 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

The aim of the present study was to evaluate the potential network of arsenic trioxide (ATO) target genes in pancreatic cancer. The DrugBank, STITCH, cBioPortal, Kaplan‑Meier plotter and Oncomine websites were used to analyze the association of ATO and its target genes with pancreatic cancer. Initially, 19 ATO target genes were identified, along with their associated protein‑protein interaction networks and Kyoto Encyclopedia of Genes and Genomes pathways. ATO was found to be associated with multiple types of cancer, and the most common solid cancer was pancreatic cancer. A total of 6 ATO target genes (namely AKT1, CCND1, CDKN2A, IKBKB, MAPK1 and MAPK3) were found to be associated with pancreatic cancer. Next, the mutation information of the 6 ATO target genes in pancreatic cancer was collected. A total of 20 ATO interacting genes were identified, which were mainly involved in hepatitis B, prostate cancer, pathways in cancer, glioma and chronic myeloid leukemia. Finally, the genes CCND1 and MAPK1 were detected to be prognostic factors in patients with pancreatic cancer. In conclusion, bioinformatics analysis may help elucidate the molecular mechanisms underlying the involvement of ATO in pancreatic cancer, enabling more effective treatment of this disease.

Introduction

Pancreatic cancer is one of the most aggressive malignancies worldwide, with a mortality rate that is nearly equal to its incidence rate (1). Pancreatic cancer is expected to become the second leading cause of cancer-associated mortality in the US by 2030 (2). Despite advances in diagnostic and therapeutic strategies over the past years, the prognosis of patients with pancreatic cancer remains unsatisfactory, with a 5-year survival rate of <8.5% (3). Pancreatic cancer has become one of the major public health concerns worldwide. In order to improve the treatment of pancreatic cancer, it is necessary to elucidate the pathological mechanism underlying its development, identify a drug-based framework and investigate the functional mechanisms involved.

Arsenic trioxide (ATO) has been reported to be an effective therapeutic agent for acute promyelocytic leukemia (APL) (4). It has been reported that ATO can induce molecular remission, as well as prolong the overall survival of patients with APL (5,6). Combined treatment with retinoic acid (RA) and ATO has been demonstrated to be curative for the majority of patients with APL (79). Furthermore, an increasing number of studies have proven the anti-carcinogenic properties of ATO in different tumors, including in gastric cancer (10), lymphoma (11), bladder cancer (12), head and neck cancer (13), and ovarian cancer (14). Although it has been reported that ATO may inhibit the progression of pancreatic cancer (15), the potential targets and molecular mechanisms of action of ATO remain unclear.

In recent years, with the continuous improvement and development of high-throughput sequencing technology, multi-omics research (such as genomics, transcriptomics, proteomics and metabolomics) has gradually improved and has been used to identify disease-associated genes, which may uncover the molecular mechanisms underlying disease development and enable effective treatment. DrugBank is a richly annotated web-based bioinformatics tool that contains multiple drug data with comprehensive drug targets information (16). It provides detailed, step-by-step information on the results of previous studies on drugs, drug targets and drug effects, allowing researchers to search, view and export data, text and image (17). Furthermore, Search Tool for Interactions of Chemicals (STITCH) is an online database of the interaction networks of chemicals and proteins (18).

In the present study, DrugBank and STITCH were used to identify the targets of ATO, in order to construct an ATO-target interaction network. Protein-protein interaction (PPI) network construction and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis demonstrated that ATO is associated with several types of malignancies, including pancreatic cancer. In total, 6 genes (AKT1, CCND1, CDKN2A, IKBKB, MAPK1 and MAPK3) were identified as the target genes of ATO in pancreatic cancer. The interconnection of these 6 ATO target genes in pancreatic cancer was then analyzed using the Search Tool for the Retrieval of Interacting Genes (STRING) database. The cBioPortal website was further used to analyze the genomics data of these 6 target genes, while online Kaplan-Meier (K-M) plotter was used to analyze the prognosis of pancreatic cancer. In addition, the Oncomine website was used to reveal the expression levels of ATO target genes in pancreatic cancer tissues. Taken together, the findings of the bioinformatics analysis performed in the present study may help determine the gene targets of ATO and provide new therapeutic approaches for pancreatic cancer.

Materials and methods

Search for target genes of ATO

DrugBank, version 5.0 (https://www.drugbank.ca), is a richly annotated web-based bioinformatics tool that contains multiple drug data with comprehensive drug target information. In the present study, the DrugBank 5.0 and STITCH (http://stitch.embl.de/) databases were used to identify the ATO-target interactions in order to obtain an ATO-target network. The information on ATO target genes was then used to construct a visualization table and analyzed by STRING for further analysis. The search term ‘arsenic trioxide’ was used to search the DrugBank 5.0 and STITCH databases, and the targets of ATO in these two websites were identified. A total of 19 targets were recorded and included in subsequent analyses.

KEGG pathway and Gene Ontology analyses, and PPI network construction of ATO targets

The STRING database (https://string-db.org/) gathers and provides known and predicted PPI data for a large number of organisms, including humans (19,20). Cytoscape is a general-purpose software platform used for integrating bimolecular interaction networks (21). CluePedia, a plugin of Cytoscape software, can be used for searching potential genes associated with the reported signaling pathway by calculating linear and non-linear statistical dependencies from experimental data (22). In the present study, the KEGG pathway and Gene Ontology (GO) analyses were conducted using the STRING database. Subsequently, the STRING website and Cytoscape software were utilized to establish a PPI network of the ATO target genes.

Genomics information of ATO targets in pancreatic cancer

The cBioPortal for Cancer Genomics (http://www.cbioportal.org/) is a free platform that is used for finding multidimensional cancer genomics data (23). OncoPrint is an online tool, which can visualize alterations of tumor samples from gene arrays. The alterations of 6 ATO target genes associated with pancreatic cancer (AKT1, CCND1, CDKN2A, IKBKB, MAPK1 and MAPK3) were visualized using cBioPortal and OncoPrint (24), and the genomic alteration frequency within the specific cancer study, listed below, was applied as a filter. In the present study, a total of 850 pancreatic cancer samples were analyzed in this section, which included International Cancer Genome Consortium (ICGC; n=99), Queensland Centre for Medical Genomics (QCMG2016; n=456), The Cancer Genome Atlas (TCGA; n=186) and University of Texas Southwestern Medical Center (UTSW; n=109) datasets (2528).

Overall survival analysis of ATO target genes in patients with pancreatic cancer

The K-M plotter (http://kmplot.com/analysis/) is a free online tool that can be used to assess the association of multiple genes with the survival of patients with cancer using different cancer samples (29). According to mRNA expression data from the aforementioned 6 ATO genes, the patients with pancreatic cancer were divided into the high- and low-expression groups based on the median expression value, and then K-M survival curves were constructed. In the present study, the information of 177 patients with pancreatic cancer was acquired from the K-M website, and the K-M survival curves were used to analyze the association between the expression of ATO target genes (AKT1, CCND1, CDKN2A, IKBKB, MAPK1 and MAPK3) and the prognosis of 177 patients with pancreatic cancer.

Determination of the expression of the 3 ATO target genes in pancreatic cancer

Oncomine (https://www.oncomine.org/) is an online cancer microarray website that facilitates the discovery of genome-wide expression analyses (30). In the present study, differential gene expression was detected by Oncomine when comparing cancer tissues with normal pancreatic tissues. The studies by Ishikawa et al (31), Iacobuzio-Donahue et al (32), Pei et al (33), Badea et al (34), Logdons et al (35) and Segara et al (36) were used to reveal the expression of 3 ATO target genes in pancreatic cancer. Differences in transcriptional expression were compared using the Student's t-test. P<0.01 was used to indicate a statistically significant difference and fold change was set to 2.

Results

Evaluation of ATO target genes

ATO is a chemotherapeutic agent of idiopathic function used to treat leukemia that is unresponsive to first-line agents. It is suspected that ATO induces cancer cells to undergo apoptosis (11). In the current study, DrugBank and STITCH tools were used to identify the target genes of ATO. As shown in Table I, a total of 19 target genes were identified, including IKBKB, TXNRD1, JUN, CCND1, MAPK3, MAPK1, AKT1, CDKN1A, HDAC1, PML, RARA, ESR1, CDKN2A, ABCC1, SP1, DNMT3B, DNMT1, HSPA4 and ABCB1.

Table I.

Target genes (n=19) of arsenic trioxide identified using DrugBank 5.0 and STITCH.

Table I.

Target genes (n=19) of arsenic trioxide identified using DrugBank 5.0 and STITCH.

Target geneSource
IKBKBDrugbank 5.0
TXNRD1Drugbank 5.0
JUNDrugbank 5.0
CCND1Drugbank 5.0
MAPK3Drugbank 5.0
MAPK1Drugbank 5.0
AKT1Drugbank 5.0
CDKN1ADrugbank 5.0
PMLDrugbank 5.0 and STITCH
HDAC1STITCH
RARASTITCH
ESR1STITCH
CDKN2ASTITCH
ABCC1STITCH
SP1STITCH
DNMT3BSTITCH
DNMT1STITCH
HSPA4STITCH
ABCB1STITCH
Gene Ontology (GO) analysis

The online STRING software was used to determine functional enrichments based on 19 ATO target genes. GO analysis revealed a significant difference in biological processes, which included ‘response to lipid’, ‘DNA-templated transcription’, ‘initiation, regulation of transcription from RNA polymerase II promoter’, ‘positive regulation of gene expression’, and ‘transcription initiation from RNA polymerase II promoter,’ among others. The molecular function terms identified included ‘enzyme binding’, ‘transcription factor binding’, ‘sequence-specific DNA binding’, ‘cyclin-dependent protein serine/threonine kinase regulator activity’ and ‘transcription corepressor activity’, among others. As regards the area of the cell components, the 19 ATO target genes were enriched in ‘chromatin’, ‘nucleoplasm’, ‘nuclear chromatin’, ‘chromosome’ and ‘nuclear lumen’. The top 5 terms for biological processes, molecular function and cellular components were shown in Table SI.

Association between ATO targets and pancreatic cancer

The PPI network (Fig. 1) and KEGG pathway analysis for the 19 ATO target genes (Table II) were obtained from the STRING database. The top 10 signaling pathways associated with the ATO target genes were as follows: MicroRNAs in cancer, pathways in cancer, chronic myeloid leukemia, acute myeloid leukemia, pancreatic cancer, glioma, melanoma, hepatitis B, pancreatic cancer and viral carcinogenesis. The most common solid tumor was pancreatic cancer (P=6.88×10−10). A total of 6 genes, namely AKT1, CCND1, CDKN2A, IKBKB, MAPK1 and MAPK3, were found to be associated with pancreatic cancer. Subsequently, CluePedia software was used to verify and visualize the association of ATO target genes with pancreatic cancer, and the results were consistent with the STRING analysis results (Fig. 2).

Table II.

Kyoto Encyclopedia of Genes and Genomes pathways associated with the arsenic trioxide target genes.

Table II.

Kyoto Encyclopedia of Genes and Genomes pathways associated with the arsenic trioxide target genes.

PathwayCountFalse discovery rateGenes
MicroRNAs in cancer10 5.09×10−15ABCB1, ABCC1, CCND1, CDKN1A, CDKN2A, DNMT1, DNMT3B, HDAC1, IKBKB, MAPK1
Pathways in cancer11 7.73×10−14AKT1, CCND1, CDKN1A, CDKN2A, HDAC1, IKBKB, JUN, MAPK1, MAPK3, PML, RARA
Chronic myeloid leukemia8 7.73×10−14AKT1, CCND1, CDKN1A, CDKN2A, HDAC1, IKBKB, MAPK1, MAPK
Acute myeloid leukemia7 2.41×10−12AKT1, CCND1, IKBKB, MAPK1, MAPK3, PML, RARA
Pancreatic cancer6 6.88×10−10AKT1, CCND1, CDKN2A, IKBKB, MAPK1, MAPK3
Glioma6 6.88×10−10AKT1, CCND1, CDKN1A, CDKN2A, MAPK1, MAPK3
Hepatitis B7 1.11×10−9AKT1, CCND1, CDKN1A, IKBKB, JUN, MAPK1, MAPK3
Melanoma6 1.11×10−9AKT1, CCND1, CDKN1A, CDKN2A, MAPK1, MAPK3
Prostate cancer6 3.56×10−9AKT1, CCND1, CDKN1A, IKBKB, MAPK1, MAPK3
Viral carcinogenesis7 4.54×10−9CCND1, CDKN1A, CDKN2A, HDAC1, JUN, MAPK1, MAPK3
Genetic data of ATO target genes in pancreatic cancer

The genetic information of these 6 genes in patients with pancreatic cancer were subsequently evaluated using the cBioPortal. The genomic alterations and clinical expression characteristics of AKT1, CCND1, CDKN2A, IKBKB, MAPK1 and MAPK3 in the aforementioned datasets ICGC (n=99), QCMG2016 (n=456), TCGA (n=186) and UTSW (n=109) were investigated (2528). As shown in Fig. 3, the alteration rate of the 6 genes in pancreatic cancer varied between 0 and 63%. AKT1 exhibited an alteration rate between 0 and 18%, CCDN1 had an alteration rate between 0 and 10%, CDKN2A had an alteration between 0 and 42%, IKBKB had an alteration of 0–7%, MAPK1 had an alteration between 0 and 8%, and MAPK3 had an alteration between 0 and 9%. Furthermore, AKT1, IKBKB and MAPK1 exhibited amplification, deep deletion and missense mutations. The alterations of CDKN2A included deep deletion, truncating, missense, inframe mutations and other mutations. The alterations of CCND1 and MAPK3 included amplification.

Interconnection of the 6 ATO target genes in pancreatic cancer

STRING analysis performed in the current study revealed that the 6 ATO target genes (AKT1, CCND1, CDKN2A, IKBKB, MAPK1 and MAPK3) were involved in chronic myeloid leukemia, pancreatic cancer, non-small cell lung cancer, acute myeloid leukemia, melanoma, glioma, prostate cancer, endocrine resistance, FoxO signaling and hepatitis B (Table SII). A PPI network was constructed to analyze the genes interconnected with AKT1, CCND1, CDKN2A, IKBKB, MAPK1 and MAPK3. A total of 20 ATO interacting genes (FOXO1, CDK6, CDK4, RICTOR, IKBKG, FOXO3, PTEN, NFKB1, PIK3CB, JUN, TP53, PIK3CA, PIK3CG, CDKN1A, CDKN1B, CDK2, PIK3CD, MTOR, NFKBIA and RB1) were identified and were mainly involved in hepatitis B, prostate cancer, pathways in cancer, glioma and chronic myeloid leukemia (Fig. 4 and Table III). The results may indicate a new therapeutic strategy for pancreatic cancer.

Table III.

Kyoto Encyclopedia of Genes and Genomes pathways in the protein-protein interaction network of the arsenic trioxide target genes interacted 20 genes.

Table III.

Kyoto Encyclopedia of Genes and Genomes pathways in the protein-protein interaction network of the arsenic trioxide target genes interacted 20 genes.

PathwayCountFalse discovery rateGene name
Hepatitis B13 4.42×10−23CDK4, CDK6, CDKN1A, CDKN1B, JUN, NFKB1, PIK3CA, PIK3CB, PIK3CD, PIK3CG, PTEN, RB1, TP53
Prostate cancer12 4.42×10−23CDKN1A, CDKN1B, FOXO1, MTOR, NFKB1, PIK3CA, PIK3CB, PIK3CD, PIK3CG, PTEN, RB1, TP53
Pathways in cancer15 4.43×10−23CDK4, CDK6, CDKN1A, CDKN1B, FOXO1, JUN, MTOR, NFKB1, PIK3CA, PIK3CB, PIK3CD, PIK3CG, PTEN, RB1, TP53
Glioma11 1.43×10−22CDK4, CDK6, CDKN1A, MTOR, PIK3CA, PIK3CB, PIK3CD, PIK3CG, PTEN, RB1, TP53
Chronic myeloid leukemia11 4.99×10−22CDK4, CDK6, CDKN1A, CDKN1B, NFKB1, PIK3CA, PIK3CB, PIK3CD, PIK3CG, RB1, TP53
Small cell lung cancer11 4.21×10−21CDK4, CDK6, CDKN1B, NFKB1, PIK3CA, PIK3CB, PIK3CD, PIK3CG, PTEN, RB1, TP53
Viral carcinogenesis12 1.16×10−19CDK4, CDK6, CDKN1A, CDKN1B, JUN, NFKB1, PIK3CA, PIK3CB, PIK3CD, PIK3CG, RB1, TP53
Melanoma10 1.48×10−19CDK4, CDK6, CDKN1A, PIK3CA, PIK3CB, PIK3CD, PIK3CG, PTEN, RB1, TP53
PI3K-AKT signaling pathway13 1.27×10−18CDK4, CDK6, CDKN1A, CDKN1B, FOXO3, MTOR, NFKB1, PIK3CA, PIK3CB, PIK3CD, PIK3CG, PTEN, TP53
Non-small cell lung cancer9 4.22×10−18CDK4, CDK6, FOXO3, PIK3CA, PIK3CB, PIK3CD, PIK3CG, RB1, TP53
Survival prediction of the 6 ATO target genes in patients with pancreatic cancer

After the 6 ATO target genes associated with pancreatic cancer were identified, the K-M plotter was used to analyze the association of these genes with the overall survival of 177 patients with pancreatic cancer. As shown in Fig. 5, a low AKT1 level indicated poorer prognosis in patients with pancreatic cancer [hazard ratio (HR)=0.60; P=0.04]. Furthermore, a high level of CCND1 was correlated with poor prognosis of patients with pancreatic cancer (HR=2.71; P=2×10−4). MAPK1 may also serve as a prognostic factor for patients with pancreatic cancer (HR=1.74; P<0.01), with high expression levels indicating poor prognosis. However, CKDN2A (P=0.17), IKBKB (P=0.33) and MAPK3 (P=0.24) were not found to be prognostic factors for patients with pancreatic cancer.

Differential expression of the 3 ATO target genes in pancreatic cancer

As aforementioned, AKT1, CCND1 and MAPK1 were demonstrated to be associated with the prognosis of pancreatic cancer. Differential expression analysis of these three ATO target genes was then conducted using the Oncomine website. As shown in Fig. 6, AKT1 was observed to be overexpressed in pancreatic cancer compared with normal pancreatic tissue in the studies of Ishikawa et al (31), Iacobuzio-Donahue et al (32) and Pei et al (33) (P<0.01, P<0.01 and <0.01, respectively). Badea et al (34), Logdons et al (35) and Pei et al (33) also reported that CCND1 was more highly expressed in pancreatic cancer as compared with the normal pancreas (P=4.04×10−6, P<0.01 and P=5.51×10−4, respectively). Furthermore, MAPK1 was reported to be overexpressed in pancreatic cancer by Pei et al (33) and Segara et al (36) (P=0.01 and P<0.01, respectively).

Discussion

Pancreatic cancer is one of the most aggressive malignancies and the fourth cause of cancer-associated mortalities worldwide. This cancer is frequently diagnosed at an advanced stage, and most tumors are unresectable at clinical presentation (37), which leads to a poor prognosis and high mortality rates among patients with pancreatic cancer. Despite the advances in diagnostic and treatment methods, there have been no significant improvements in the prognosis of patients with pancreatic cancer in the past years. Therefore, more effective treatment methods for pancreatic cancer are urgently required.

The antitumor properties of ATO have been identified in a number of tumors (1014). ATO has also been shown to inhibit the viability of pancreatic cancer stem cells in vitro and in vivo (38). In addition, Gao et al (15) reported that ATO exerted anticancer effects on pancreatic cancer. The combination of ATO with hypoxia-inducible factor-1 inhibitor was also found to be effective (39). However, the potential targets and molecular mechanisms of action of ATO in pancreatic cancer remain elusive.

In the present study, the molecular targets and associated genes of ATO were investigated by bioinformatics analysis. First, the 19 target genes of ATO were identified by DrugBank 5.0 and STITCH, which included IKBKB, TXNRD1, JUN, CCND1, MAPK3, MAPK1, AKT1, CDKN1A, HDAC1, PML, RARA, ESR1, CDKN2A, ABCC1, SP1, DNMT3B, DNMT1, HSPA4 and ABCB1. Next, the STRING website was used to construct the PPI network and conduct KEGG pathway analysis for the ATO target genes. A total of 6 target genes were demonstrated to be associated with pancreatic cancer, including AKT1, CCND1, CDKN2A, IKBKB, MAPK1 and MAPK3. Subsequently, the cBioPortal database was used to detect the alterations of these 6 genes, and the results were as follows: AKT1, IKBKB and MAPK1 were found to display amplification, deep deletion and missense mutations; CDKN2A exhibited deep deletion, truncating, missense, inframe and other mutations; and CCND1 and MAPK3 exhibited amplification. Furthermore, 20 genes that are interconnected with the 6 ATO target genes in pancreatic cancer were identified. These genes were mainly involved in hepatitis B, pancreatic cancer, pathways in cancer, glioma and chronic myeloid leukemia. Finally, the K-M plotter was used to analyze the association of the 6 target genes with the overall survival of patients with pancreatic cancer and Oncomine was used to analyze differential expression of these genes in pancreatic cancer. The results demonstrated that CCND1 and MAPK1 were overexpressed in pancreatic cancer and may be of prognostic value for patients with pancreatic cancer.

By conducting bioinformatics analysis, 6 ATO target genes were identified in the current study. Among these 6 genes, AKT1 is a member of the PI3K/AKT pathway and serves an important role in multiple types of cancer, including pancreatic cancer (40). CCND1, also referred to as cyclin D1, forms a complex with CDK4, thus phosphorylating and inhibiting RB1, and regulating the cell cycle during G1/S transition. The activation of the CCND1/CDK4 complex also promotes pancreatic cancer cell growth (41). Furthermore, it is well-established that the increased risk of pancreatic cancer is partly due to germline mutation carrier of CDKN2A (42). A previous study demonstrated that nuclear factor (NF)-κB promoted the proliferation and growth, and inhibited the apoptosis of pancreatic cancer cells (43). Inhibition of NF-κB also blocked metastasis in pancreatic cancer-derived xenograft tumors (44). In another study, the MAPK/ERK signaling pathway was revealed to be correlated with cancer cell growth, proliferation and apoptosis, including pancreatic cancer (45). Hu et al (46) used integrative analysis and identified MAPK3 as a significant hub gene in pancreatic cancer. In the present study, AKT1, CCND1 and MAPK1 were found to be overexpressed in pancreatic cancer, while high CCND1 and MAPK1 levels indicated poor prognosis.

STRING analysis performed in the current study revealed that the 6 ATO target genes (AKT1, CCND1, CDKN2A, IKBKB, MAPK1 and MAPK3) were involved in chronic myeloid leukemia, pancreatic cancer, non-small cell lung cancer, acute myeloid leukemia, melanoma, glioma, prostate cancer, endocine resistance FoxO signaling and hepatitis B. The PPI network was also used to analyze the genes interconnected with the 6 ATO target genes, and the 20 interconnected genes were observed to be involved in hepatitis B, prostate cancer, pathways in cancer, glioma and chronic myeloid leukemia. These results may help develop new therapeutic strategies for cancer.

However, there were certain limitations to the present study. Firstly, this research was based on bioinformatics analysis, and experimental results are required to support the conclusions. Secondly, since the NF-κB pathway serves an important role in multiple cancer development, a significant association of IKBKB with poor prognosis of patients with pancreatic cancer was not observed by Kaplan-Meier analysis. Furthermore, the key genes implicated in pancreatic cancer should also be analyzed to confirm the reliability of the results. The present study showed that 6 ATO target genes (AKT1, CCND1, CDKN2A, IKBKB, MAPK1 and MAPK3) were associated with pancreatic cancer, and that high CCND1 and MAPK1 levels indicated poor prognosis. However, since tissue samples were not collected, these 6 proteins could not be verified in pancreatic cancer samples. Next, immunohistochemistry will be used to validate the expression of these proteins in pancreatic cancer and evaluate their association with the prognosis of patients with pancreatic cancer.

In conclusion, the bioinformatics analysis was used to elucidate the molecular mechanism of action of ATO in pancreatic cancer. The results not only suggest that ATO may be an effective drug for pancreatic cancer, but also provide a novel approach for identifying potential treatments for other diseases by bioinformatics analysis.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from the National Natural Science Foundation of China (no. 81672921) and the Innovation Capacity Support Plan of Shaanxi Province (no. 2018TD-002).

Availability of data and materials

The datasets generated and analyzed during the present study are available in the public databases.

Authors' contributions

QZ and SXH designed the study. CYZ acquired and analyzed the data, drafted the manuscript and revised it. LYG was involved in data analysis and revised the manuscript. RL, NNW, YYF, YPD, HZ, YQZ and YYZ performed the data analysis. All authors read and approved the final manuscript.

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:

ATO

arsenic trioxide

PPI

protein-protein interaction

KEGG

Kyoto Encyclopedia of Genes and Genomes

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Saad AM, Turk T, Al-Husseini MJ and Abdel-Rahman O: Trends in pancreatic adenocarcinoma incidence and mortality in the United States in the last four decades; a SEER-based study. BMC Cancer. 18:6882018. View Article : Google Scholar : PubMed/NCBI

3 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Soignet SL, Frankel SR, Douer D, Tallman MS, Kantarjian H, Calleja E, Stone RM, Kalaycio M, Scheinberg DA, Steinherz P, et al: United States multicenter study of arsenic trioxide in relapsed acute promyelocytic leukemia. J Clin Oncol. 19:3852–3860. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Mathews V, George B, Chendamarai E, Lakshmi KM, Desire S, Balasubramanian P, Viswabandya A, Thirugnanam R, Abraham A, Shaji RV, et al: Single-agent arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia: Long-term follow-up data. J Clin Oncol. 28:3866–3871. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Ghavamzadeh A, Alimoghaddam K, Rostami S, Ghaffari SH, Jahani M, Iravani M, Mousavi SA, Bahar B and Jalili M: Phase II study of single-agent arsenic trioxide for the front-line therapy of acute promyelocytic leukemia. J Clin Oncol. 29:2753–2757. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Lo-Coco F, Avvisati G, Vignetti M, Thiede C, Orlando SM, Iacobelli S, Ferrara F, Fazi P, Cicconi L, Di Bona E, et al: Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N Engl J Med. 369:111–121. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Burnett AK, Russell NH, Hills RK, Bowen D, Kell J, Knapper S, Morgan YG, Lok J, Grech A, Jones G, et al: Arsenic trioxide and all-trans retinoic acid treatment for acute promyelocytic leukaemia in all risk groups (AML17): Results of a randomised, controlled, phase 3 trial. Lancet Oncol. 16:1295–1305. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Seftel MD, Barnett MJ, Couban S, Leber B, Storring J, Assaily W, Fuerth B, Christofides A and Schuh AC: A Canadian consensus on the management of newly diagnosed and relapsed acute promyelocytic leukemia in adults. Curr Oncol. 21:234–250. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Zhang L, Liu L, Zhan S, Chen L, Wang Y, Zhang Y, Du J, Wu Y and Gu L: Arsenic trioxide suppressed migration and angiogenesis by targeting FOXO3a in gastric cancer cells. Int J Mol Sci. 19(pii): E37392018. View Article : Google Scholar : PubMed/NCBI

11 

Zhong L, Xu F and Chen F: Arsenic trioxide induces the apoptosis and decreases NF-KB expression in lymphoma cell lines. Oncol Lett. 16:6267–6274. 2018.PubMed/NCBI

12 

Mao MH, Huang HB, Zhang XL, Li K, Liu YL and Wang P: Additive antitumor effect of arsenic trioxide combined with intravesical bacillus Calmette-Guerin immunotherapy against bladder cancer through blockade of the IER3/Nrf2 pathway. Biomed Pharmacother. 107:1093–1103. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Du S, Liu K, Gao P, Li Z and Zheng J: Differential anticancer activities of arsenic trioxide on head and neck cancer cells with different human papillomavirus status. Life Sci. 212:182–193. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Luo D, Zhang X, Du R, Gao W, Luo N, Zhao S, Li Y, Chen R, Wang H, Bao Y, et al: Low dosage of arsenic trioxide (As2O3) inhibits angiogenesis in epithelial ovarian cancer without cell apoptosis. J Biol Inorg Chem. 23:939–947. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Gao JK, Wang LX, Long B, Ye XT, Su JN, Yin XY, Zhou XX and Wang ZW: Arsenic Trioxide inhibits cell growth and invasion via Down-Regulation of Skp2 in pancreatic cancer cells. Asian Pac J Cancer Prev. 16:3805–3810. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Huang YA, You ZH and Chen X: A systematic prediction of drug-target interactions using molecular fingerprints and protein sequences. Curr Protein Pept Sci. 19:468–478. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Wishart DS, Knox C, Guo AC, Cheng D, Shrivastava S, Tzur D, Gautam B and Hassanali M: DrugBank: A knowledgebase for drugs, drug actions and drug targets. Nucleic Acids Res. 36:D901–D906. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Kuhn M, von Mering C, Campillos M, Jensen LJ and Bork P: STITCH: Interaction networks of chemicals and proteins. Nucleic Acids Res. 36:D684–D688. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, et al: STRING v10: Protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 43:D447–D452. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, Santos A, Doncheva NT, Roth A, Bork P, et al: The STRING database in 2017: Quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 45:D362–D368. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Bindea G, Galon J and Mlecnik B: CluePedia Cytoscape plugin: Pathway insights using integrated experimental and in silico data. Bioinformatics. 29:661–663. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6:pl12013. View Article : Google Scholar : PubMed/NCBI

25 

Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK, Wilson PJ, Patch AM, Wu J, et al: Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 491:399–405. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, Miller DK, Christ AN, Bruxner TJ, Quinn MC, et al: Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 531:47–52. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, Dimitriadoy S, Liu DL, Kantheti HS, Saghafinia S, et al: Oncogenic signaling pathways in the cancer genome atlas. Cell. 173:321–337.e10. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, Mollaee M, Wagner KU, Koduru P, Yopp A, et al: Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 6:67442015. View Article : Google Scholar : PubMed/NCBI

29 

Lanczky A, Nagy A, Bottai G, Munkácsy G, Szabó A, Santarpia L and Győrffy B: miRpower: A web-tool to validate survival-associated miRNAs utilizing expression data from 2178 breast cancer patients. Breast Cancer Res Treat. 160:439–446. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Rhodes DR, Kalyana-Sundaram S, Mahavisno V, Varambally R, Yu J, Briggs BB, Barrette TR, Anstet MJ, Kincead-Beal C, Kulkarni P, et al: Oncomine 3.0: Genes, pathways, and networks in a collection of 18,000 cancer gene expression profiles. Neoplasia. 9:166–180. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Ishikawa M, Yoshida K, Yamashita Y, Ota J, Takada S, Kisanuki H, Koinuma K, Choi YL, Kaneda R, Iwao T, et al: Experimental trial for diagnosis of pancreatic ductal carcinoma based on gene expression profiles of pancreatic ductal cells. Cancer Sci. 96:387–393. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Iacobuzio-Donahue CA, Maitra A, Olsen M, Lowe AW, van Heek NT, Rosty C, Walter K, Sato N, Parker A, Ashfaq R, et al: Exploration of global gene expression patterns in pancreatic adenocarcinoma using cDNA microarrays. Am J Pathol. 162:1151–1162. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Pei H, Li L, Fridley BL, Jenkins GD, Kalari KR, Lingle W, Petersen G, Lou Z and Wang L: FKBP51 affects cancer cell response to chemotherapy by negatively regulating Akt. Cancer Cell. 16:259–266. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Badea L, Herlea V, Dima SO, Dumitrascu T and Popescu I: Combined gene expression analysis of whole-tissue and microdissected pancreatic ductal adenocarcinoma identifies genes specifically overexpressed in tumor epithelia. Hepatogastroenterology. 55:2016–2027. 2008.PubMed/NCBI

35 

Logsdon CD, Simeone DM, Binkley C, Arumugam T, Greenson JK, Giordano TJ, Misek DE, Kuick R and Hanash S: Molecular profiling of pancreatic adenocarcinoma and chronic pancreatitis identifies multiple genes differentially regulated in pancreatic cancer. Cancer Res. 63:2649–2657. 2003.PubMed/NCBI

36 

Segara D, Biankin AV, Kench JG, Langusch CC, Dawson AC, Skalicky DA, Gotley DC, Coleman MJ, Sutherland RL and Henshall SM: Expression of HOXB2, a retinoic acid signaling target in pancreatic cancer and pancreatic intraepithelial neoplasia. Clin Cancer Res. 11:3587–3596. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Chari ST: Detecting early pancreatic cancer: Problems and prospects. Semin Oncol. 34:284–294. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Han JB, Sang F, Chang JJ, Hua YQ, Shi WD, Tang LH and Liu LM: Arsenic trioxide inhibits viability of pancreatic cancer stem cells in culture and in a xenograft model via binding to SHH-Gli. Onco Targets Ther. 6:1129–1138. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Lang M, Wang X, Wang H, Dong J, Lan C, Hao J, Huang C, Li X, Yu M, Yang Y, et al: Arsenic trioxide plus PX-478 achieves effective treatment in pancreatic ductal adenocarcinoma. Cancer Lett. 378:87–96. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Michl P and Downward J: Mechanisms of disease: PI3K/AKT signaling in gastrointestinal cancers. Z Gastroenterol. 43:1133–1139. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Yan L, Wang Y, Wang ZZ, Rong YT, Chen LL, Li Q, Liu T, Chen YH, Li YD, Huang ZH and Peng J: Cell motility and spreading promoted by CEACAM6 through cyclin D1/CDK4 in human pancreatic carcinoma. Oncol Rep. 35:418–426. 2016. View Article : Google Scholar : PubMed/NCBI

42 

McWilliams RR, Wieben ED, Chaffee KG, Antwi SO, Raskin L, Olopade OI, Li D, Highsmith WE Jr, Colon-Otero G, Khanna LG, et al: CDKN2A Germline rare coding variants and risk of pancreatic cancer in minority populations. Cancer Epidemiol Biomarkers Prev. 27:1364–1370. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Liptay S, Weber CK, Ludwig L, Wagner M, Adler G and Schmid RM: Mitogenic and antiapoptotic role of constitutive NF-kappaB/Rel activity in pancreatic cancer. Int J Cancer. 105:735–746. 2003. View Article : Google Scholar : PubMed/NCBI

44 

Fujioka S, Sclabas GM, Schmidt C, Frederick WA, Dong QG, Abbruzzese JL, Evans DB, Baker C and Chiao PJ: Function of nuclear factor kappaB in pancreatic cancer metastasis. Clin Cancer Res. 9:346–354. 2003.PubMed/NCBI

45 

Hu Y, Yang H, Lu XQ, Xu F, Li J and Qian J: ARHI suppresses pancreatic cancer by regulating MAPK/ERK 1/2 pathway. Pancreas. 44:342–343. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Hu B, Shi C, Jiang HX and Qin SY: Identification of novel therapeutic target genes and pathway in pancreatic cancer by integrative analysis. Medicine (Baltimore). 96:e82612017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 18 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou CY, Gong LY, Liao R, Weng NN, Feng YY, Dong YP, Zhu H, Zhao YQ, Zhang YY, Zhu Q, Zhu Q, et al: Evaluation of the target genes of arsenic trioxide in pancreatic cancer by bioinformatics analysis. Oncol Lett 18: 5163-5172, 2019
APA
Zhou, C., Gong, L., Liao, R., Weng, N., Feng, Y., Dong, Y. ... Han, S. (2019). Evaluation of the target genes of arsenic trioxide in pancreatic cancer by bioinformatics analysis. Oncology Letters, 18, 5163-5172. https://doi.org/10.3892/ol.2019.10889
MLA
Zhou, C., Gong, L., Liao, R., Weng, N., Feng, Y., Dong, Y., Zhu, H., Zhao, Y., Zhang, Y., Zhu, Q., Han, S."Evaluation of the target genes of arsenic trioxide in pancreatic cancer by bioinformatics analysis". Oncology Letters 18.5 (2019): 5163-5172.
Chicago
Zhou, C., Gong, L., Liao, R., Weng, N., Feng, Y., Dong, Y., Zhu, H., Zhao, Y., Zhang, Y., Zhu, Q., Han, S."Evaluation of the target genes of arsenic trioxide in pancreatic cancer by bioinformatics analysis". Oncology Letters 18, no. 5 (2019): 5163-5172. https://doi.org/10.3892/ol.2019.10889