Next Article in Journal
Potential for a Plant-Made SARS-CoV-2 Neutralizing Monoclonal Antibody as a Synergetic Cocktail Component
Previous Article in Journal
Persistent Enterovirus Infection: Little Deletions, Long Infections
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Disclosing Potential Key Genes, Therapeutic Targets and Agents for Non-Small Cell Lung Cancer: Evidence from Integrative Bioinformatics Analysis

by
Md. Parvez Mosharaf
1,2,
Md. Selim Reza
1,3,
Esra Gov
4,
Rashidul Alam Mahumud
5 and
Md. Nurul Haque Mollah
1,*
1
Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh
2
School of Commerce, Faculty of Business, Education, Law and Arts, University of Southern Queensland, Toowoomba, QLD 4350, Australia
3
Centre for High Performance Computing, Joint Engineering Research Centre for Health Big Data Intelligent Analysis Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
4
Department of Bioengineering, Faculty of Engineering, Adana AlparslanTurkes Science and Technology University, Adana 01250, Turkey
5
NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
*
Author to whom correspondence should be addressed.
Submission received: 6 March 2022 / Revised: 7 May 2022 / Accepted: 8 May 2022 / Published: 12 May 2022

Abstract

:
Non-small-cell lung cancer (NSCLC) is considered as one of the malignant cancers that causes premature death. The present study aimed to identify a few potential novel genes highlighting their functions, pathways, and regulators for diagnosis, prognosis, and therapies of NSCLC by using the integrated bioinformatics approaches. At first, we picked out 1943 DEGs between NSCLC and control samples by using the statistical LIMMA approach. Then we selected 11 DEGs (CDK1, EGFR, FYN, UBC, MYC, CCNB1, FOS, RHOB, CDC6, CDC20, and CHEK1) as the hub-DEGs (potential key genes) by the protein–protein interaction network analysis of DEGs. The DEGs and hub-DEGs regulatory network analysis commonly revealed four transcription factors (FOXC1, GATA2, YY1, and NFIC) and five miRNAs (miR-335-5p, miR-26b-5p, miR-92a-3p, miR-155-5p, and miR-16-5p) as the key transcriptional and post-transcriptional regulators of DEGs as well as hub-DEGs. We also disclosed the pathogenetic processes of NSCLC by investigating the biological processes, molecular function, cellular components, and KEGG pathways of DEGs. The multivariate survival probability curves based on the expression of hub-DEGs in the SurvExpress web-tool and database showed the significant differences between the low- and high-risk groups, which indicates strong prognostic power of hub-DEGs. Then, we explored top-ranked 5-hub-DEGs-guided repurposable drugs based on the Connectivity Map (CMap) database. Out of the selected drugs, we validated six FDA-approved launched drugs (Dinaciclib, Afatinib, Icotinib, Bosutinib, Dasatinib, and TWS-119) by molecular docking interaction analysis with the respective target proteins for the treatment against NSCLC. The detected therapeutic targets and repurposable drugs require further attention by experimental studies to establish them as potential biomarkers for precision medicine in NSCLC treatment.

1. Introduction

Lung cancer is treated as the leading cause of cancer-related death worldwide among human cancer, which causes the dynamic degradation of the lung [1]. The most common type of bronchial tumor is non-small-cell lung cancer (NSCLC), which accounts for approximately 75% of all lung cancers [2]. The NSCLC is more deadly than the small-cell lung cancer (SCLC), though it grows and spreads slowly compared with the SCLC since it progresses to the advanced stage with few or without any symptoms. Although the targeted therapy has achieved substantial development, the increasing mortality rate associated with lung cancer lays emphasis on both prevention and early detection of lung cancer. Traditional cancer diagnosis methods including histopathology and cytopathologyare practiced in the case of adenocarcinoma, squamous cell carcinoma, and large-cell carcinoma of NSCLC [3,4,5]. The morphological judgment for the tumors has some limitations, including the lack of significant morphological features, which leads to the identification ambiguities [6,7,8,9,10]. Several non-causal risk factors (e.g., smoking, alcohol consumption, and high air pollution) of lung cancer have been detected by several independent studies [11,12,13,14,15]. However, so far, there are no in-depth studies that explore the causal risk factors of NSCLC highlighting their pathogenetic processes and associated candidate drugs for the treatment against NSCLC. The causal risk factors are known as the mutated genes that drive the cancer progression. Usually, non-causal risk factors are assumed to be responsible for genetic mutation and some of them stimulate cancer progression. Cancer-causing mutated genes are utilized for diagnosis, prognosis, and therapies of cancer [16,17]. Moreover, the DNA vaccine is part of a new era of modern therapeutics where the gene-based prophylactic vaccines are being developed [18,19,20]. The plasmid DNA vaccines and viral-vectored vaccines are two types of gene-based vaccines on which many animal trials are being practiced all over the world [21,22]. Therefore, the cancer-causing genes also might be a great therapeutics target for the gene-based DNA vaccine development.
Gene expression profile analysis is now considered as one of the most promising approaches for exploring cancer-causing mutated genes, which yields relevant information for diagnosis, prognosis, and therapies of cancers. [23,24,25,26]. Computationally, mutated genes (potential key genes) are predicted by the analysis of differential gene expression patterns [16,17,23,24,25,26,27,28,29]. Therefore, in this study, an attempt was made to explore NSCLC-causing key genes from the publicly available gene expression profiles, highlighting their functions, pathways, and regulators, which yield relevant information for diagnosis, prognosis, and therapies of NSCLC, by using the integrated bioinformatics approaches.

2. Materials and Methods

To reach the goal of this study, we analyzed a publicly available gene expression dataset by using integrated bioinformatics approaches [16,28,29]. The global working flowchart of this study is displayed in Figure 1.

2.1. Collection of Gene Expression Profiles for NSCLC

To explore NSCLC-causing key genes, the Affymetrix Human Genome U133 plus 2.0 microarray gene expression dataset was retrieved from the NCBI Gene Expression Omnibus (GEO) database [30] with accession number GSE19804, which contained 60 tumor samples and 60 control samples on 54,675 genes. The dataset was generated by a previous study [31]. The sample unit was aged from 37 to 80 years, with nine different tumor stages (i.e., 1, 1A, 1B, 2, 2A, 2B, 3A, 3B, 4).

2.2. Differentially Expressed Genes (DEGs) Identification

At first, the gene expression dataset was normalized for identifying DEGs through the Robust Multi-Array Average (RMA) expression measure and it was implemented by the NCBI-GEO2R (https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/geo/geo2r/, accessed on 5 October 2021) web-tool. Then, the LIMMA [32] statistical test was utilized to identify the DEGs between NSCLC and control samples. To control the false discovery rate in multiple-testing, the p-values were adjusted by Benjamini Hochberg’s [33] method. Both the adjusted p-value and log2FC values were considered for identifying the upregulated and downregulated DEGs as follows:
DEGs = { Upregulated   DEGs ,   if   adjusted   p   value < 0.001   &   log 2 FC > 1   Downregulated   DEGs ,   if   adjusted   p   value < 0.001   &   log 2 FC < 1

2.3. DEGs-Set Enrichment Analysis

The bioinformatics resources, Database for Annotation, Visualization and Integrated Discovery (DAVID) (version v6.8) [34,35] was utilized to discern molecular function, biological process, and molecular pathway annotations related to the identified DEGs. Besides, the KEGG pathways identification was conducted through the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database [36,37,38]. For statistical significance, the adjusted p-value < 0.05 was considered, determined from Fisher Exact test and Benjamini–Hochberg’s correction was used for the multiple testing correction techniques.

2.4. Protein-Protein Interaction Network Analysis of DEGs

The STRING database [39] was used to construct the protein–protein interaction (PPI) network of the proteins encoded by DEGs. The STRING database uses a score combiner depending on the product of probabilities [40]. To visualize and perform topological analyses of the PPI network, the NetworkAnalyst [41] was utilized. The topological analysis was applied to determine hub-DEGs/proteins through the CytoHubba plugin [42] in Cytoscape 3.8.2 using degree (connectivity) and betweenness metrics simultaneously [43]. The minimum degree of 10 was considered as the cut off criterion in CytoHubba. Furthermore, the Molecular Complex Detection (MCODE), a novel clustering algorithm [44] along with the CytoHubba was used to identify the sub-modules from the PPI network. The top-scored modules are presented in this analysis.

2.5. Mutation Analysis of Hub-DEGs

To investigate the genomic alterations/mutations of the hub-genes, the online cBioPortal (https://www.cbioportal.org, accessed on 28 March 2022) was used over the NSCLC datasets of the server [45,46]. The OncoPrint output was used to represent the most important alteration frequency of genes.

2.6. Physicochemical Properties of Hub Proteins

The physicochemical properties of the detected hub proteins were reported from the online tool ProtParam (https://web.expasy.org/protparam/, accessed on 10 November 2021), which allows the computation of various physical and chemical parameters for a given protein. The physiochemical properties of molecular weight, theoretical pI, extinction coefficient, instability index, aliphatic index, and grand average of hydropathicity (GRAVY) were checked for the reported hub protein in this study.

2.7. Regulatory Biomolecules Selection

To explore transcriptional and post-transcriptional regulators of DEGs, we performed TFs-DEGs and miRNA-DEGs interaction network analysis, respectively. The TarBase and miRTarBase [47,48] databases were used to identify the significant miRNAs. The JASPAR database [49] retrieved the key regulatory transcription factors (TFs). The entire analysis was conducted through the NetworkAnalyst [41].

2.8. Cross-Validation and Evaluation of the Performance of Reported Biomolecules

At first, patients were divided into a low-risk group (control group) and high-risk group (SCLC group) in the SurvExpress online server [50]. Then, the differences between the risk groups from the expression levels of hub-DEGs were investigated by using box plots and survival probability curves.The statistical significance of the differences in the box plots were evaluated through the t-test. Survival signatures of the reported biomolecules were evaluated through Kaplan–Meier plots, and a log-rank p-value < 0.01 for the statistical significance in all survival analyses.

2.9. Drug Repositioning

The hub-DEGs-guided probable drugs or drug candidate molecules were retrieved through the online drug-repositioning tool and database Connectivity Map (CMap) [51]. This is an integrative platform that accumulates the information of the drug or drug candidate molecules from published data sources in clinical experimental stages, investigational stages, and approved for treatment stages. Furthermore, the molecular docking simulation study [52] was conducted for the target biomolecules with the repositioned drug to identify the best-fitted position with binding affinity. The highest docking score with the best-fit pose was considered for the drug–protein interaction affinity. An important type of molecular docking is protein–ligand docking because of its therapeutic applications in modern structure-based drug design [52]. Here, have performed some vital protein ligand docking and studied the interacting amino acids of the same complex. The 3D structure of the target proteins was obtained from Protein Data bank (PDB). The chemical structure of drugs was retrieved from PubChem database (https://pubchem.ncbi.nlm.nih.gov/, accessed on 5 December 2021). All generated chemical compound structures were energy minimized by the MMFF94 force field [53]. For the binding sites, predictions of target proteins were analyzed through 3DLigandSite—Ligand-binding site prediction Server [54]. Docking analysis was carried out using Autodock 4.2 [55] and AutoDock Vina [56]. The interactions like Hydrogen Bonding and other non-bonded terms between all drug and target proteins were carried out using the Accelrys Discovery Studio Visualizer software [57].

3. Results

3.1. Differentially Expressed Genes (DEGs) Identification

At first, we normalized the genes expression profiles by using RMA. Then, we analyzed the normalized dataset by the statistical LIMMA approach and isolated 1943 DEGs between NSCLC and control samples with the cutoff at adjusted p-value < 0.001 and |log2FC| < 1 (Figure 2A). Among those, 1367 DEGs were upregulated, and the remaining 576 DEGs were downregulated (Figure 2B). Further analysis was conducted based on these DEGs.

3.2. Protein-Protein Interaction Analysis

The PPI network analysis was conducted to reveal the central highly connected proteins which are called hub-DEGs, or proteins or key genes/proteins based on the degree measures (Figure 3) through Cytoscope 3.7.2 with CytoHubba. The degree was considered as ≥10 along with the other default parameters. The proposed top hub proteins are CDK1, EGFR, FYN, UBC, MYC, CCNB1, FOS, RHOB, CDC6, CDC20, and CHEK1, which could be the main proteins in the NSCLC pathogenesis mechanism. By using the MCODE algorithm, 19 sub-network modules were selected considering the default parameters such as node score cutoff of 0.2, K-Core value of 2, and maximum depth from the seed node of 100 along with the other default parameters. Based on the score, the top four modules are represented in Figure 4 and details of analysis results are provided in Supplementary Figure S1. The sub-modules were checked and the presence of the proposed hub proteins was found. The presence of the hub proteins indicates that these are more reliable to treat as potential therapeutic targets.

3.3. Mutation Analysis of Hub-DEGs

The genomic alteration/mutation analysis of 11 hub-DEGs revealed that the EGFR, MYC, and CHEK1 genes had 12%, 8%, and 1.3% genomic alteration/mutation over the four lung cancer studies. Other genes were consistent among the studies. For details of the genomic alteration/mutation summary, see Supplementary Figure S1.
The physicochemical properties of the identified hub proteins are reported in this study. These properties are essential for deeper investigation of the significant biomolecules. The EGFR protein had the highest molecular weight (MW) of 134,277.4 kda, where the UBC reflected the lowest 18,006.82 kda MW. The isoelectric point ranged from 4.77 (FOS) to 9.64 (CDC6) among the reported hub proteins. The detailed information is summarized in Table 1.

3.4. Biological Importance of DEGs

DAVID (version v6.8) revealed the molecular function, biological process, and molecular pathway annotations of the identified DEGs through the gene over-representation analysis. The significant GO terms were retrieved, which included the biological processes, molecular function, and cellular components (Table 2). The significant GO terms are summarized and presented in Table 2 for upregulated and downregulated genes separately. The significant functional pathways obtained from the KEGG Pathway analysis are also shown in Figure 5 for the hub-DEGs. The pathways in cancer, cytokine–cytokine receptor interaction, chemokine signaling pathway, cell-adhesion molecules (CAMs), cAMP signaling pathway, MAPK signaling pathway, and TNF signaling pathway are the significant pathways shared by the upregulated genes (Figure 5A). The metabolic pathways, cell cycle, PI3K-Akt signaling pathway, focal adhesion, and ECM-receptor interaction key pathways are exhibited by the downregulated genes (Figure 5B).

3.5. Regulatory Transcriptional/Post Transcriptional Candidates in in NSCLC

The TFs-DEGs interaction network and the miRNA-DEGs interaction network revealed the substantial TFs and the miRNAs (Figure 6) that may significantly regulate the DEGs. The transcription factors (FOXC1, GATA2, YY1, E2F1, FOXL1, NFIC, NFKB1, PPARG, TFAP2A, USF2) and miRNA (miR-335-5p, miR-26b-5p, miR-16-5p, miR-124-3p, miR-92a-3p, miR-7b-5p, miR-93-5p, miR-17-5p, miR-155-5p) were selected as the key transcriptional and post-transcriptional regulatory biomolecules of DEGs. Furthermore, the interaction network of hub proteins with TFs and miRNA were constructed (Figure 7). The hub-proteins versus TFs interaction network reflected four TFs (FOXC1, GATA2, YY1, and NFIC) as the key regulatory TFs of the drug target hub-DEGs/proteins (Figure 7A). On the other hand, five miRNAs (miR-335-5p, miR-26b-5p, miR-92a-3p, miR-155-5p, and miR-16-5p) were found as the key regulatory miRNAs of hub-DEGs/proteins (Figure 7B). These regulatory biomolecules were also found from the interaction network analysis of DEGs-TF and all DEGs-miRNA, respectively (Figure 6).

3.6. Risk Discrimination Performance of Reporter Biomolecules

The risk discrimination performance and the differential expression pattern were observed by the online gene validation website SurvExpress. The analysis was conducted through the TCGA Lung squamous cell carcinoma survival information for the hub genes and the key transcription factors. The survival curve for the high- and low-risk group and the box plot of their gene expressions are shown in (Figure 8). For both analyses, the prognostic index, log-rank test, and hazard ratio are shown (Figure 8). All hub proteins and reported TFs showed statistically significant performances in terms of survival probabilities in all datasets, in both the high- and low-risk groups.

3.7. Drug Repositioning

The identification of the drug candidate molecules through CMap database revealed the repurposed drugs for the top hub drug-target proteins. The CMap database reflected the drug candidate molecules for the submitted hub proteins. Among the top hub proteins, for the CDK1, EGFR, FYN, and MYC, we found repurposable drugs in pre-clinical trials, FDA-approved drugs, and those in other experimental stages (Table 3).
The molecular docking analysis for the FDA-approved, launched drugs with the hub proteins was conducted. The best pose with the highest docking score was considered to select the drug–protein interaction. The potential repositioned drug candidates need deeper attention for further experimental validation, which leads to the development of more efficient therapy for NSCLC treatment. The molecular docking analysis results are summarized in Figure 9, where (i) indicates the protein–drug complex and (ii) indicates the 2D diagram with interacting amino acid. For the Dinaciclib–CDK1 complex, interaction in the substrate-binding site (SBS-1) of CDK1 generated a binding-free energy of −9.3 Kcal/mol. Residues such as THR14, TYR15, VAL18, LYS33, GLN132, ASN133, ALA145, ASP146, and VAL165 surround the amino acid and THR14, GLN132, GLN132, ASN133, and ASP146 are involved in the hydrogen-bond interaction while the other surrounding amino acid residues are involved in hydrophobic interactions (Figure 9A). The docking simulation of EGFR inhibitor was performed with three compounds, including Afatinib, Erlotinib, and Gefitinib (Figure 9B–D). The highest affinity for substrate binding sites (SBS-2), with a binding free energy of −9.0 Kcal/mol, was found for Afatinib in the EGFR open conformation model, and binding-free energies of −8.5 Kcal/mol and −8.2 Kcal/mol were found for for Erlotinib and Gefitinib compounds in EGFR conformations respectively. Therefore, the chemical compound of Afatinib was strongly bound with EGFR conformation. LEU718, LYS745, MET793, CYS797, ARG841, ASN842, ASP855, and LEU858 are the surrounding residues for the Afatinib–EGFR complex. MET793, ASN842, ASP855, and LEU718 are involved in the hydrogen-bond interaction, while the other surrounding amino acids such as LYS745, LEU718, LEU858, and ARG841, CYS797, and ARG841 are involved in Pi–Cation, Alkyl, and Pi–Alkyl interactions respectively. The docking simulation of FYN inhibitor was performed with two compounds including Bosutinib and Dasatinib (Figure 9E,F). The highest affinity for SBS-3, with a binding-free energy of −7.1 Kcal/mol, was found for Bosutinib in FYN conformation, and a binding-free energy of −6.9 Kcal/mol was found for the Dasatinib—EGFR complex. Therefore, the compound of Bosutinib was strongly bound with the FYN conformation. Trp149, Tyr150, Arg176, Leu224, and Gln225 are surrounding residues for the Bosutinib–FYN complex. TRP149, GLN225, and ARG176 are involved in the hydrogen-bond interaction, while the other surrounding amino acids such as TRP149 and TRP149, and TRP149, TYR150, and LEU224 are involved in C-H and Pi-Orbital’s interactions respectively.
For the TWS-119–MYC complex, the interaction in SBS-4 of MYC generated a binding-free energy of −7.9 Kcal/mol. Residues such as Ser952, Val953, Glu956, Arg254, His258, and Gln261 are surrounding amino acids and GLN261, GLU956, and HIS258 are involved in the hydrogen-bond interaction while the other surrounding amino acid residues are involved in hydrophobic interactions (Figure 9G). The ultimate potential of the drugs with the molecular signatures of the NSCLC demanded close attention for experimental validation for developing effective and safe medications.

4. Discussion

Identification of disease-causing crucial biomarkers may shed light on a deeper understanding of the molecular mechanism of disease [58,59,60,61,62,63]. The present study was conducted to analyze the NSCLC gene expression data to determine the DEGs, extensive molecular pathways, significant hub proteins, and associated regulatory biomolecules in order to pick up the potential therapeutic targets for NSCLC through a multi-omics data integration framework. Through the gene expression patterns analysis, we identified 1943 DEGs, including 1367 upregulated and 576 downregulated genes. The functional enrichment analysis revealed that the proposed upregulated DEGs are significantly involved with some cancer-causing molecular functions and pathways, including cytokine–cytokine receptor interaction, chemokine signaling pathway, cell-adhesion molecules (CAMs), cAMP signaling pathway, MAPK signaling pathway, TNF signaling pathway, cGMP-PKG signaling pathway, Proteoglycans in cancer, and Rap1 signaling pathway (Figure 5). The downregulated genes are shared metabolic pathways, cell cycle, PI3K-Akt signaling pathway, focal adhesion, ECM-receptor interaction, p53 signaling pathway, and protein digestion and absorption pathways. All of these functions and pathways are significantly related to cancer development and play crucial roles in the NSCLC microenvironment. Recent studies indicated the importance of the tumor microenvironment as a decisive factor in tumorigenesis in various cancers [64,65,66,67,68]. Therefore, the physicochemical properties will be helpful to explore the further analysis of the reported proteins as a therapeutic target for NSCLC.
To detect the basic mechanism of disease, the protein–protein interaction network analysis is becoming a promising approach [69]. The PPI network analysis in this study revealed the hub-DEGs’ encoded hub-proteins. The CDK1 is related to the cell cycle activities. Up-regulation of CDK1 genes may be indicative of poor survival rates and a higher risk for cancer recurrence. The CDK1 gene is also related to several other cancer diseases [70,71]. The EGFR gene is associated with cell growth and had a contribution in lung cancer studied before [72,73]. The study revealed that the growth is suppressed and the radiosensitivity is amplified by the activities of ubiquitin C (UBC) in NSCLC cells [74,75,76,77]. The CDC6, CDC20, and CHEK1 genes are closely related to the occurrence and development of small-cell lung cancer, and CHEK1 is treated as a therapeutic target for lung cancer [78]. Eight hub genes (CDK1, EGFR, UBC, MYC, CCNB1, RHOB, CDC6, and CDC20) have tumor suppressor functions, while five hub genes (CDK1, EGFR, FYN, UBC, and CCNB1) are protein kinases as well. The MCODE cluster analysis clearly showed that the hub genes were distributed among the distinguished sub network (Figure 4) modules, which provided the strong evidence about the proposed signature biomolecules that these are reliable as therapeutic targets. Thus, the predicted hub-DEGs and relevant information might be useful in early detection of NSCLC. On the other hand, the genomic alteration/mutation analysis of the hub-DEGs reflected that most mutation for the EGFR occurred across the four lung cancer studies and was followed by the MYC and CHEK1 genes, since EGFR is a highly mutant/altered gene for lung cancer and NSCLC as well [79]. The alteration/mutation frequency revealed that the EGFR showed the highest alteration frequency relative to others, including the mutation, where CHEK1 represented mutation and deletion across the studies (Supplementary Figure S1), which may be a concern of investigation in future research.
The DEGs and hub-DEGs regulatory network analysis commonly revealed four transcription factors (FOXC1, GATA2, YY1, and NFIC) and five miRNAs (miR-335-5p, miR-26b-5p, miR-92a-3p, miR-155-5p, and miR-16-5p) as the key transcriptional and post-transcriptional regulators of DEGs as well as hub-DEGs. A study reported that various tumor-associated genes are regulated by FOXC1 and maintain several cancer-related pathways [80]. The GATA2 is treated as a therapeutic target in NSCLC treatment development and it also related to breast and kidney cancer [81,82]. The higher expression pattern of YY1 transcription factor triggered the patients having larger tumor size, differentiation, higher TNM stage, and lymph node metastasis [83]. The reported TFs are also involved in other cancer diseases [58,59,60,61,62,63]. In various types of cancer tissues, the miR-26b-5p acts as a tumor suppressor [84]. Currently, as one of the diagnostic tools for lung cancer identification, the miR-92a-3p expression measurement is being used [85,86]. The miR-155-5p is significantly associated with a higher risk for progression in adenocarcinoma patients [87,88] and miR-16-5p showed higher expression pattern in NSCLC cells [86].
The prognostic power of the reported biomolecules in discriminating the high- and low-risk conditions were exhibited by using the multivariate survival probability curves and box plots (Figure 8). The survival curves clearly demonstrated that the reported biomolecules played a significant role in patient survival. The box plot of the gene expression data of the molecular candidate also showed clear differences between the high- and low-risk groups (Figure 8B).
Finally, we selected the top-ranked five hub-DEGs-guided candidate drugs from the Connectivity Map (CMap) database (Table 3). Out of the selected drugs, we validated FDA approved six launched drugs (Dinaciclib, Afatinib, Icotinib, Bosutinib, Dasatinib, and TWS-119) by molecular docking simulation with the top-ranked five hub-DEGs-mediated target proteins for the treatment against NSCLC. The drug-target binding affinity scores (less than −7.0 Kcal/mol) suggested that the aforementioned six FDA approved launched drugs might be effective for the treatment against NSCLC. Thus, the findings of this study might be useful resources in prevention and early detection of NSCLC.

5. Conclusions

The current study focused on identifying the significant biomolecules along with their molecular mechanisms through integrative bioinformatics analysis. Among 1943 DEGs, 11 DEGs (CDK1, EGFR, FYN, UBC, MYC, CCNB1, FOS, RHOB, CDC6, CDC20, and CHEK1) were reported as the hub-DEGs/proteins that may play the key roles in NSCLC progression. The DEGs set enrichment analysis with the gene ontology (GO) database showed that DEGs are significantly involved with the cell adhesion, cell division, inflammatory response, signal transduction, protein binding, and plasma membrane extracellular region. The enrichment analysis with the KEGG pathway database showed that DEGs are significantly associated with the metabolic pathways, cell cycle, ECM-receptor interaction, and pathways in cancer. The inevitable regulatory TFs (FOXC1, GATA2, YY1, and FOXL1) and miRNA (miR-335-5p, miR-26b-5p, miR-92a-3p, miR-155-5pm and miR-16-5p) were identified as potential regulatory biomarkers for both DEGs and hub-DEGs. The strong prognostic performance of the reported biomolecules was observed between the high- and low-risk groups through the survival curves and box plots. The top-ranked hub-DEG-guided repurposable drug analysis revealed that the Dinaciclib, Afatinib, Icotinib, Bosutinib, Dasatiniband, and TWS-119 might be suggested as novel putative drugs for NSCLC treatment. The molecular docking analysis between the drug-target hub proteins and the repurposed drugs were conducted to investigate their molecular interaction mechanism. Thus, the findings of this study might be useful resources for NSCLC diagnosis, prognosis, and therapies, including gene-based DNA-vaccine development.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/vaccines10050771/s1, Figure S1: The genomic alteration of the proposed 11 hub-DEGs among the NSCLC.

Author Contributions

For this research article, M.P.M. and M.N.H.M. conceptualized the research design. M.P.M. collected the data and analyzed. M.S.R. did the molecular docking simulation part. M.P.M. and M.S.R. drafted the manuscript. R.A.M., E.G. and M.N.H.M. revised, edited the manuscript. M.N.H.M. supervised the research work. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors have declared no competing interests.

References

  1. Jiang, H.; Xu, A.; Li, M.; Han, R.; Wang, E.; Wu, D.; Fei, G.; Zhou, S.; Wang, R. Seven autophagy-related lncRNAs are associated with the tumor immune microenvironment in predicting survival risk of nonsmall cell lung cancer. Brief. Funct. Genom. 2021. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 145–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Qu, Y.; Emoto, K.; Eguchi, T.; Aly, R.G.; Zheng, H.; Chaft, J.E.; Tan, K.S.; Jones, D.R.; Kris, M.G.; Adusumilli, P.S.; et al. Pathologic Assessment after Neoadjuvant Chemotherapy for NSCLC: Importance and Implications of Distinguishing Adenocarcinoma From Squamous Cell Carcinoma. J. Thorac. Oncol. 2019, 14, 482–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Travis, W.D. Pathology of Lung Cancer. Clin. Chest Med. 2011, 32, 669–692. [Google Scholar] [CrossRef]
  5. Milovanovic, I.; Stjepanovic, M.; Mitrovic, D. Distribution patterns of the metastases of the lung carcinoma in relation to histological type of the primary tumor: An autopsy study. Ann. Thorac. Med. 2017, 12, 191–198. [Google Scholar] [CrossRef]
  6. D’Amico, T.A. Molecular Biologic Staging of Lung Cancer. Ann. Thorac. Surg. 2008, 85, S737–S742. [Google Scholar] [CrossRef]
  7. Alibolandi, M.; Ramezani, M.; Abnous, K.; Sadeghi, F.; Atyabi, F.; Asouri, M.; Ahmadi, A.A.; Hadizadeh, F. In vitro and in vivo evaluation of therapy targeting epithelial-cell adhesion-molecule aptamers for non-small cell lung cancer. J. Control. Release 2015, 209, 88–100. [Google Scholar] [CrossRef]
  8. Morgan, K.M.; Riedlinger, G.M.; Rosenfeld, J.; Ganesan, S.; Pine, S.R. Patient-derived xenograft models of non-small cell lung cancer and their potential utility in personalized medicine. Front. Oncol. 2017, 7, 2. [Google Scholar] [CrossRef] [Green Version]
  9. Desai, N.; Neyaz, A.; Szabolcs, A.; Shih, A.R.; Chen, J.H.; Thapar, V.; Nieman, L.T.; Solovyov, A.; Mehta, A.; Lieb, D.J.; et al. Temporal and spatial heterogeneity of host response to SARS-CoV-2 pulmonary infection. Nat. Commun. 2020, 11, 6319. [Google Scholar] [CrossRef]
  10. Kriegsmann, K.; Zgorzelski, C.; Muley, T.; Christopoulos, P.; Thomas, M.; Winter, H.; Eichhorn, M.; Eichhorn, F.; von Winterfeld, M.; Herpel, E.; et al. Role of Synaptophysin, Chromogranin and CD56 in adenocarcinoma and squamous cell carcinoma of the lung lacking morphological features of neuroendocrine differentiation: A retrospective large-scale study on 1170 tissue samples. BMC Cancer 2021, 21, 486. [Google Scholar] [CrossRef]
  11. Alberg, A.J.; Brock, M.V.; Ford, J.G.; Samet, J.M.; Spivack, S.D. Epidemiology of lung cancer: Diagnosis and management of lung cancer, 3rd ed.: American college of chest physicians evidence-based clinical practice guidelines. Chest 2013, 143, e1S–e29S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Cho, J.Y.; Kim, J.; Lee, J.S.; Kim, Y.J.; Kim, S.H.; Lee, Y.J.; Cho, Y.-J.; Yoon, H.I.; Lee, J.H.; Lee, C.-T.; et al. Characteristics, incidence, and risk factors of immune checkpoint inhibitor-related pneumonitis in patients with non-small cell lung cancer. Lung Cancer 2018, 125, 150–156. [Google Scholar] [CrossRef] [PubMed]
  13. Akhtar, N.; Bansal, J.G. Risk factors of Lung Cancer in nonsmoker. Curr. Probl. Cancer 2017, 41, 328–339. [Google Scholar] [CrossRef] [PubMed]
  14. Malhotra, J.; Malvezzi, M.; Negri, E.; La Vecchia, C.; Boffetta, P. Risk factors for lung cancer worldwide. Eur. Respir. J. 2016, 48, 889–902. Available online: https://erj.ersjournals.com/content/48/3/889 (accessed on 27 February 2022). [CrossRef] [PubMed] [Green Version]
  15. Zappa, C.; Mousa, S.A. Non-small cell lung cancer: Current treatment and future advances. Transl. Lung Cancer Res. 2016, 5, 288. Available online: https://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/pmc/articles/PMC4931124/ (accessed on 27 February 2022). [CrossRef] [Green Version]
  16. Reza, M.S.; Harun-Or-Roshid, M.; Islam, M.A.; Hossen, M.A.; Hossain, M.T.; Feng, S.; Xi, W.; Mollah, N.H.; Wei, Y. Bioinformatics Screening of Potential Biomarkers from mRNA Expression Profiles to Discover Drug Targets and Agents for Cervical Cancer. Int. J. Mol. Sci. 2022, 23, 3968. Available online: https://0-www-mdpi-com.brum.beds.ac.uk/1422-0067/23/7/3968/htm (accessed on 6 May 2022). [CrossRef]
  17. Alam, M.S.; Rahaman, M.M.; Sultana, A.; Wang, G.; Mollah, M.N.H. Statistics and network-based approaches to identify molecular mechanisms that drive the progression of breast cancer. Comput. Biol. Med. 2022, 145, 105508. Available online: https://pubmed.ncbi.nlm.nih.gov/35447458/ (accessed on 6 May 2022). [CrossRef]
  18. Delany, I.; Rappuoli, R.; De Gregorio, E. Vaccines for the 21st century. EMBO Mol. Med. 2014, 6, 708–720. [Google Scholar] [CrossRef]
  19. Okuda, K.; Wada, Y.; Shimada, M. Recent developments in preclinical DNA vaccination. Vaccines 2014, 2, 89–106. [Google Scholar] [CrossRef]
  20. Wahren, B.; Liu, M.A. Dna vaccines: Recent developments and the future. Vaccines 2014, 2, 785–796. [Google Scholar] [CrossRef] [Green Version]
  21. Nakayama, Y.; Aruga, A. Comparison of Current Regulatory Status for Gene-Based Vaccines in the U.S.; Europe and Japan. Vaccines 2015, 3, 186–202. Available online: https://0-www-mdpi-com.brum.beds.ac.uk/2076-393X/3/1/186/htm (accessed on 6 May 2022). [CrossRef] [PubMed] [Green Version]
  22. Ulmer, J.B.; Wahren, B.; Liu, M.A. Gene-based vaccines: Recent technical and clinical advances. Trends Mol. Med. 2006, 12, 216–222. [Google Scholar] [CrossRef] [PubMed]
  23. Xu, H.; Huang, J.; Hua, S.; Liang, L.; He, X.; Zhan, M.; Lu, L.; Chu, J. Interactome analysis of gene expression profiles identifies CDC6 as a potential therapeutic target modified by miR-215-5p in hepatocellular carcinoma. Int. J. Med. Sci. 2020, 17, 2926–2940. [Google Scholar] [CrossRef] [PubMed]
  24. Ennishi, D.; Takata, K.; Béguelin, W.; Duns, G.; Mottok, A.; Farinha, P.; Bashashati, A.; Saberi, S.; Boyle, M.; Meissner, B.; et al. Molecular and genetic characterization of MHC deficiency identifies ezh2 as therapeutic target for enhancing immune recognition. Cancer Discov. 2019, 9, 546–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Xu, J.; Zhu, C.; Yu, Y.; Wu, W.; Cao, J.; Li, Z.; Dai, J.; Wang, C.; Tang, Y.; Zhu, Q.; et al. Systematic cancer-testis gene expression analysis identified CDCA5 as a potential therapeutic target in esophageal squamous cell carcinoma. eBioMedicine 2019, 46, 54–65. [Google Scholar] [CrossRef] [Green Version]
  26. Wei, Z.; Liu, Y.; Qiao, S.; Li, X.; Li, Q.; Zhao, J.; Hu, J.; Wei, Z.; Shan, A.; Sun, X.; et al. Identification of the potential therapeutic target gene ube2c in human hepatocellular carcinoma: An investigation based on geo and tcga databases. Oncol. Lett. 2019, 17, 5409–5418. [Google Scholar] [CrossRef] [Green Version]
  27. Islam, T.; Rahman, R.; Gov, E.; Turanli, B.; Gulfidan, G.; Haque, A.; Arga, K.Y.; Mollah, N.H. Drug Targeting and Biomarkers in Head and Neck Cancers: Insights from Systems Biology Analyses. Omics J. Integr. Biol. 2018, 22, 422–436. [Google Scholar] [CrossRef]
  28. Mosharaf, M.P.; Reza, M.S.; Kibria, M.K.; Ahmed, F.F.; Kabir, M.H.; Hasan, S.; Mollah, N.H. Computational identification of host genomic biomarkers highlighting their functions, pathways and regulators that influence SARS-CoV-2 infections and drug repurposing. Sci. Rep. 2022, 12, 4279. Available online: https://0-www-nature-com.brum.beds.ac.uk/articles/s41598-022-08073-8 (accessed on 21 April 2022). [CrossRef]
  29. Faysal Ahmed, F.; Selim Reza, M.; Shahin Sarker, M.; Samiul Islam, M.I.; Parvez Mosharaf, M.; Hasan, S.; Mollah, N.H. Identification of host transcriptome-guided repurposable drugs for SARS-CoV-1 infections and their validation with SARS-CoV-2 infections by using the integrated bioinformatics approaches. PLoS ONE 2022, 17, e0266124. Available online: https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0266124 (accessed on 21 April 2022).
  30. Barrett, T.; Wilhite, S.E.; Ledoux, P.; Evangelista, C.; Kim, I.F.; Tomashevsky, M.; Marshall, K.A.; Phillippy, K.H.; Sherman, P.M.; Holko, M.; et al. NCBI GEO: Archive for functional genomics data sets—Update. Nucleic Acids Res. 2013, 41, D991–D995. [Google Scholar] [CrossRef] [Green Version]
  31. Lu, T.P.; Tsai, M.H.; Lee, J.M.; Hsu, C.P.; Chen, P.C.; Lin, C.W.; Shih, J.Y.; Yang, P.C.; Hsiao, C.K.; Lai, L.C.; et al. Identification of a novel biomarker, SEMA5A, for non-small cell lung carcinoma in nonsmoking women. Cancer Epidemiol. Biomarkers Prev. 2010, 19, 2590–2597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Gentleman, R.C.; Carey, V.J.; Bates, D.M.; Bolstad, B.; Dettling, M.; Dudoit, S.; Ellis, B.; Gautier, L.; Ge, Y.; Gentry, J.; et al. Bioconductor: Open software development for computational biology and bioinformatics. Genome Biol. 2004, 5, R80. [Google Scholar] [CrossRef] [Green Version]
  33. Benjamini, Y.; Hochberg, Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J. R. Stat. Soc. Ser. B 1995, 57, 289–300. [Google Scholar] [CrossRef]
  34. Huang, D.W.; Sherman, B.T.; Lempicki, R.A. Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009, 37, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Huang, D.W.; Sherman, B.T.; Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2009, 4, 44–57. [Google Scholar] [CrossRef] [PubMed]
  36. Kanehisa, M.; Furumichi, M.; Tanabe, M.; Sato, Y.; Morishima, K. KEGG: New perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017, 45, D353–D361. Available online: https://pubmed.ncbi.nlm.nih.gov/27899662/ (accessed on 8 October 2021). [CrossRef] [Green Version]
  37. Kanehisa, M.; Sato, Y.; Kawashima, M.; Furumichi, M.; Tanabe, M. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res. 2016, 44, D457–D462. [Google Scholar] [CrossRef] [Green Version]
  38. Kanehisa, M.; Goto, S. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res. 2000, 28, 27–30. [Google Scholar] [CrossRef]
  39. Szklarczyk, D.; Morris, J.H.; Cook, H.; Kuhn, M.; Wyder, S.; Simonovic, M.; Santos, A.; Doncheva, N.T.; Roth, A.; Bork, P.; et al. The STRING database in 2017: Quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017, 45, D362–D368. [Google Scholar] [CrossRef]
  40. Li, X.; Li, W.; Zeng, M.; Zheng, R.; Li, M. Network-based methods for predicting essential genes or proteins: A survey. Brief. Bioinform. 2020, 21, 566–583. [Google Scholar] [CrossRef]
  41. Xia, J.; Gill, E.E.; Hancock, R.E.W. NetworkAnalyst for statistical, visual and network-based meta-analysis of gene expression data. Nat. Protoc. 2015, 10, 823–844. [Google Scholar] [CrossRef] [PubMed]
  42. Chin, C.H.; Chen, S.H.; Wu, H.H.; Ho, C.W.; Ko, M.T.; Lin, C.Y. cytoHubba: Identifying hub objects and sub-networks from complex interactome. BMC Syst. Biol. 2014, 8, S11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Calimlioglu, B.; Karagoz, K.; Sevimoglu, T.; Kilic, E.; Gov, E.; Arga, K.Y. Tissue-Specific Molecular Biomarker Signatures of Type 2 Diabetes: An Integrative Analysis of Transcriptomics and Protein-Protein Interaction Data. Omics J. Integr. Biol. 2015, 19, 563–573. [Google Scholar] [CrossRef] [PubMed]
  44. Bandettini, W.P.; Kellman, P.; Mancini, C.; Booker, O.J.; Vasu, S.; Leung, S.W.; Wilson, J.R.; Shanbhag, S.M.; Chen, M.Y.; Arai, A.E. MultiContrast Delayed Enhancement (MCODE) improves detection of subendocardial myocardial infarction by late gadolinium enhancement cardiovascular magnetic resonance: A clinical validation study. J. Cardiovasc. Magn. Reson. 2012, 14, 83. Available online: https://pubmed.ncbi.nlm.nih.gov/23199362/ (accessed on 20 September 2021). [CrossRef] [Green Version]
  45. Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 2013, 6, pl1. Available online: https://pubmed.ncbi.nlm.nih.gov/23550210/ (accessed on 6 May 2022). [CrossRef] [Green Version]
  46. Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.O.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E.; et al. The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data. Cancer Discov. 2012, 2, 401–404. Available online: https://aacrjournals.org/cancerdiscovery/article/2/5/401/3246/The-cBio-Cancer-Genomics-Portal-An-Open-Platform (accessed on 6 May 2022). [CrossRef] [Green Version]
  47. Karagkouni, D.; Paraskevopoulou, M.D.; Chatzopoulos, S.; Vlachos, I.S.; Tastsoglou, S.; Kanellos, I.; Papadimitriou, D.; Kavakiotis, I.; Maniou, S.; Skoufos, G.; et al. DIANA-TarBase v8: A decade-long collection of experimentally supported miRNA-gene interactions. Nucleic Acids Res. 2018, 46, D239–D245. [Google Scholar] [CrossRef] [Green Version]
  48. Chou, C.H.; Chang, N.W.; Shrestha, S.; Hsu, S.D.; Lin, Y.L.; Lee, W.H.; Yang, C.D.; Hong, H.C.; Wei, T.Y.; Tu, S.J.; et al. miRTarBase 2016: Updates to the experimentally validated miRNA-target interactions database. Nucleic Acids Res. 2016, 44, D239–D247. Available online: https://pubmed.ncbi.nlm.nih.gov/26590260/ (accessed on 20 September 2021). [CrossRef]
  49. Khan, A.; Fornes, O.; Stigliani, A.; Gheorghe, M.; Castro-Mondragon, J.A.; Van Der Lee, R.; Bessy, A.; Cheneby, J.; Kulkarni, S.R.; Tan, G.; et al. JASPAR 2018: Update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res. 2018, 46, D260–D266. [Google Scholar] [CrossRef] [Green Version]
  50. Aguirre-Gamboa, R.; Gomez-Rueda, H.; Martínez-Ledesma, E.; Martínez-Torteya, A.; Chacolla-Huaringa, R.; Rodriguez-Barrientos, A.; Tamez-Pena, J.G.; Treviño, V. SurvExpress: An Online Biomarker Validation Tool and Database for Cancer Gene Expression Data Using Survival Analysis. PLoS ONE 2013, 8, e74250. [Google Scholar] [CrossRef] [Green Version]
  51. Lamb, J.; Crawford, E.D.; Peck, D.; Modell, J.W.; Blat, I.C.; Wrobel, M.J.; Lerner, J.; Brunet, J.-P.; Subramanian, A.; Ross, K.N.; et al. The connectivity map: Using gene-expression signatures to connect small molecules, genes, and disease. Science 2006, 313, 1929–1935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Meng, X.-Y.; Zhang, H.-X.; Mezei, M.; Cui, M. Molecular Docking: A Powerful Approach for Structure-Based Drug Discovery. Curr. Comput. Aided-Drug Des. 2012, 7, 146–157. [Google Scholar] [CrossRef] [PubMed]
  53. Halgren, T.A. Merck molecular force field. I. Basis, form, scope, parameterization, and performance of MMFF94. J. Comput. Chem. 1996, 17, 490–519. [Google Scholar] [CrossRef]
  54. Wass, M.N.; Kelley, L.A.; Sternberg, M.J.E. 3DLigandSite: Predicting ligand-binding sites using similar structures. Nucleic Acids Res. 2010, 38, W469–W473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J. Comput. Chem. 2009, 30, 2785–2791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Oleg, T.; Arthur, J.O. AutoDock Vina: Improving the Speed and Accuracy of Docking with a New Scoring Function, Efficient Optimization, and Multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar]
  57. Accelrys Software Inc. Visualizer DS, Version 4. 0. 100. 13345; Accelrys Software Inc.: San Diego, CA, USA, 2016. [Google Scholar]
  58. Rahman, R.; Islam, T.; Gov, E.; Turanli, B.; Gulfidan, G.; Shahjaman, M.; Banu, N.A.; Mollah, N.H.; Arga, K.Y.; Moni, M.A. Identification of prognostic biomarker signatures and candidate drugs in colorectal cancer: Insights from systems biology analysis. Medicina 2019, 55, 20. [Google Scholar] [CrossRef] [Green Version]
  59. Islam, T.; Rahman, M.R.; Aydin, B.; Beklen, H.; Arga, K.Y.; Shahjaman, M. Integrative transcriptomics analysis of lung epithelial cells and identification of repurposable drug candidates for COVID-19. Eur. J. Pharmacol. 2020, 887, 173594. [Google Scholar] [CrossRef]
  60. Rahman, M.R.; Islam, T.; Turanli, B.; Zaman, T.; Faruquee, H.M.; Rahman, M.M.; Mollah, M.N.H.; Nanda, R.K.; Arga, K.Y.; Gov, E.; et al. Network-based approach to identify molecular signatures and therapeutic agents in Alzheimer’s disease. Comput. Biol. Chem. 2019, 78, 431–439. [Google Scholar] [CrossRef]
  61. Moni, M.A.; Islam, M.B.; Rahman, M.R.; Rashed-Al-Mahfuz, M.; Awal, M.A.; Islam, S.M.S.; Mollah, N.H.; Quinn, M.W. Network-Based Computational Approach to Identify Delineating Common Cell Pathways Influencing Type 2 Diabetes and Diseases of Bone and Joints. IEEE Access 2020, 8, 1486–1497. [Google Scholar] [CrossRef]
  62. Satu, S.; Khan, I.; Rahman, R.; Howlader, K.C.; Roy, S.; Roy, S.S.; Quinn, J.M.W.; Ali Moni, M. Diseasome and comorbidities complexities of SARS-CoV-2 infection with common malignant diseases. Brief Bioinform. 2021, 22, 1415–1429. [Google Scholar] [CrossRef] [PubMed]
  63. Shahjaman, M.; Rezanur Rahman, M.; Shahinul Islam, S.M.; Nurul Haque Mollah, M. A robust approach for identification of cancer biomarkers and candidate drugs. Medicina 2019, 55, 269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Dzobo, K.; Senthebane, D.A.; Rowe, A.; Thomford, N.E.; Mwapagha, L.; Al-Awwad, N.; Dandara, C.; Parker, M.I. Cancer Stem Cell Hypothesis for Therapeutic Innovation in Clinical Oncology? Taking the Root Out, Not Chopping the Leaf. Omics J. Integr. Biol. 2016, 20, 681–691. [Google Scholar] [CrossRef]
  65. Gollapalli, K.; Ghantasala, S.; Atak, A.; Rapole, S.; Moiyadi, A.; Epari, S.; Srivastava, S. Tissue Proteome Analysis of Different Grades of Human Gliomas Provides Major Cues for Glioma Pathogenesis. Omics J. Integr. Biol. 2017, 21, 275–284. [Google Scholar] [CrossRef] [PubMed]
  66. Gov, E.; Kori, M.; Arga, K.Y. Multiomics Analysis of Tumor Microenvironment Reveals Gata2 and miRNA-124-3p as Potential Novel Biomarkers in Ovarian Cancer. Omics J. Integr. Biol. 2017, 21, 603–615. [Google Scholar] [CrossRef] [PubMed]
  67. Hu, M.; Qian, C.; Hu, Z.; Fei, B.; Zhou, H. Biomarkers in tumor microenvironment? Upregulation of fibroblast activation protein-α correlates with gastric cancer progression and poor prognosis. Omics J. Integr. Biol. 2017, 21, 38–44. [Google Scholar] [CrossRef]
  68. Miskolczi, Z.; Smith, M.P.; Rowling, E.J.; Ferguson, J.; Barriuso, J.; Wellbrock, C. Collagen abundance controls melanoma phenotypes through lineage-specific microenvironment sensing. Oncogene 2018, 37, 3166–3182. [Google Scholar] [CrossRef] [Green Version]
  69. Sevimoglu, T.; Arga, K.Y. The role of protein interaction networks in systems biomedicine. Comput. Struct. Biotechnol. J. 2014, 11, 22–27. [Google Scholar] [CrossRef] [Green Version]
  70. Ni, Z.; Wang, X.; Zhang, T.; Li, L.; Li, J. Comprehensive analysis of differential expression profiles reveals potential biomarkers associated with the cell cycle and regulated by p53 in human small cell lung cancer. Exp. Ther. Med. 2018, 15, 3273–3782. [Google Scholar] [CrossRef] [Green Version]
  71. Shi, Y.-X.; Zhu, T.; Zou, T.; Zhuo, W.; Chen, Y.-X.; Huang, M.-S.; Zheng, W.; Wang, C.-J.; Li, X.; Mao, X.-Y.; et al. Prognostic and predictive values of CDK1 and MAD2L1 in lung adenocarcinoma. Oncotarget 2016, 7, 85235–85243. [Google Scholar] [CrossRef] [Green Version]
  72. Bethune, G.; Bethune, D.; Ridgway, N.; Xu, Z. Epidermal growth factor receptor (EGFR) in lung cancer: An overview and update. J. Thorac. Dis. 2010, 2, 48–51. [Google Scholar] [PubMed]
  73. Sousa, A.; Silveira, C.; Janeiro, A.; Malveiro, S.; Oliveira, A.R.; Felizardo, M.; Nogueira, F.; Teixeira, E.; Martins, J.; Carmo-Fonseca, M. Detection of rare and novel EGFR mutations in NSCLC patients: Implications for treatment-decision. Lung Cancer 2020, 139, 35–40. [Google Scholar] [CrossRef] [PubMed]
  74. Tang, Y.; Geng, Y.; Luo, J.; Shen, W.; Zhu, W.; Meng, C.; Li, M.; Zhou, X.; Zhang, S.; Cao, J. Downregulation of ubiquitin inhibits the proliferation and radioresistance of non-small cell lung cancer cells in vitro and in vivo. Sci. Rep. 2015, 5, 9476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Zou, Y.; Jing, L. Identification of key modules and prognostic markers in adrenocortical carcinoma by weighted gene co-expression network analysis. Oncol. Lett. 2019, 18, 3673–3681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Mohammed, A.; Abdulhadi, S. Evaluation of the Concentration of Ubiquitin C Protein (UBC) in Patients of Lung Cancer and Comparing with Healthy Subjects. Eng. Technol. J. 2020, 38, 61–65. [Google Scholar] [CrossRef]
  77. Hao, S.; Li, S.; Wang, J.; Zhao, L.; Yan, Y.; Cao, Q.; Wu, T.; Liu, L.; Wang, C. Transcriptome analysis of phycocyanin-mediated inhibitory functions on non-small cell lung cancer A549 cell growth. Mar. Drugs 2018, 16, 511. [Google Scholar] [CrossRef] [Green Version]
  78. Wen, P.; Chidanguro, T.; Shi, Z.; Gu, H.; Wang, N.; Wang, T.; Li, Y.; Gao, J. Identification of candidate biomarkers and pathways associated with SCLC by bioinformatics analysis. Mol. Med. Rep. 2018, 18, 1538–1550. [Google Scholar] [CrossRef] [Green Version]
  79. Chinnappan, J.; Ramu, A.; Vidhya Rajalakshmi, V.; Akil Kavya, S. Integrative Bioinformatics approaches to therapeutic gene target selection in various cancers for Nitroglycerin. Sci. Rep. 2021, 11, 22036. [Google Scholar] [CrossRef]
  80. Yang, Z.; Jiang, S.; Cheng, Y.; Li, T.; Hu, W.; Ma, Z.; Chen, F.; Yang, Y. FOXC1 in cancer development and therapy: Deciphering its emerging and divergent roles. Ther. Adv. Med. Oncol. 2017, 9, 797–816. [Google Scholar] [CrossRef]
  81. Tessema, M.; Yingling, C.M.; Snider, A.M.; Do, K.; Juri, D.E.; Picchi, M.A.; Zhang, X.; Liu, Y.; Leng, S.; Tellez, C.S.; et al. GATA2 is epigenetically repressed in human and mouse lung tumors and is not requisite for survival of KRAS mutant lung cancer. J. Thorac. Oncol. 2014, 9, 784–793. [Google Scholar] [CrossRef] [Green Version]
  82. Yu, S.; Jiang, X.; Li, J.; Li, C.; Guo, M.; Ye, F.; Zhang, M.; Jiao, Y.; Guo, B. Comprehensive analysis of the GATA transcription factor gene family in breast carcinoma using gene microarrays, online databases and integrated bioinformatics. Sci. Rep. 2019, 9, 4467. [Google Scholar] [CrossRef] [PubMed]
  83. Huang, T.; Wang, G.; Yang, L.; Peng, B.; Wen, Y.; Ding, G.; Wang, Z. Transcription Factor YY1 Modulates Lung Cancer Progression by Activating lncRNA-PVT1. DNA Cell Biol. 2017, 36, 947–958. [Google Scholar] [CrossRef] [PubMed]
  84. Miyamoto, K.; Seki, N.; Matsushita, R.; Yonemori, M.; Yoshino, H.; Nakagawa, M.; Enokida, H. Tumour-suppressive miRNA-26a-5p and miR-26b-5p inhibit cell aggressiveness by regulating PLOD2 in bladder cancer. Br. J. Cancer 2016, 115, 354–363. [Google Scholar] [CrossRef] [Green Version]
  85. Wu, K.L.; Tsai, Y.M.; Lien, C.T.; Kuo, P.L.; Hung, J.Y. The roles of microRNA in lung cancer. Int. J. Mol. Sci. 2019, 20, 1611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Fan, L.; Sha, J.; Teng, J.; Li, D.; Wang, C.; Xia, Q.; Chen, H.; Su, B.; Qi, H. Evaluation of Serum Paired MicroRNA Ratios for Differential Diagnosis of Non-Small Cell Lung Cancer and Benign Pulmonary Diseases. Mol. Diagn. Ther. 2018, 22, 493–502. [Google Scholar] [CrossRef]
  87. Sanfiorenzo, C.; Ilie, M.I.; Belaid, A.; Barlési, F.; Mouroux, J.; Marquette, C.-H.; Brest, P.; Hofman, P. Two Panels of Plasma MicroRNAs as Non-Invasive Biomarkers for Prediction of Recurrence in Resectable NSCLC. PLoS ONE. 2013, 8, e54596. [Google Scholar] [CrossRef] [Green Version]
  88. Pasculli, B.; Barbano, R.; Fontana, A.; Biagini, T.; Di Viesti, M.P.; Rendina, M.; Valori, V.M.; Morritti, M.; Bravaccini, S.; Ravaioli, S.; et al. Hsa-miR-155-5p Up-Regulation in Breast Cancer and Its Relevance for Treatment with Poly[ADP-Ribose] Polymerase 1 (PARP-1) Inhibitors. Front. Oncol. 2020, 10, 1415. [Google Scholar] [CrossRef]
Figure 1. The schematic diagram of the integrative bioinformatics analysis of this study.
Figure 1. The schematic diagram of the integrative bioinformatics analysis of this study.
Vaccines 10 00771 g001
Figure 2. Gene expression profile of microarray data. (A) The volcano plot which represents the scatter plot of log2FC values versus −log10(adjusted p-values). (B) The volcano plot highlighting DEGs, where green bullets represent the upregulated (adjusted p-value < 0.001 and log2FC > 1) and downregulated (adjusted p-value < 0.001 and log2FC < −1) DEGs selected based on the described criteria.
Figure 2. Gene expression profile of microarray data. (A) The volcano plot which represents the scatter plot of log2FC values versus −log10(adjusted p-values). (B) The volcano plot highlighting DEGs, where green bullets represent the upregulated (adjusted p-value < 0.001 and log2FC > 1) and downregulated (adjusted p-value < 0.001 and log2FC < −1) DEGs selected based on the described criteria.
Vaccines 10 00771 g002
Figure 3. NSCLC-specific protein–protein interaction network. The redder color represents the higher degree measured by CytoHubba. The hub-DEGs are represented only with the different colors in the PPI. Green nodes represent the associated proteins.
Figure 3. NSCLC-specific protein–protein interaction network. The redder color represents the higher degree measured by CytoHubba. The hub-DEGs are represented only with the different colors in the PPI. Green nodes represent the associated proteins.
Vaccines 10 00771 g003
Figure 4. The first four sub networks based on score, identified by the MCODE algorithm. The scores of 6.071, 3.76, 3.684, and 3.4 were exhibited by the (A) first, (B) second, (C) third, and (D) forth sub modules, respectively.
Figure 4. The first four sub networks based on score, identified by the MCODE algorithm. The scores of 6.071, 3.76, 3.684, and 3.4 were exhibited by the (A) first, (B) second, (C) third, and (D) forth sub modules, respectively.
Vaccines 10 00771 g004
Figure 5. The KEGG pathways (A) for upregulated DEGs and (B) downregulated DEGs.
Figure 5. The KEGG pathways (A) for upregulated DEGs and (B) downregulated DEGs.
Vaccines 10 00771 g005
Figure 6. (A) The TFs-DEGs interaction network and (B) the miRNA-DEGs interaction network. The TFs and miRNAs are marked as blue-shape square in the interactions. The larger square means a higher degree of connectivity among the nodes. The circle-shaped nodes represent the DE genes.
Figure 6. (A) The TFs-DEGs interaction network and (B) the miRNA-DEGs interaction network. The TFs and miRNAs are marked as blue-shape square in the interactions. The larger square means a higher degree of connectivity among the nodes. The circle-shaped nodes represent the DE genes.
Vaccines 10 00771 g006
Figure 7. (A) The hub proteins–TFs interaction network, and the TFs are marked as blue-shaped square in the interactions. (B) The hub proteins–miRNA interaction network, and the hub proteins are marked as red circles in interaction network. The larger significant miRNAs are labeled and marked as pink-colored circles.
Figure 7. (A) The hub proteins–TFs interaction network, and the TFs are marked as blue-shaped square in the interactions. (B) The hub proteins–miRNA interaction network, and the hub proteins are marked as red circles in interaction network. The larger significant miRNAs are labeled and marked as pink-colored circles.
Vaccines 10 00771 g007
Figure 8. The risk group discrimination performance by the multivariate survival probability curves (left) and box plots (right) based on (A) hub-DEGs/proteins and (B) key TFs (transcription factors) proteins.
Figure 8. The risk group discrimination performance by the multivariate survival probability curves (left) and box plots (right) based on (A) hub-DEGs/proteins and (B) key TFs (transcription factors) proteins.
Vaccines 10 00771 g008
Figure 9. The molecular docking poses for the selected repurposed drugs and potential target proteins. The figure showed the best docking pose between protein and drug, like in (A) between CDK1-Dinaciclib; in (B) between EGFR-Afatinib; in (C) between EGFR-Erlotinib; in (D) between EGFR-Gefitinib; in (E) between FYN-Bosutinib; in (F) between FYN-Dasatinib and in (G) between MYC-TWS119 respectively.
Figure 9. The molecular docking poses for the selected repurposed drugs and potential target proteins. The figure showed the best docking pose between protein and drug, like in (A) between CDK1-Dinaciclib; in (B) between EGFR-Afatinib; in (C) between EGFR-Erlotinib; in (D) between EGFR-Gefitinib; in (E) between FYN-Bosutinib; in (F) between FYN-Dasatinib and in (G) between MYC-TWS119 respectively.
Vaccines 10 00771 g009
Table 1. The physicochemical properties of the reported hub proteins.
Table 1. The physicochemical properties of the reported hub proteins.
Hub Protein’s NameNumber of Amino AcidsMolecular Weight (kda)Theoretical pINumber of Negatively Charged Residues (Asp + Glu)Number of Positively Charged Residues (Arg + Lys)* Extinction CoefficientInstability IndexAliphatic IndexGrand Average of Hydropathicity (GRAVY)
CDK129734,095.458.38373942,86039.2697.78−0.281
EGFR1210134,277.46.26138126128,89044.5980.74−0.316
FYN53760,761.96.23686394,24036.4175.36−0.489
UBC15818,006.828.87182229,70045.7872.91−0.533
MYC43948,804.085.33645129,50592.2366.42−0.772
CCNB143348,337.437.09525230,62050.5990.09−0.239
FOS38040,695.414.77513321,93078.8265.32−0.369
RHOB19622,123.395.1322621,93046.3587.96−0.26
CDC656062,720.289.64589120,94048.5794.89−0.383
CDC2049954,722.599.334254106,25547.7276.31−0.483
CHEK147654,433.578.5616676,48542.2684.75−0.459
Note: * Extinction coefficients are in units of M−1 cm−1, at 280 nm measured in water.
Table 2. The functional enrichment analysis of the DEGs to clarify the gene ontology terms in the NSCLC disease. The top GO terms are summarized and presented here.
Table 2. The functional enrichment analysis of the DEGs to clarify the gene ontology terms in the NSCLC disease. The top GO terms are summarized and presented here.
Upregulated Genes
GO TermNumber of GenesCoverage (%)p-Value
GOTERM_BP_DIRECT
GO:0001525 angiogenesis404.271.77 × 10−12
GO:0007155 cell adhesion596.31.28 × 10−11
GO:0006954 inflammatory response505.32.33 × 10−10
GO:0007166 cell-surface receptor signaling pathway414.42.97 × 10−10
GO:0006955 immune response495.22.29 × 10−8
GO:0032496 response to lipopolysaccharide262.82.80 × 10−7
GO:0006935 chemotaxis222.33.17 × 10−7
GO:0007165 signal transduction9410.05.91 × 10−7
GOTERM_CC_DIRECT
GO:0005886 plasma membrane29531.58.30 ×10−16
GO:0005576 extracellular region14515.51.69 ×10−14
GO:0005615 extracellular space12713.53.91× 10−14
GO:0045121 membrane raft343.66.18 × 10−10
GO:0070062 extracellular exosome18519.79.29 × 10−7
GO:0009986 cell surface525.52.02 × 10−6
GO:0005925 focal adhesion414.43.45 × 10−6
GO:0016021 integral component of membrane29731.72.91 × 10−5
GOTERM_MF_DIRECT
GO:0008201 heparin binding293.11.15 × 10−9
GO:0030246 carbohydrate binding272.91.36 × 10−6
GO:0005178 integrin binding192.01.46 × 10−6
GO:0005509 calcium ion binding596.32.60 × 10−5
GO:0051015 actin filament binding192.03.86 × 10−5
GO:0004872 receptor activity252.77.30 × 10−5
GO:0005515 protein binding46049.18.91 × 10−5
GO:0003779 actin binding283.02.41 × 10−4
Down Regulated Genes
GO TermNumber of GenesCoverage (%)p-Value
GOTERM_BP_DIRECT
GO:0030574collagen catabolic process153.41.70 × 10−10
GO:0007067mitotic nuclear division265.97.35 × 10−10
GO:0051301cell division296.51.30 × 10−8
GO:0007062sister chromatid cohesion143.27.36 × 10−7
GO:0030198extracellular matrix organization194.37.37 × 10−7
GO:0000082G1/S transition of mitotic cell cycle133.04.17 × 10−6
GO:0030199collagen fibril organization81.82.75 × 10−5
GO:0001649osteoblast differentiation122.72.90 × 10−5
GO:0000281mitotic cytokinesis71.64.50 × 10−5
GO:0006508proteolysis276.11.12 × 10−4
GOTERM_CC_DIRECT
GO:0005615extracellular space6314.25.08 × 10−8
GO:0070062extracellular exosome10122.81.18 × 10−6
GO:0005578proteinaceous extracellular matrix214.73.05 × 10−6
GO:0000777condensed chromosome kinetochore122.73.95 × 10−6
GO:0005581collagen trimer122.76.85 × 10−6
GO:0030496midbody143.26.95 × 10−6
GO:0005576extracellular region6414.41.01 × 10−5
GO:0005819spindle122.79.10 × 10−5
GOTERM_MF_DIRECT
GO:0004222metalloendopeptidase activity132.97.55 × 10−6
GO:0004252serine-type endopeptidase activity194.31.56 × 10−5
GO:0005201extracellular matrix structural constituent102.21.57 × 10−5
GO:0042802identical protein binding327.26.18 × 10−4
GO:0019901protein kinase binding194.30.0019
GO:0005524ATP binding5111.50.0021
Table 3. The repurposed drugs that were found from the CMap database.
Table 3. The repurposed drugs that were found from the CMap database.
Target ProteinsName of DrugMechanism of ActionPhase
CDK1aminopurvalanol-aCDK inhibitor, tyrosine kinase inhibitorPre-clinical
BMS-265246CDK inhibitorPre-clinical
CDK1-5-inhibitorCDK inhibitor, glycogen synthase kinase inhibitorPre-clinical
CGP-60474CDK inhibitorPre-clinical
CGP-74514CDK inhibitorPre-clinical
CHIR-99021glycogen synthase kinase inhibitorPre-clinical
dinaciclibCDK inhibitorPhase 3
indirubin-3-monoximeCDK inhibitor, glycogen synthase kinase inhibitorPre-clinical
JNJ-7706621CDK inhibitorPre-clinical
kenpaulloneCDK inhibitor, glycogen synthase kinase inhibitorPre-clinical
olomoucineCDK inhibitorPre-clinical
PF-573228focal adhesion kinase inhibitorPre-clinical
PHA-767491CDC inhibitorPre-clinical
purvalanol-aCDK inhibitorPre-clinical
Ro-3306CDK inhibitorPre-clinical
SU9516CDK inhibitorPre-clinical
1-azakenpaulloneglycogen synthase kinase inhibitorPre-clinical
8-hydroxy-DPATserotonin receptor agonistPre-clinical
EGFRafatinibEGFR inhibitorLaunched
brigatinibALK tyrosine kinase receptor inhibitor, EGFR inhibitorLaunched
erlotinibEGFR inhibitorLaunched
gefitinibEGFR inhibitorLaunched
icotinibEGFR inhibitorLaunched
lapatinibEGFR inhibitorLaunched
lidocainehistamine receptor agonistLaunched
olmutinibEGFR inhibitor, Bruton’s tyrosine kinase (BTK) inhibitorLaunched
osimertinibEGFR inhibitorLaunched
vandetanibEGFR inhibitor, RET tyrosine kinase inhibitor, VEGFR inhibitorLaunched
FYNbosutinibAbl kinase inhibitor, Bcr-Abl kinase inhibitor, src inhibitorLaunched
dasatinibBcr-Abl kinase inhibitor, ephrin inhibitor, KIT inhibitor, PDGFR tyrosine kinase receptor inhibitor, src inhibitor, tyrosine kinase inhibitorLaunched
MYCTWS-119glycogen synthase kinase inhibitorPre-clinical
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mosharaf, M.P.; Reza, M.S.; Gov, E.; Mahumud, R.A.; Mollah, M.N.H. Disclosing Potential Key Genes, Therapeutic Targets and Agents for Non-Small Cell Lung Cancer: Evidence from Integrative Bioinformatics Analysis. Vaccines 2022, 10, 771. https://0-doi-org.brum.beds.ac.uk/10.3390/vaccines10050771

AMA Style

Mosharaf MP, Reza MS, Gov E, Mahumud RA, Mollah MNH. Disclosing Potential Key Genes, Therapeutic Targets and Agents for Non-Small Cell Lung Cancer: Evidence from Integrative Bioinformatics Analysis. Vaccines. 2022; 10(5):771. https://0-doi-org.brum.beds.ac.uk/10.3390/vaccines10050771

Chicago/Turabian Style

Mosharaf, Md. Parvez, Md. Selim Reza, Esra Gov, Rashidul Alam Mahumud, and Md. Nurul Haque Mollah. 2022. "Disclosing Potential Key Genes, Therapeutic Targets and Agents for Non-Small Cell Lung Cancer: Evidence from Integrative Bioinformatics Analysis" Vaccines 10, no. 5: 771. https://0-doi-org.brum.beds.ac.uk/10.3390/vaccines10050771

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