Next Article in Journal
The Interaction of DMRTA2 with HSP90β Inhibits p53 Ubiquitination and Activates the p53 Pathway to Suppress the Malignant Progression of Non-Small-Cell Lung Cancer
Previous Article in Journal
Molecular Docking and Drug-Likeness of Salicornia-Derived Phytochemicals Against HER Receptors
Previous Article in Special Issue
Regulatory Roles of miR-155-5p, miR-21-5p, miR-93-5p, and miR-140-5p in Breast Cancer Progression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Construction of ceRNA Regulatory Network Unraveled Prognostic Biomarkers and Repositioned Drug Candidates for the Management of Pancreatic Ductal Adenocarcinoma

1
Department of Bioengineering, Faculty of Engineering and Architecture, Konya Food and Agriculture University, Konya 42700, Turkey
2
Department of Biotechnology, Institute of Postgraduate Education, Konya Food and Agriculture University, Konya 42700, Turkey
3
Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul 34854, Turkey
4
Department of Molecular and Precision Medicine, Penn State College of Medicine, Hershey, PA 17033, USA
*
Authors to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2025, 47(7), 496; https://doi.org/10.3390/cimb47070496
Submission received: 7 May 2025 / Revised: 23 June 2025 / Accepted: 26 June 2025 / Published: 27 June 2025

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancer types due to its late diagnosis, low survival rates, and high frequency of metastasis. Considering the molecular mechanism of PDAC development has not been fully elucidated, this study aimed to shed more light on the molecular regulatory signatures of circular RNAs (circRNAs) in PDAC progression and provide a different perspective to identify potential biomarkers as well as discover candidate repositioned drug molecules for the prevention or treatment of PDAC with network-based integrative analysis. The mRNA, miRNA, and circRNA expression profiles of PDAC were obtained from nine microarray datasets. Differentially expressed genes (DEGs), microRNAs (DEmiRNAs), and circular RNAs (DEcircRNAs) were identified. The competing endogenous RNA (ceRNA; DEG–DEmiRNA–DEcircRNA) regulatory network was constructed, which included 12 DEcircRNAs, 64 DEGs, and 6 miRNAs specific to PDAC. The ADAM12, MET, QKI, SEC23A, and ZEB2 were identified as hub genes and demonstrated significant survival probability for PDAC. In addition to providing novel biomarkers for diagnosis that can be detected non-invasively, the secretion levels of hub genes-associated proteins were found in plasma, serum, and oral epithelium. The drug repositioning analysis revealed vorinostat, meclocycline sulfosalicylate, and trichostatin A, which exhibited significant binding affinities to the hub genes compared to their inhibitors via molecular docking analysis.

1. Introduction

Pancreatic cancer is one of the diseases with the worst prognosis [1]. It ranks as the sixth leading cause of cancer deaths in both sexes combined [2]. Pancreatic cancer is responsible for 5% of all cancer mortality worldwide [2]. In 2022, 511,000 new cases of pancreatic cancer were diagnosed, and 467,000 deaths occurred [2]. Pancreatic ductal adenocarcinomas (PDAC) are among the most prevalent exocrine cell tumors, accounting for approximately 95% of cases of pancreatic cancer [3]. It is important to differentiate between advanced chronic pancreatitis, primary pancreatic neoplasms, and ductal adenocarcinoma [4]. In contrast to histological criteria of chronic pancreatitis, which is characterized by regular, lobular formations, intrapancreatic localization, intact ducts, and preserved nuclear polarity, PDAC exhibits characteristics such as irregular distribution, perineural and extrapancreatic dissemination, nuclear pleomorphism, and commonly lost nuclear polarity [4]. The various subtypes of PDAC are classified as ductal adenocarcinoma, adenosquamous carcinoma, colloid carcinoma, anaplastic carcinoma, sarcomatoid carcinoma, and osteoclastic giant cell carcinoma, according to the World Health Organization [5]. The reasons for the poor prognosis of PDAC include delayed diagnosis, lack of effective drugs, and/or resistance of neoplastic cells to conventional chemotherapy [6,7].
Circular RNA (circRNA) is one of the most significant and highly stable single-stranded noncoding RNAs (ncRNAs) because of its covalently closed loop structure [8]. circRNAs are endogenous biomolecules that lack both 5′ end caps or 3′ poly(A) tails [9]. The stability of circRNAs makes them crucial for the development and diagnosis of human diseases and malignancies [10]. circRNAs serve as molecular sponges that sequester microRNAs (miRNAs) from interacting with their target messenger RNAs (mRNAs) [11]. They can control the availability and activity of miRNAs through the sponging mechanism, which impacts many cellular pathways and activities, implicating the development of cancer [12]. For example, circRNA transcriptional adaptor 2A (circTADA2A) has been shown to modulate the microRNA-203a-3p/suppressor of cytokine signaling 3 (SOCS3) axis in breast cancer, thereby suppressing cell proliferation, migration, and invasion [13]. In another example, circ-0001823 suppresses miR-613, which influences RAB8A expression and encourages proliferation and metastasis in cervical cancer [14]. circRNAs control the epigenetic gene expressions through miRNAs, which establish a circRNA–miRNA–mRNA network, known as competing endogenous RNA (ceRNA) [15].
Despite effortful studies having been conducted to understand the mechanism behind the pathogenesis of PDAC, as well as the suggestion of several small molecules for treatment of the disease, there is an unmet need for the development of omics-oriented drugs. Although several signs of progress have been made in the treatment of pancreatic cancer [16,17,18], clinical trial data are still largely missing and are urgently needed to verify relevant effects and for the development of more personalized treatment approaches. This study compared the microarray datasets of PDAC patients as well as controls for the differentially expressed circRNAs, miRNAs, and mRNAs (DEcircRNAs, DEmiRNAs, and DEGs). Furthermore, a detailed investigation of the “DEcircRNA–DEmiRNA–DEGs”-competing endogenous RNA (ceRNA) network was performed for PDAC to reveal novel disease signatures (Figure 1). Several potential repositioned drugs for PDAC were identified via hub gene–drug interaction, and molecular docking analysis was used to validate these in silico. Altogether, this investigation improves the current understanding of the ceRNA regulatory network and the mechanisms involved in the regulation of PDAC carcinogenesis and provides novel candidate biomarkers for prognosis and therapeutic agents.

2. Materials and Methods

2.1. Selection of Microarray Datasets

Data from PDAC and healthy tissues were obtained from the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (NCBI-GEO) database [19]. These datasets included four mRNA expression profiles (GSE15471, GSE41368, GSE71989, and GSE62165), three miRNA expression profiles (GSE60978, GSE32678, and GSE41369), and two circRNA expression profiles (GSE69362 and GSE79634) (Table 1).

2.2. Identification of DEGs, DEmiRNAs, and DEcircRNAs

The mRNA, miRNA, and circRNA datasets were analyzed using the online statistical tool GEO2R (https://www.ncbi.nlm.nih.gov/geo/geo2r/, accessed on 10 February 2025) [19], which combines R/Bioconductor 4.2.2, Biobase 2.58.0, GEOquery 2.66.0, and Linear Models for Microarray Analysis (limma) 3.54.0. The expression values were normalized using the quantile normalization method, which is embedded in the limma package. The Benjamini–Hochberg method was used to control the false discovery rate. To acquire the differentially expressed genes (DEGs), the differentially expressed miRNAs (DEmiRNAs), and the differentially expressed circRNAs (DEcircRNAs) from the dataset, the cut-off criterion for |log2(fold change)| > 1 and p-value < 0.05 were determined for all datasets. The directions of expressions were designated as follows: log2FC ≤ −1 as downregulated and log2FC ≥ 1 as upregulated. To identify common DEGs, DEmiRNAs, and DEcircRNAs, Venn diagrams were constructed for expression profiles separately through Jvenn (https://jvenn.toulouse.inrae.fr/app/example.html, accessed on 17 February 2025) [31].

2.3. Functional Enrichment Analysis

The functional enrichment analysis for common DEGs was performed using GeneCodis v4 [32]. The Kyoto Encyclopedia of Genes and Genomes (KEGG) [33] database and the Gene Ontology (GO) [34] terminology were used for pathway analysis and determination of biological processes, cellular processes, and molecular functions. Results with p < 0.05 were considered statistically significant.

2.4. Construction of ceRNA Network

The relationships between common DEcircRNAs and DEmiRNAs were found in data downloaded from the circBank [35], an extensive human circRNA database. Afterwards, data was downloaded from miRTarBase [36] to construct interactions between common DEmiRNAs and DEGs. The ceRNA (DEGs–DEmiRNAs–DEcircRNAs) regulatory network was constructed by combining DEcircRNA–DEmiRNA pairings with DEmiRNA–DEG pairs. Then, Cytoscape (3.9.1) [37] was used to depict the ceRNA regulatory network. The topological analysis of the network was conducted using both local and global metrics including degree and betweenness centrality features. Using Metascape (v3.5.20240901), GO and KEGG pathway analyses were performed on target DEGs in the ceRNA regulatory network.

2.5. Survival Analysis

Cox regression was performed to elucidate survival-associated genes. The results were visualized and compared through Kaplan–Meier (KM) plots using the follow-up times of high-risk or low-risk groups. The prognostic power of the module was determined using the log-rank test p-value, the hazard ratio (HR), and its confidence intervals (CIs). Survival analysis was performed with the DEGs in ceRNA network construction through the KM-Plotter tool (https://kmplot.com/analysis/index.php?p=service, accessed on 5 March 2025) [38]. The log-rank p-value < 0.05 was accepted as statistically significant.

2.6. Determination of Secretion Levels of Hub Genes-Associated Proteins in Different Tissues

The secretion levels (ppm) of common hub genes-associated proteins were obtained through protein expression data available in the GeneCards database [39]. GeneCards is periodically curated and integrates the protein expression data from external proteomics databases such as ProteomicsDB [40], MaxQB [41], and MOPED [42]. The different tissues/biological fluids such as the pancreas, pancreatic juice, plasma, serum, urine, oral epithelium, and saliva were used, and these were investigated through protein expression data accessible in the above databases.

2.7. Drug Repositioning

Potential drugs and small compounds based on the expression signatures of target DEGs were repositioned through the Library of Integrated Network-based Cellular Signatures (LINCS)–L1000CDS2 data [43]. The resultant repositioned drugs were selected based on their cosine distance scores (1-cos α), the Food and Drug Administration (FDA) approval status, and limitations. The cosine distance search allowed us to perform a similarity search among input genes when these were exposed to candidate repositioned drugs. The largest cosine distance scores represented reverse interactions between input genes and candidate drugs. The mechanism of action and indications of these drugs were cross-checked through published literature and publicly available datasets such as PubChem [44], Comparative Toxicogenomics Database (CTD) [45], and Drug Bank [46].

2.8. Molecular Docking

The three-dimensional (3D) structures of the target DEGs of DEcircRNAs were obtained from the Protein Data Bank (PDB) [47] and UniProt [48]. Inhibitors of each prognostic biomarker were identified from the CTD [45], and the 3D structures of potential drugs and inhibitors were acquired from PubChem [44]. Molecular docking analyses were performed using the Cavity detection-guided Blind Docking (CB-Dock v2) [49] online tool.

3. Results

3.1. Identification of DEGs, DEcircRNAs, DEmiRNAs, and Enrichments of DEGs

A total of 817 DEGs were identified from four sets of mRNA expression profiles (GSE15471, GSE41368, GSE71989, and GSE62165). A total of 37 DEcircRNAs were found in circRNA expression profiles (GSE69362 and GSE79634), and 7 DEmiRNAs were obtained in miRNA expression profiles (GSE60978, GSE32678, and GSE41369) according to the thresholds as indicated in Venn diagrams (Figure 2).
The enrichment analysis revealed distinctive biological characteristics and molecular functions of the common DEGs. According to pathway analysis, the DEGs were enriched in the phagosome, complement, and coagulation cascades, Staphylococcus aureus infection, focal adhesion, leishmaniasis, extracellular matrix (ECM)–receptor interaction, amoebiasis, cytoskeleton in muscle cells, toxoplasmosis, and viral myocarditis. The GO analysis highlighted significant enrichment in cell adhesion, ECM, virus response, angiogenesis, cell migration regulation, endodermal cell differentiation, defense response to virus, collagen fibril organization, and immune response (Figure 3A,B). The functional annotations of the DEmiRNAs were significantly enriched with positive regulation of osteoblast differentiation (Figure 3C). DEmiRNAs were enriched in cancer-related pathways such as microRNAs and proteoglycans in cancer and the chemical carcinogenesis receptor activation pathway (Figure 3D).

3.2. Visualization of ceRNA Network

The PDAC-specific differential expression network was constructed using the interactions of common 64 DEGs, 6 DEmiRNAs, and 12 DEcircRNAs. This network revealed 80 nodes and 138 links between differentially expressed elements (Figure 4A). The topological analysis of the ceRNA network was carried out based on the local and global metrics including degree and betweenness centrality. The significant hub elements (MET, SEC23A, STC2, ZEB1, ZEB2, hsa-miR-130b-5p, hsa-miR-135b-3p, hsa-miR-135b-5p, hsa-miR-199a-3p, hsa-miR-200c-3p, hsa-miR-200c-5p) are depicted in Figure 4B. This network represents the topologically significant hub elements and differs from the previous network by interactions with 79 links around 80 nodes. It was named the ceRNA expression network of the PDAC. To narrow down the interaction list and elucidate the most specific subnetwork composed of nodes that are more closely related than the rest of the components, we constructed a PDAC-specific core network (Figure 4C). This network represents the most likely small circuits preferred by cells in PDAC pathogenesis regarding information flow. The PDAC-specific core network was composed of 15 nodes and 14 edges, and ADAM12, MET, QKI, THBS2, SEC23A, ZEB1, and ZEB2 were identified as hub elements of this network (Figure 4C). Also, there were circRNAs, including hsa_circRNA_102465 and hsa_circRNA_100904, that act as signal distributors of the PDAC-specific core network. The miRNAs such as hsa-miR-135b-5p, hsa-miR-200c-5p, hsa-miR-199a-3p, hsa-miR-130b-5p, hsa-miR-135b-3p, and hsa-miR-200c-3p were also identified in the PDAC-specific core network.

3.3. Survival Analysis and Subnetwork Construction of ceRNA

The KM-Plotter tool uses gene expression data, relapse-free, as well as overall survival information from the most commonly preferred sources of high-throughput datasets such as Gene Expression Omnibus (GEO), European Genome–Phenome Archive (EGA), and The Cancer Genome Atlas (TCGA). To analyze the prognostic power of an individual gene, the patient samples are divided into two groups according to varied quantile expressions of the proposed biomarker. The survival probabilities were compared using the KM plots of the hub gene expressions (Figure 5) between the two different groups to determine prognostic biomarkers. Among these, 5 hub genes ADAM12 (HR = 1.28, p = 9.29 × 10−4), MET (HR = 1.77, p = 4.78 × 10−4), QKI (HR = 1.16, p = 1.33 × 10−4), SEC23A (HR = 1.23, p = 0.00548), and ZEB2 (HR = 1.24, p = 0.00561) were identified and these had higher expression and significant correlation with overall survival time. The prognostic biomarkers that demonstrated an effect on PDAC patient survival are listed in Table 2.

3.4. Secretion Levels of Hub Genes-Associated Proteins in Different Tissues

QKI protein is expressed highly in both plasma and pancreas, and their expression levels are comparable. MET is expressed in the pancreas; however, the expression level is significantly higher in serum. In contrast, SEC23A protein has elevated levels in the pancreas and it is also expressed in oral epithelium.

3.5. Drug Repositioning with ceRNA Subnetwork

Drug repositioning was carried out through L1000CDS2 depending on 5 hub genes and their fold change values. While circRNAs were crucial in the construction of the ceRNA network and the elucidation of upstream regulatory pathways, our subsequent analyses focused on mRNAs for drug repositioning purposes. This decision was methodologically driven, as current drug repositioning platforms—such as L1000CDS2 and CMAP—operate primarily at the mRNA level and are not yet optimized for circRNA-based screening. Therefore, we prioritized mRNAs that are central nodes within the ceRNA network and involved in relevant signaling pathways as therapeutic targets. Nevertheless, circRNAs played a foundational role in identifying these mRNAs within the regulatory network, and their expression profiles were considered in the mechanistic inference.
After using 5 hub genes as a query, the 50 drugs that could reverse the PDAC scenario were suggested via the search engine, and they were eliminated depending on their approval status, mechanism of action, indications, and antineoplastic activities. Among these 50 drugs, 4 drugs (vorinostat, trichostatin A, meclocycline sulfosalicylate, and guanabenz acetate) were selected, and 1 of them was approved, whereas 3 drugs were in the investigational status according to the FDA. Table 3 summarizes the detailed properties of the four repurposed drugs.

3.6. Molecular Docking Analysis with Candidate Repositioned Drugs

The interactions of selected drugs (vorinostat, trichostatin A, meclocycline sulfosalicylate, and guanabenz acetate) with target hub genes of PDAC were analyzed through the molecular docking method (Figure 6). The inhibitors of the target hub genes are abrine (ADAM12), acetaminophen (MET), aristolochic acid I (QKI), 1,2-Dimethylhydrazine (SEC23A), and antimycin A (ZEB2), which were considered as the control group, and their binding affinities (kcal/mol) were compared against the selected drugs. Accordingly, trichostatin A and meclocycline sulfosalicylate had the highest binding affinities with 80% (n = 4 out of 5 hubs) compared to vorinostat (60%, n = 3) and guanabenz acetate (40%, n = 2). The binding affinities of each target hub gene with repurposed drugs were ADAM12 at 75%, MET at 100%, QKI at 0%, SEC23A at 100%, and ZEB2 at 50% compared to the control group. MET and SEC23A were the most significant hubs among the target hub genes regarding molecular docking results.

3.7. Cross-Validation of Protein and Gene Expressions of Proposed Diseased Signatures

We systematically compared expression profiles between diseased and healthy tissues, leveraging publicly available high-resolution transcriptomic and proteomic datasets. Specifically, we assessed the spatial and cell-type-specific expression of the proposed marker genes across multiple tissue types (including normal and PDAC tissues), ensuring a comprehensive validation framework. This analysis provides crucial mechanistic insights by establishing the differential expression patterns that underpin disease-associated molecular signatures. In the immunohistochemical (IHC) stainings, the proposed biomarkers demonstrated distinct protein expression values in PDAC tissues (Figure 7A). The proposed disease signatures exhibit significantly altered gene expression in diseased tissues compared to their healthy counterparts, consistent with their hypothesized role in disease pathogenesis (Figure 7B). The study employed a systems biology-driven approach that integrates transcriptomic data and network-based inference to prioritize candidate targets. Such computational predictions inherently reflect probabilistic associations rather than deterministic outcomes. Discrepancies between mRNA and protein expression are not uncommon, as they can result from various layers of post-transcriptional, translational, or post-translational regulation, as well as tissue-specific expression dynamics and technical limitations of detection methods such as IHC.

4. Discussion

PDAC is the most prevalent pancreas malignant neoplasm [69] and the most lethal among solid malignancies [70]. PDAC is a heterogeneous neoplasm that exhibits varied morphologic and genetic features as well as a poor clinical prognosis [69]. Cytotoxic chemotherapy continues to be the cornerstone of systemic therapy for PDAC, making treatment a significant challenge [71]. Nevertheless, the available first-line therapies might have serious side effects, and there is an urgent need for new treatment solutions. Today, the field of ‘omics’ uses systems-oriented approaches to better understand the predictive and prognostic phenotypes of various diseases, including cancer [72,73,74].
circRNAs have been observed in the initiation and progression stages of various malignancies [75,76]. These circRNAs also have the ability to target miRNAs, which are known as “miRNA sponges”, in order to decrease their levels and remove their target mRNA inhibitions, which in turn controls the gene expressions that code for proteins [77,78]. The primary function of circRNA in different types of cancer is thought to be the sponging of miRNA by ceRNA [79]. The majority of research has been focused on how circRNAs control the growth of tumors through miRNAs [80,81,82].
In this study, we constructed a ceRNA network related to circRNA by thoroughly analyzing the mRNA, miRNA, and circRNA expression patterns of PDAC profiles. The ceRNA network comprised 12 DEcircRNAs (Supplementary Table S1), 64 DEGs (Supplementary Table S2), and 6 DEmiRNAs. Depending on the ceRNA network, circRNAs and mRNAs function as ceRNAs and have the same miRNA binding sites. Our results indicated that hsa_circRNA_102465 and hsa_circRNA_100904 have the most miRNA interactions, which might be crucial for the progression of PDAC.
Regarding our findings, several hub genes, including ADAM12, MET, THBS2, ZEB1, and ZEB2, have been previously implicated in PDAC progression, tumor invasiveness, and epithelial–mesenchymal transition (EMT). ZEB1 and ZEB2 are established transcriptional repressors of E-cadherin and are well-documented drivers of EMT and metastasis in PDAC [83,84]. Yang et al. showed that miR-135-5p expression increased in PDAC patients [85]. A study conducted in 2023 reported an interaction between miR-135-5p and the ADAM12 gene [86]. miR-135-5p expression was detected in placenta and was associated with preeclampsia, and could be used as a potential molecular marker. Furthermore, the elevated miR-135-5p levels interact with the ADAM12 gene and prevent migration and invasion in trophic cells [86]. In another report, miR-135-5p was shown to inhibit cell proliferation by targeting the ADAM12 gene in glioma cells [87,88].
Another candidate, miRNA-200c, has been shown to associate with the metastasis of various tumors [89] and prevent the invasion of tumor tissue in melanoma, breast, and pancreatic cancers [90,91,92]. It has been reported that miRNA-200c closely interacts with the ZEB1 and ZEB2 proteins and prevents invasion in cancer cells through this complex [93,94]. It has also been shown to be effective in directing the EMT mechanism, which is crucial for the metastasis of tumor cells and the progression of cancer [95].
MET proto-oncogene is negatively regulated by miRNA-199a. It is thought that there is potential for preventing the metastasis and progression of cancer cells inhibiting the MET signaling pathway and inducing apotosis in several cancer types [96].
In parallel, several of the identified miRNAs, such as miR-200c-5p, miR-135b-5p, and miR-130b-5p, have been shown to regulate these hub genes in PDAC or related cancer types. miR-200c-5p, for instance, is a well-characterized suppressor of ZEB1/ZEB2, and its downregulation is associated with EMT activation in pancreatic cancer [97]. Similarly, miR-135b-5p has been linked to the regulation of MET and ADAM12, affecting invasion and proliferation in multiple cancers including PDAC [98,99].
Although functional studies on circRNAs such as hsa_circRNA_102465 and hsa_circRNA_100904 remain limited, computational predictions and prior expression data suggest their potential involvement in gastrointestinal cancers and their capability to act as sponges for miRNAs relevant to PDAC regulatory circuits [100].
The protein expression levels in different tissues play a key role in the early diagnosis, monitoring, and treatment of diseases. Elevated or reduced levels of these proteins could be used as a biomarker of some diseases like cancer. They provide information about the presence, progression, or response to treatment. For this reason, we performed an analysis to discover the secretion level of hub genes-associated proteins in different tissues. In this context, the protein expressions in plasma, serum, urine, and saliva could provide an opportunity for diagnosis and response to treatment of PDAC. The significantly high levels of MET, QKI, and SEC23A expressions could be utilized as novel biomarkers for the early diagnosis, monitoring, and effective treatment of PDAC. Also, supportive information in the literature shows that signaling from MET, the receptor for hepatocyte growth factor, promotes pancreatic tumorigenesis and poor patient prognosis by enhancing tumor cell growth, survival, and motility [101]. QKI has been identified as a splicing signature that can distinguish basal-like PDAC subtypes and may predict worse clinical outcomes in pancreatic cancer [102]. SEC23A has been indicated as a potential prognostic marker in bladder cancer [103]. In addition, other probable markers like ADAM12 have been reported as a circulating marker for stromal activation in pancreatic cancer and could predict responses to chemotherapy [104]. Moreover, THBS2 was recently evaluated as a prognostic marker in PDAC metastatic patients [105]. However, it is critical to find these proteins in tissues/biofluids where biological samples can easily be obtained, thus providing a non-invasive method of diagnosis.
Drug repositioning was carried out regarding five hub genes ADAM12, MET, QKI, SEC23A, and ZEB2. This approach provides a significant advantage for identifying novel pancreatic drugs by decreasing drug development costs and time. Among the 50 drugs, 4 drugs (vorinostat, trichostatin A, meclocycline sulfosalicylate, and guanabenz acetate) were selected to reverse PDAC expression depending on the hub genes’ drug repositioning (Table 3). The drug repositioning approach employed in our study was based on transcriptomic reversal signatures using the L1000CDS2 platform. This method does not rely on the traditional mechanism of action alignment but instead uses the ability of small molecules to reverse disease-specific gene expression patterns. Accordingly, the identification of an antimicrobial drug, an antihypertensive agent, and histone deacetylase inhibitors as repositioned drug candidates was not based on their function per se, but rather on their predicted capacity to modulate the expression of oncogenic hub genes implicated in our network analysis. The docking analyses were performed with these drugs to verify results and the binding affinities to target hub genes (ADAM12, MET, QKI, SEC23A, and ZEB2) via in silico analyses.
Vorinostat (Zolinza®, Merck Research Laboratories) belongs to an inhibitor of the histone deacetylase (HDAC) drug class [64]. It is an FDA-approved drug and is used for cutaneous T-cell lymphoma treatment [64]. The HDAC enzyme family catalyzes the removal of acetyl groups from the lysine residues of histone tails, thus resulting in a more compacted chromatin [106]. HDAC inhibitors have a variety of particular impacts on distinct cell types both in vivo and in vitro, including stopping the proliferation of malignant cells, altering their differentiation, regulating migration and angiogenesis, and triggering their apoptosis [107]. Furthermore, BCL6, E2F1, GATA1, P53, SMAD7, and YY1 are transcriptional factors that regulate the genes involved in cell cycle progression, apoptosis, and immune response, and can be directly or indirectly affected by vorinostat [108,109,110,111,112]. Vorinostat was also effective against parasitic diseases such as amoebiasis, cryptosporidiosis, leishmaniasis, malaria, and schistosomiasis [113]. It has been indicated that vorinostat has antiproliferative activity in breast cancer [114], ovarian or primary peritoneal carcinoma [115], lung cancer [116], and gastric cancer [117].
Trichostatin A (TSA) inhibits a hydroxamic acid Class I HDAC inhibitor (HDAC1, 2, 3), Class IIa HDAC inhibitor (HDAC4, 7, 9), and Class IIb inhibitor (HDAC6). It has antineoplastic, antibacterial, and antifungal properties [66]. TSA was first identified as an antifungal antibiotic, isolated from a Streptomyces platensis culture broth [118]. TSA can induce differentiation, cell cycle arrest (G1 and G2 phases), and reverse transform cell morphology to a more normal state in culture [119]. Platta et al. indicated that TSA inhibits the proliferation of small-cell lung cancer cells by inducing morphological differentiation, cell cycle arrest, and apoptosis, which inhibits cell growth in a dose-dependent manner [120]. TSA has also been investigated in PDAC in vitro [121].
Meclocycline sulfosalicylate is a tetracycline, a class of antibiotics used to treat various bacterial infections such as acne vulgaris [122,123]. It binds to the 30S ribosomal subunit and inhibits protein synthesis in bacteria [67]. Thus, adding amino acids to the growing peptide chain is hindered. Also, a wide range of Gram-positive and -negative bacteria are susceptible to the action of meclocycline sulfosalicylate [67].
Guanabenz acetate (GA, WytensinTM) was identified as an antihypertensive drug for the treatment of high blood pressure and functions as an α-2 adrenergic agonist [68]. It also shows anti-prion activity [124]. The mechanism of action of GA works by inhibiting the dephosphorylation of eukaryotic initiation factor-2 alpha (eIF2α) caused by stress and thus causing suppression of endoplasmic reticulum stress [125]. The Rac1-GTPase signaling pathway is downregulated due to increased eIF2α phosphorylation [126]. Overexpression of Rac1-GTPase was observed in triple-negative breast cancer [127].
According to docking results, trichostatin A, meclocycline sulfosalicylate, and vorinostat had significant binding affinities with the hub genes regarding their inhibitors. Moreover, MET and SEC23A were effectively docked with all five repurposed drugs.
MET is a proto-oncogene and the transmembrane tyrosine kinase receptor for scatter factor/hepatocyte growth factor (SF/HGF), which are pleiotropic cytokines [128,129]. When HGF binds to the MET receptor, it triggers cell motility, division, survival, and differentiation [130]. MET has been linked to various biological processes, including tissue repair, morphogenesis, and embryonic and neural development [54].
The SEC23A gene is a main component of COPII, which controls the vesicle budding from the ER [59,131]. Face dysmorphisms, skeletal abnormalities, sutural cataracts, late-closing fontanels, and other symptoms are the main characteristics of cranio-lenticulo-sutural dysplasia (CLSD), which is caused by two missense mutations in SEC23A [60,132]. Zhu et al. indicated the complete absence of SEC23A, which causes it to be fatal during mid-embryogenesis [133]. The molecular mechanism(s) for the role of suggested prognostic markers as well as the impact of drug combinations will be studied in the future, using in vitro experiments utilizing pancreatic ductal adenocarcinoma cells lines.
While the proposed drug candidates demonstrate promising therapeutic potential based on in silico analyses, their actual efficacy in biological systems remains contingent upon a range of pharmacokinetic and pharmacodynamic parameters—including absorption, distribution, metabolism, excretion, and toxicity (ADMET)—which necessitate rigorous evaluation through both in vitro and in vivo experimental models. Additionally, the biological relevance of these findings requires further validation. Specifically, corroborating the differential expression, the construction of the PDAC-specific ceRNA network and functional roles of these hub genes at both the transcriptomic (e.g., quantitative PCR) and proteomic (e.g., Western blotting, immunohistochemistry) levels in clinically relevant samples from PDAC patients would significantly enhance the translational value and scientific robustness of our conclusions.
Collectively, this study offers a novel integrative framework that combines circRNA-associated ceRNA network construction with transcriptomics-driven drug repositioning to uncover previously uncharacterized regulatory mechanisms and therapeutic opportunities in PDAC. Through the construction of a PDAC-specific ceRNA network, we identified key regulatory axes involving hsa_circRNA_102465, hsa_circRNA_100904, miR-135b-5p, miR-200c-5p, and miR-130b-5p, which interact with oncogenic drivers such as ADAM12, MET, QKI, SEC23A, and ZEB2. These interactions offer insight into the post-transcriptional regulation of key oncogenic drivers involved in EMT and tumor progression. Additionally, our use of the L1000CDS2-based transcriptomic reversal approach for drug repositioning represents an innovative strategy in PDAC, distinct from conventional mechanism-of-action-based repurposing. This enabled the identification of non-traditional candidates—some with no prior links to PDAC—such as meclocycline sulfosalicylate and guanabenz acetate, alongside the HDAC inhibitors vorinostat and trichostatin A. Taken together, these findings not only expand the current understanding of circRNA-mediated regulation in PDAC but also provide a promising foundation for non-invasive biomarker development and the preclinical evaluation of repositioned therapeutics. Altogether, these findings provide a strong rationale for further experimental validation and may contribute to the development of novel diagnostic and therapeutic strategies in PDAC.

5. Conclusions

This ceRNA network elucidated that hsa_circRNA_102465 and hsa_circRNA_100904 were crucial players in regulating the gene expression orchestra in PDAC as well as hub microRNAs, including hsa-miR-200c-5p, hsa-miR-199a-3p, hsa-miR-200c-3p, hsa-miR-130b-5p, hsa-miR-135b-3p, and hsa-miR-135b-5p. Under the effect of ceRNA regulation, cascades ADAM12, MET, QKI, SEC23A, and ZEB2 were reported as PDAC disease signatures that significantly impacted the survival rates of pancreatic cancer patients. Based on these signatures, a drug repositioning analysis was conducted, which resulted in the identification of potential drug candidates that aimed to reverse the expression of these signatures to converge the healthy-state gene expressions. As a result, therapeutics such as trichostatin A, vorinostat, and meclocycline sulfosalicylate are being proposed as potential repositioned drug candidates for the therapeutic management of PDAC.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cimb47070496/s1.

Author Contributions

Conceptualization, K.O., B.A., B.T. and R.S.; methodology, K.O. and B.A.; software, K.O. and B.A.; validation, K.O., B.A. and B.T.; formal analysis, K.O. and B.A.; investigation, K.O., B.A., B.T. and O.O.A.; resources, B.T. and O.O.A.; data curation, B.T. and O.O.A.; writing—original draft preparation, K.O. and B.A.; writing—review and editing, B.A. and R.S.; visualization, K.O. and B.A.; supervision, B.A. and R.S.; funding acquisition, R.S.; B.A. and R.S. are responsible for the overall content as guarantors. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Siegel, R.L.; Kratzer, T.B.; Giaquinto, A.N.; Sung, H.; Jemal, A. Cancer statistics, 2025. CA Cancer J Clin. 2025, 75, 10–45. [Google Scholar] [CrossRef] [PubMed]
  2. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global Cancer Statistics 2022: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  3. Grossberg, A.J.; Chu, L.C.; Deig, C.R.; Fishman, E.K.; Hwang, W.L.; Maitra, A.; Marks, D.L.; Mehta, A.; Nabavizadeh, N.; Simeone, D.M.; et al. Multidisciplinary Standards of Care and Recent Progress in Pancreatic Ductal Adenocarcinoma. CA Cancer J. Clin. 2020, 70, 375–403. [Google Scholar] [CrossRef]
  4. Klöppel, G.; Adsay, N.V. Chronic Pancreatitis and the Differential Diagnosis versus Pancreatic Cancer. Arch. Pathol. Lab. Med. 2009, 133, 382–387. [Google Scholar] [CrossRef] [PubMed]
  5. Nagtegaal, I.D.; Odze, R.D.; Klimstra, D.; Paradis, V.; Rugge, M.; Schirmacher, P.; Washington, K.M.; Carneiro, F.; Cree, I.A. The 2019 WHO Classification of Tumours of the Digestive System. Histopathology 2020, 76, 182–188. [Google Scholar] [CrossRef]
  6. de Wilde, R.F.; Hruban, R.H.; Maitra, A.; Offerhaus, G.J.A. Reporting Precursors to Invasive Pancreatic Cancer: Pancreatic Intraepithelial Neoplasia, Intraductal Neoplasms and Mucinous Cystic Neoplasm. Diagn. Histopathol. 2012, 18, 17–30. [Google Scholar] [CrossRef]
  7. Samuel, N.; Hudson, T.J. The Molecular and Cellular Heterogeneity of Pancreatic Ductal Adenocarcinoma. Nat. Rev. Gastroenterol. Hepatol. 2011, 9, 77–87. [Google Scholar] [CrossRef]
  8. Chen, L.-L.; Yang, L. Regulation of CircRNA Biogenesis. RNA Biol. 2015, 12, 381–388. [Google Scholar] [CrossRef]
  9. Adelman, K.; Egan, E. Non-Coding RNA: More Uses for Genomic Junk. Nature 2017, 543, 183–185. [Google Scholar] [CrossRef]
  10. Hashemi, M.; Khosroshahi, E.M.; Daneii, P.; Hassanpoor, A.; Eslami, M.; Koohpar, Z.K.; Asadi, S.; Zabihi, A.; Jamali, B.; Ghorbani, A.; et al. Emerging Roles of CircRNA-MiRNA Networks in Cancer Development and Therapeutic Response. Non-Coding RNA Res. 2025, 10, 98–115. [Google Scholar] [CrossRef]
  11. Kristensen, L.S.; Andersen, M.S.; Stagsted, L.V.W.; Ebbesen, K.K.; Hansen, T.B.; Kjems, J. The Biogenesis, Biology and Characterization of Circular RNAs. Nat. Rev. Genet. 2019, 20, 675–691. [Google Scholar] [CrossRef] [PubMed]
  12. Choi, S.S.; Kim, S.E.; Oh, S.Y.; Ahn, Y.-H. Clinical Implications of Circulating Circular RNAs in Lung Cancer. Biomedicines 2022, 10, 871. [Google Scholar] [CrossRef]
  13. Xu, J.-Z.; Shao, C.-C.; Wang, X.-J.; Zhao, X.; Chen, J.-Q.; Ouyang, Y.-X.; Feng, J.; Zhang, F.; Huang, W.-H.; Ying, Q.; et al. circTADA2As Suppress Breast Cancer Progression and Metastasis via Targeting MiR-203a-3p/SOCS3 Axis. Cell Death Dis. 2019, 10, 175. [Google Scholar] [CrossRef]
  14. Ji, H.; Hu, N. Circular RNA 0001823 Aggravates the Growth and Metastasis of the Cervical Cancer Cells through Modulating the MicroRNA-613/RAB8A Axis. Bioengineered 2022, 13, 10335–10349. [Google Scholar] [CrossRef]
  15. Salmena, L.; Poliseno, L.; Tay, Y.; Kats, L.; Pandolfi, P.P. A CeRNA Hypothesis: The Rosetta Stone of a Hidden RNA Language? Cell 2011, 146, 353–358. [Google Scholar] [CrossRef]
  16. Wooten, D.J.; Sinha, I.; Sinha, R. Selenium Induces Pancreatic Cancer Cell Death Alone and in Combination with Gemcitabine. Biomedicines 2022, 10, 149. [Google Scholar] [CrossRef]
  17. Carbone, D.; Parrino, B.; Cascioferro, S.; Pecoraro, C.; Giovannetti, E.; Di Sarno, V.; Musella, S.; Auriemma, G.; Cirrincione, G.; Diana, P. 1,2,4-Oxadiazole Topsentin Analogs with Antiproliferative Activity against Pancreatic Cancer Cells, Targeting GSK3β Kinase. Chem. Med. Chem. 2021, 16, 537–554. [Google Scholar] [CrossRef]
  18. Carbone, D.; Pecoraro, C.; Panzeca, G.; Xu, G.; Roeten, M.S.F.; Cascioferro, S.; Giovannetti, E.; Diana, P.; Parrino, B. 1,3,4-Oxadiazole and 1,3,4-Thiadiazole Nortopsentin Derivatives against Pancreatic Ductal Adenocarcinoma: Synthesis, Cytotoxic Activity, and Inhibition of CDK1. Mar. Drugs 2023, 21, 412. [Google Scholar] [CrossRef]
  19. Clough, E.; Barrett, T.; Wilhite, S.E.; Ledoux, P.; Evangelista, C.; Kim, I.F.; Tomashevsky, M.; Marshall, K.A.; Phillippy, K.H.; Sherman, P.M.; et al. NCBI GEO: Archive for Gene Expression and Epigenomics Data Sets: 23-Year Update. Nucleic Acids Res. 2024, 52, D138–D144. [Google Scholar] [CrossRef]
  20. Badea, L.; Herlea, V.; Dima, S.O.; Dumitrascu, T.; Popescu, I. Combined Gene Expression Analysis of Whole-Tissue and Microdissected Pancreatic Ductal Adenocarcinoma Identifies Genes Specifically Overexpressed in Tumor Epithelia. Hepatogastroenterology 2008, 55, 2016–2027. [Google Scholar]
  21. Idichi, T.; Seki, N.; Kurahara, H.; Yonemori, K.; Osako, Y.; Arai, T.; Okato, A.; Kita, Y.; Arigami, T.; Mataki, Y.; et al. Regulation of Actin-Binding Protein ANLN by Antitumor MiR-217 Inhibits Cancer Cell Aggressiveness in Pancreatic Ductal Adenocarcinoma. Oncotarget 2017, 8, 53180–53193. [Google Scholar] [CrossRef]
  22. Frampton, A.E.; Castellano, L.; Colombo, T.; Giovannetti, E.; Krell, J.; Jacob, J.; Pellegrino, L.; Roca-Alonso, L.; Funel, N.; Gall, T.M.H.; et al. MicroRNAs Cooperatively Inhibit a Network of Tumor Suppressor Genes to Promote Pancreatic Tumor Growth and Progression. Gastroenterology 2014, 146, 268–277.e18. [Google Scholar] [CrossRef] [PubMed]
  23. Jiang, J.; Azevedo-Pouly, A.C.P.; Redis, R.S.; Lee, E.J.; Gusev, Y.; Allard, D.; Sutaria, D.S.; Badawi, M.; Elgamal, O.A.; Lerner, M.R.; et al. Globally Increased Ultraconserved Noncoding RNA Expression in Pancreatic Adenocarcinoma. Oncotarget 2016, 7, 53165–53177. [Google Scholar] [CrossRef] [PubMed]
  24. Janky, R.; Binda, M.M.; Allemeersch, J.; Van den Broeck, A.; Govaere, O.; Swinnen, J.V.; Roskams, T.; Aerts, S.; Topal, B. Prognostic Relevance of Molecular Subtypes and Master Regulators in Pancreatic Ductal Adenocarcinoma. BMC Cancer 2016, 16, 632. [Google Scholar] [CrossRef] [PubMed]
  25. Sandhu, V.; Bowitz Lothe, I.M.; Labori, K.J.; Lingjærde, O.C.; Buanes, T.; Dalsgaard, A.M.; Skrede, M.L.; Hamfjord, J.; Haaland, T.; Eide, T.J.; et al. Molecular Signatures of MRNAs and MiRNAs as Prognostic Biomarkers in Pancreatobiliary and Intestinal Types of Periampullary Adenocarcinomas. Mol. Oncol. 2015, 9, 758–771. [Google Scholar] [CrossRef]
  26. Donahue, T.R.; Tran, L.M.; Hill, R.; Li, Y.; Kovochich, A.; Calvopina, J.H.; Patel, S.G.; Wu, N.; Hindoyan, A.; Farrell, J.J.; et al. Integrative Survival-Based Molecular Profiling of Human Pancreatic Cancer. Clin. Cancer Res. 2012, 18, 1352–1363. [Google Scholar] [CrossRef]
  27. Toste, P.A.; Li, L.; Kadera, B.E.; Nguyen, A.H.; Tran, L.M.; Wu, N.; Madnick, D.L.; Patel, S.G.; Dawson, D.W.; Donahue, T.R. P85α Is a MicroRNA Target and Affects Chemosensitivity in Pancreatic Cancer. J. Surg. Res. 2015, 196, 285–293. [Google Scholar] [CrossRef]
  28. Li, H.; Hao, X.; Wang, H.; Liu, Z.; He, Y.; Pu, M.; Zhang, H.; Yu, H.; Duan, J.; Qu, S. Circular RNA Expression Profile of Pancreatic Ductal Adenocarcinoma Revealed by Microarray. Cell. Physiol. Biochem. 2016, 40, 1334–1344. [Google Scholar] [CrossRef]
  29. Qu, S.; Song, W.; Yang, X.; Wang, J.; Zhang, R.; Zhang, Z.; Zhang, H.; Li, H. Microarray Expression Profile of Circular RNAs in Human Pancreatic Ductal Adenocarcinoma. Genom. Data 2015, 5, 385–387. [Google Scholar] [CrossRef]
  30. Guo, S.; Xu, X.; Ouyang, Y.; Wang, Y.; Yang, J.; Yin, L.; Ge, J.; Wang, H. Microarray Expression Profile Analysis of Circular RNAs in Pancreatic Cancer. Mol. Med. Rep. 2018, 17, 7661–7671. [Google Scholar] [CrossRef]
  31. Bardou, P.; Mariette, J.; Escudié, F.; Djemiel, C.; Klopp, C. Jvenn: An Interactive Venn Diagram Viewer. BMC Bioinform. 2014, 15, 293. [Google Scholar] [CrossRef] [PubMed]
  32. Garcia-Moreno, A.; López-Domínguez, R.; Villatoro-García, J.A.; Ramirez-Mena, A.; Aparicio-Puerta, E.; Hackenberg, M.; Pascual-Montano, A.; Carmona-Saez, P. Functional Enrichment Analysis of Regulatory Elements. Biomedicines 2022, 10, 590. [Google Scholar] [CrossRef] [PubMed]
  33. 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. [Google Scholar] [CrossRef] [PubMed]
  34. Ashburner, M.; Ball, C.A.; Blake, J.A.; Botstein, D.; Butler, H.; Cherry, J.M.; Davis, A.P.; Dolinski, K.; Dwight, S.S.; Eppig, J.T.; et al. Gene Ontology: Tool for the Unification of Biology. The Gene Ontology Consortium. Nat. Genet. 2000, 25, 25–29. [Google Scholar] [CrossRef]
  35. Liu, M.; Wang, Q.; Shen, J.; Yang, B.B.; Ding, X. Circbank: A Comprehensive Database for CircRNA with Standard Nomenclature. RNA Biol. 2019, 16, 899–905. [Google Scholar] [CrossRef]
  36. Huang, H.-Y.; Lin, Y.-C.-D.; Cui, S.; Huang, Y.; Tang, Y.; Xu, J.; Bao, J.; Li, Y.; Wen, J.; Zuo, H.; et al. MiRTarBase Update 2022: An Informative Resource for Experimentally Validated MiRNA-Target Interactions. Nucleic Acids Res. 2022, 50, D222–D230. [Google Scholar] [CrossRef]
  37. Shannon, P.; Markiel, A.; Ozier, O.; Baliga, N.S.; Wang, J.T.; Ramage, D.; Amin, N.; Schwikowski, B.; Ideker, T. Cytoscape: A Software Environment for Integrated Models of Biomolecular Interaction Networks. Genome Res. 2003, 13, 2498–2504. [Google Scholar] [CrossRef]
  38. Posta, M.; Győrffy, B. Analysis of a Large Cohort of Pancreatic Cancer Transcriptomic Profiles to Reveal the Strongest Prognostic Factors. Clin. Transl. Sci. 2023, 16, 1479–1491. [Google Scholar] [CrossRef]
  39. Stelzer, G.; Rosen, N.; Plaschkes, I.; Zimmerman, S.; Twik, M.; Fishilevich, S.; Stein, T.I.; Nudel, R.; Lieder, I.; Mazor, Y.; et al. The GeneCards Suite: From Gene Data Mining to Disease Genome Sequence Analyses. Curr. Protoc. Bioinform. 2016, 54, 1.30.1–1.30.33. [Google Scholar] [CrossRef]
  40. Samaras, P.; Schmidt, T.; Frejno, M.; Gessulat, S.; Reinecke, M.; Jarzab, A.; Zecha, J.; Mergner, J.; Giansanti, P.; Ehrlich, H.-C.; et al. ProteomicsDB: A Multi-Omics and Multi-Organism Resource for Life Science Research. Nucleic Acids Res. 2020, 48, D1153–D1163. [Google Scholar] [CrossRef]
  41. Schaab, C.; Geiger, T.; Stoehr, G.; Cox, J.; Mann, M. Analysis of High Accuracy, Quantitative Proteomics Data in the MaxQB Database. Mol. Cell. Proteom. 2012, 11, M111.014068. [Google Scholar] [CrossRef] [PubMed]
  42. Montague, E.; Stanberry, L.; Higdon, R.; Janko, I.; Lee, E.; Anderson, N.; Choiniere, J.; Stewart, E.; Yandl, G.; Broomall, W.; et al. MOPED 2.5—An Integrated Multi-Omics Resource: Multi-Omics Profiling Expression Database Now Includes Transcriptomics Data. OMICS J. Integr. Biol. 2014, 18, 335–343. [Google Scholar] [CrossRef]
  43. Duan, Q.; Reid, S.P.; Clark, N.R.; Wang, Z.; Fernandez, N.F.; Rouillard, A.D.; Readhead, B.; Tritsch, S.R.; Hodos, R.; Hafner, M.; et al. L1000CDS2: LINCS L1000 Characteristic Direction Signatures Search Engine. npj Syst. Biol. Appl. 2016, 2, 16015. [Google Scholar] [CrossRef] [PubMed]
  44. Kim, S.; Chen, J.; Cheng, T.; Gindulyte, A.; He, J.; He, S.; Li, Q.; Shoemaker, B.A.; Thiessen, P.A.; Yu, B.; et al. PubChem 2023 Update. Nucleic Acids Res. 2023, 51, D1373–D1380. [Google Scholar] [CrossRef] [PubMed]
  45. Davis, A.P.; Wiegers, T.C.; Johnson, R.J.; Sciaky, D.; Wiegers, J.; Mattingly, C.J. Comparative Toxicogenomics Database (CTD): Update 2023. Nucleic Acids Res. 2023, 51, D1257–D1262. [Google Scholar] [CrossRef]
  46. Knox, C.; Wilson, M.; Klinger, C.M.; Franklin, M.; Oler, E.; Wilson, A.; Pon, A.; Cox, J.; Chin, N.E.L.; Strawbridge, S.A.; et al. DrugBank 6.0: The DrugBank Knowledgebase for 2024. Nucleic Acids Res. 2024, 52, D1265–D1275. [Google Scholar] [CrossRef]
  47. Berman, H.M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T.N.; Weissig, H.; Shindyalov, I.N.; Bourne, P.E. The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235–242. [Google Scholar] [CrossRef]
  48. The UniProt Consortium. UniProt: The Universal Protein Knowledgebase in 2023. Nucleic Acids Res. 2023, 51, D523–D531. [Google Scholar] [CrossRef]
  49. Liu, Y.; Grimm, M.; Dai, W.-T.; Hou, M.-C.; Xiao, Z.-X.; Cao, Y. CB-Dock: A Web Server for Cavity Detection-Guided Protein-Ligand Blind Docking. Acta Pharmacol. Sin. 2020, 41, 138–144. [Google Scholar] [CrossRef]
  50. Wewer, U.M. Barrett, A.J., Rawlings, N.D., Woessner, J.F., Eds.; 212—ADAM12. In Handbook of Proteolytic Enzymes; Academic Press: London, UK, 2004; pp. 724–726. [Google Scholar] [CrossRef]
  51. Mendaza, S.; Ulazia-Garmendia, A.; Monreal-Santesteban, I.; Córdoba, A.; de Azúa, Y.R.; Aguiar, B.; Beloqui, R.; Armendáriz, P.; Arriola, M.; Martín-Sánchez, E.; et al. ADAM12 Is A Potential Therapeutic Target Regulated by Hypomethylation in Triple-Negative Breast Cancer. Int. J. Mol. Sci. 2020, 21, 903. [Google Scholar] [CrossRef]
  52. Christiansen, M.; Spencer, K.; Laigaard, J.; Cowans, N.J.; Larsen, S.O.; Wewer, U.M. ADAM 12 as a Second-Trimester Maternal Serum Marker in Screening for Down Syndrome. Prenat. Diagn. 2007, 27, 611–615. [Google Scholar] [CrossRef] [PubMed]
  53. Nyren-Erickson, E.K.; Jones, J.M.; Srivastava, D.K.; Mallik, S. A Disintegrin and Metalloproteinase-12 (ADAM12): Function, Roles in Disease Progression, and Clinical Implications. Biochim. Biophys. Acta 2013, 1830, 4445–4455. [Google Scholar] [CrossRef] [PubMed]
  54. Malik, R.; Mambetsariev, I.; Fricke, J.; Chawla, N.; Nam, A.; Pharaon, R.; Salgia, R. Kumar, R., Fisher, P.B., Eds.; Chapter Seven—MET Receptor in Oncology: From Biomarker to Therapeutic Target. In Advances in Cancer Research; Academic Press: London, UK, 2020; Volume 147, pp. 259–301. [Google Scholar] [CrossRef]
  55. Furlan, A.; Kherrouche, Z.; Montagne, R.; Copin, M.-C.; Tulasne, D. Thirty Years of Research on Met Receptor to Move a Biomarker from Bench to Bedside. Cancer Res. 2014, 74, 6737–6744. [Google Scholar] [CrossRef]
  56. Benvenuti, S.; Comoglio, P.M. The MET Receptor Tyrosine Kinase in Invasion and Metastasis. J. Cell. Physiol. 2007, 213, 316–325. [Google Scholar] [CrossRef]
  57. Neumann, D.P.; Goodall, G.J.; Gregory, P.A. The Quaking RNA-Binding Proteins as Regulators of Cell Differentiation. WIREs RNA 2022, 13, e1724. [Google Scholar] [CrossRef]
  58. Darbelli, L.; Richard, S. Emerging Functions of the Quaking RNA-Binding Proteins and Link to Human Diseases. WIREs RNA 2016, 7, 399–412. [Google Scholar] [CrossRef]
  59. Lang, M.R.; Lapierre, L.A.; Frotscher, M.; Goldenring, J.R.; Knapik, E.W. Secretory COPII Coat Component Sec23a Is Essential for Craniofacial Chondrocyte Maturation. Nat. Genet. 2006, 38, 1198–1203. [Google Scholar] [CrossRef]
  60. Boyadjiev, S.A.; Fromme, J.C.; Ben, J.; Chong, S.S.; Nauta, C.; Hur, D.J.; Zhang, G.; Hamamoto, S.; Schekman, R.; Ravazzola, M.; et al. Cranio-Lenticulo-Sutural Dysplasia Is Caused by a SEC23A Mutation Leading to Abnormal Endoplasmic-Reticulum-to-Golgi Trafficking. Nat. Genet. 2006, 38, 1192–1197. [Google Scholar] [CrossRef]
  61. Grabitz, A.L.; Duncan, M.K. Focus on Molecules: Smad Interacting Protein 1 (Sip1, ZEB2, ZFHX1B). Exp. Eye Res. 2012, 101, 105–106. [Google Scholar] [CrossRef]
  62. Hegarty, S.V.; Sullivan, A.M.; O’Keeffe, G.W. Zeb2: A Multifunctional Regulator of Nervous System Development. Prog. Neurobiol. 2015, 132, 81–95. [Google Scholar] [CrossRef]
  63. Cui, J.; Pan, G.; He, Q.; Yin, L.; Guo, R.; Bi, H. MicroRNA-545 Targets ZEB2 to Inhibit the Development of Non-small Cell Lung Cancer by Inactivating Wnt/β-Catenin Pathway. Oncol. Lett. 2019, 18, 2931–2938. [Google Scholar] [CrossRef] [PubMed]
  64. Mann, B.S.; Johnson, J.R.; Cohen, M.H.; Justice, R.; Pazdur, R. FDA Approval Summary: Vorinostat for Treatment of Advanced Primary Cutaneous T-Cell Lymphoma. Oncologist 2007, 12, 1247–1252. [Google Scholar] [CrossRef] [PubMed]
  65. Kozeretska, I.A.; Serga, S.V.; Koliada, A.K.; Vaiserman, A.M. Verlinden, H., Ed.; Chapter Four—Epigenetic Regulation of Longevity in Insects. In Advances in Insect Physiology; Academic Press: London, UK, 2017; Volume 53, pp. 87–114. [Google Scholar] [CrossRef]
  66. Cacabelos, R.; Torrellas, C. Tollefsbol, T.O., Ed.; Chapter 32—Pharmacoepigenomics. In Medical Epigenetics; Academic Press: Boston, MA, USA, 2016; pp. 585–617. [Google Scholar] [CrossRef]
  67. Gelmetti, C. Local Antibiotics in Dermatology. Dermatol. Ther. 2008, 21, 187–195. [Google Scholar] [CrossRef] [PubMed]
  68. Holmes, B.; Brogden, R.N.; Heel, R.C.; Speight, T.M.; Avery, G.S. Guanabenz: A Review of its Pharmacodynamic Properties and Therapeutic Efficacy in Hypertension. Drugs 1983, 26, 212–229. [Google Scholar] [CrossRef]
  69. Rishi, A.; Goggins, M.; Wood, L.D.; Hruban, R.H. Pathological and Molecular Evaluation of Pancreatic Neoplasms. Semin. Oncol. 2015, 42, 28–39. [Google Scholar] [CrossRef]
  70. Ferlay, J.; Partensky, C.; Bray, F. More Deaths from Pancreatic Cancer than Breast Cancer in the EU by 2017. Acta Oncol. 2016, 55, 1158–1160. [Google Scholar] [CrossRef]
  71. Rebelo, R.; Polónia, B.; Santos, L.L.; Vasconcelos, M.H.; Xavier, C.P.R. Drug Repurposing Opportunities in Pancreatic Ductal Adenocarcinoma. Pharmaceuticals 2021, 14, 280. [Google Scholar] [CrossRef]
  72. Aydin, B.; Arga, K.Y.; Karadag, A.S. Omics-Driven Biomarkers of Psoriasis: Recent Insights, Current Challenges, and Future Prospects. Clin. Cosmet. Investig. Dermatol. 2020, 13, 611–625. [Google Scholar] [CrossRef]
  73. Beklen, H.; Gulfidan, G.; Arga, K.Y.; Mardinoglu, A.; Turanli, B. Drug Repositioning for P-Glycoprotein Mediated Co-Expression Networks in Colorectal Cancer. Front. Oncol. 2020, 10, 1273. [Google Scholar] [CrossRef]
  74. Turanli, B.; Yildirim, E.; Gulfidan, G.; Arga, K.Y.; Sinha, R. Current State of “Omics” Biomarkers in Pancreatic Cancer. J. Pers. Med. 2021, 11, 127. [Google Scholar] [CrossRef]
  75. Jiang, F.; Shen, X. Current Prevalence Status of Gastric Cancer and Recent Studies on the Roles of Circular RNAs and Methods Used to Investigate Circular RNAs. Cell. Mol. Biol. Lett. 2019, 24, 53. [Google Scholar] [CrossRef] [PubMed]
  76. Li, Z.; Chen, Z.; Hu, G.; Jiang, Y. Roles of Circular RNA in Breast Cancer: Present and Future. Am. J. Transl. Res. 2019, 11, 3945–3954. [Google Scholar]
  77. Chen, J.; Chen, T.; Zhu, Y.; Li, Y.; Zhang, Y.; Wang, Y.; Li, X.; Xie, X.; Wang, J.; Huang, M.; et al. CircPTN Sponges MiR-145-5p/MiR-330-5p to Promote Proliferation and Stemness in Glioma. J. Exp. Clin. Cancer Res. 2019, 38, 398. [Google Scholar] [CrossRef]
  78. Ren, S.; Liu, J.; Feng, Y.; Li, Z.; He, L.; Li, L.; Cao, X.; Wang, Z.; Zhang, Y. Knockdown of CircDENND4C Inhibits Glycolysis, Migration and Invasion by up-Regulating MiR-200b/c in Breast Cancer under Hypoxia. J. Exp. Clin. Cancer Res. 2019, 38, 388. [Google Scholar] [CrossRef]
  79. Gong, Y.; Jiao, Y.; Qi, X.; Fu, J.; Qian, J.; Zhu, J.; Yang, H.; Tang, L. Construction of a CircRNA-MiRNA-MRNA Network Based on Differentially Co-Expressed Circular RNA in Gastric Cancer Tissue and Plasma by Bioinformatics Analysis. World J. Surg. Oncol. 2022, 20, 34. [Google Scholar] [CrossRef]
  80. Jin, J.; Xu, B.; Hu, Z.; He, X. Circular RNA CircTADA2A Promotes the Proliferation, Invasion, and Migration of Non-Small Cell Lung Cancer Cells via the MiR-450b-3p/HMGN5 Signaling Pathway. Transl. Cancer Res. 2022, 11, 242–251. [Google Scholar] [CrossRef]
  81. Zhang, H.; Jiang, L.; Hou, J.; Zhong, S.; Zhou, S.; Zhu, L.; Li, J.; Wang, D.; Sun, D.; Ji, Z.; et al. Circular RNA Hsa_circ_0052112 Promotes Cell Migration and Invasion by Acting as Sponge for MiR-125a-5p in Breast Cancer. Biomed. Pharmacother. 2018, 107, 1342–1353. [Google Scholar] [CrossRef]
  82. Jing, X.F.; Ren, M.Y.; Fan, Y.S.; Fu, Y.H.; Wang, C.F. Circular RNA_0001073 (Circ_0001073) Suppresses The Progression of Non-Small Cell Lung Cancer via MiR-582-3p/RGMB Axis. Cell J. 2021, 23, 684–691. [Google Scholar] [CrossRef] [PubMed]
  83. Kurahara, H.; Takao, S.; Maemura, K.; Mataki, Y.; Kuwahata, T.; Maeda, K.; Sakoda, M.; Iino, S.; Ueno, S.; Natsugoe, S. Epithelial–Mesenchymal Transition and Mesenchymal–Epithelial Transition via Regulation of ZEB-1 and ZEB-2 Expression in Pancreatic Cancer. J. Surg. Oncol. 2012, 105, 655–661. [Google Scholar] [CrossRef]
  84. Krebs, A.M.; Mitschke, J.; Lasierra Losada, M.; Schmalhofer, O.; Boerries, M.; Busch, H.; Boettcher, M.; Mougiakakos, D.; Reichardt, W.; Bronsert, P.; et al. The EMT-Activator Zeb1 Is a Key Factor for Cell Plasticity and Promotes Metastasis in Pancreatic Cancer. Nat. Cell Biol. 2017, 19, 518–529. [Google Scholar] [CrossRef]
  85. Yang, Y.; Ishak Gabra, M.B.; Hanse, E.A.; Lowman, X.H.; Tran, T.Q.; Li, H.; Milman, N.; Liu, J.; Reid, M.A.; Locasale, J.W.; et al. MiR-135 Suppresses Glycolysis and Promotes Pancreatic Cancer Cell Adaptation to Metabolic Stress by Targeting Phosphofructokinase-1. Nat. Commun. 2019, 10, 809. [Google Scholar] [CrossRef] [PubMed]
  86. Sun, B.; Jiang, T.; Yong, J.; Peng, J.; Dong, S.; Gu, Y.; Ji, X.; Luo, L.; Chang, W.-L. MiR-135b-5p Targets ADAM12 to Suppress Invasion and Accelerate Trophoblast Apoptosis in Preeclampsia. Placenta 2023, 143, 69–79. [Google Scholar] [CrossRef]
  87. Lulli, V.; Buccarelli, M.; Martini, M.; Signore, M.; Biffoni, M.; Giannetti, S.; Morgante, L.; Marziali, G.; Ilari, R.; Pagliuca, A.; et al. MiR-135b Suppresses Tumorigenesis in Glioblastoma Stem-like Cells Impairing Proliferation, Migration and Self-Renewal. Oncotarget 2015, 6, 37241–37256. [Google Scholar] [CrossRef]
  88. Kapplingattu, S.V.; Bhattacharya, S.; Adlakha, Y.K. MiRNAs as Major Players in Brain Health and Disease: Current Knowledge and Future Perspectives. Cell Death Discov. 2025, 11, 7. [Google Scholar] [CrossRef]
  89. Katoch, A.; Jamwal, V.L.; Faheem, M.M.; Kumar, S.; Senapati, S.; Yadav, G.; Gandhi, S.G.; Goswami, A. Overlapping Targets Exist between the Par-4 and MiR-200c Axis Which Regulate EMT and Proliferation of Pancreatic Cancer Cells. Transl. Oncol. 2021, 14, 100879. [Google Scholar] [CrossRef]
  90. Yu, J.; Ohuchida, K.; Mizumoto, K.; Sato, N.; Kayashima, T.; Fujita, H.; Nakata, K.; Tanaka, M. MicroRNA, Hsa-MiR-200c, Is an Independent Prognostic Factor in Pancreatic Cancer and Its Upregulation Inhibits Pancreatic Cancer Invasion but Increases Cell Proliferation. Mol. Cancer 2010, 9, 169. [Google Scholar] [CrossRef]
  91. Ahmad, A.; Aboukameel, A.; Kong, D.; Wang, Z.; Sethi, S.; Chen, W.; Sarkar, F.H.; Raz, A. Phosphoglucose Isomerase/Autocrine Motility Factor Mediates Epithelial-Mesenchymal Transition Regulated by MiR-200 in Breast Cancer Cells. Cancer Res. 2011, 71, 3400–3409. [Google Scholar] [CrossRef]
  92. Hurteau, G.J.; Spivack, S.D.; Brock, G.J. Potential MRNA Degradation Targets of Hsa-MiR-200c, Identified Using Informatics and QRT-PCR. Cell Cycle 2006, 5, 1951–1956. [Google Scholar] [CrossRef]
  93. Korpal, M.; Lee, E.S.; Hu, G.; Kang, Y. The MiR-200 Family Inhibits Epithelial-Mesenchymal Transition and Cancer Cell Migration by Direct Targeting of E-Cadherin Transcriptional Repressors ZEB1 and ZEB2. J. Biol. Chem. 2008, 283, 14910–14914. [Google Scholar] [CrossRef]
  94. Sundararajan, V.; Gengenbacher, N.; Stemmler, M.P.; Kleemann, J.A.; Brabletz, T.; Brabletz, S. The ZEB1/MiR-200c Feedback Loop Regulates Invasion via Actin Interacting Proteins MYLK and TKS5. Oncotarget 2015, 6, 27083–27096. [Google Scholar] [CrossRef]
  95. Title, A.C.; Hong, S.-J.; Pires, N.D.; Hasenöhrl, L.; Godbersen, S.; Stokar-Regenscheit, N.; Bartel, D.P.; Stoffel, M. Genetic Dissection of the MiR-200–Zeb1 Axis Reveals Its Importance in Tumor Differentiation and Invasion. Nat. Commun. 2018, 9, 4671. [Google Scholar] [CrossRef] [PubMed]
  96. Kim, S.; Lee, U.J.; Kim, M.N.; Lee, E.-J.; Kim, J.Y.; Lee, M.Y.; Choung, S.; Kim, Y.J.; Choi, Y.-C. MicroRNA MiR-199a* Regulates the MET Proto-Oncogene and the Downstream Extracellular Signal-Regulated Kinase 2 (ERK2). J. Biol. Chem. 2008, 283, 18158–18166. [Google Scholar] [CrossRef] [PubMed]
  97. Würdinger, T.; Tannous, B.A.; Saydam, O.; Skog, J.; Grau, S.; Soutschek, J.; Weissleder, R.; Breakefield, X.O.; Krichevsky, A.M. MiR-296 Regulates Growth Factor Receptor Overexpression in Angiogenic Endothelial Cells. Cancer Cell. 2008, 14, 382–393. [Google Scholar] [CrossRef] [PubMed]
  98. Li, S.; Ma, J.; Si, Y.; Cheng, S.; Hu, M.; Zhi, X.; Li, B.; Yu, H.; Jiang, W.G. Differential Expression and Functions of Ehm2 Transcript Variants in Lung Adenocarcinoma. Int. J. Oncol. 2019, 54, 1747–1758. [Google Scholar] [CrossRef]
  99. Shao, Y.; Zhang, S.; Pan, Y.; Peng, Z.; Dong, Y. miR-135b: A Key Role in Cancer Biology and Therapeutic Targets. Non-Coding RNA Res. 2025, 12, 67–80. [Google Scholar] [CrossRef]
  100. Guo, S.; Zhang, J.; Tao, J.; Zhu, J.; Zheng, R.; Wang, H. Emerging Roles of Circular RNAs in Cancer: A Narrative Review. J. Pancreatol. 2022, 5, 41–48. [Google Scholar] [CrossRef]
  101. Hill, K.S.; Lorinczi, M.; Elferink, L.A. Altered down Regulation of the Receptor Tyrosine Kinase Met in Pancreatic Adenocarcinoma Cells. J. Exp. Ther. Oncol. 2010, 8, 297–312. [Google Scholar]
  102. Ruta, V.; Naro, C.; Pieraccioli, M.; Leccese, A.; Archibugi, L.; Cesari, E.; Panzeri, V.; Allgöwer, C.; Arcidiacono, P.G.; Falconi, M.; et al. An Alternative Splicing Signature Defines the Basal-like Phenotype and Predicts Worse Clinical Outcome in Pancreatic Cancer. Cell Rep. Med. 2024, 5, 101411. [Google Scholar] [CrossRef]
  103. Zeng, B.; Zhao, Q.; Sun, Z.; Liu, D.; Chen, H.; Li, X.; Wang, J.; Xing, H.R. SEC23A Is an Independent Prognostic Biomarker in Bladder Cancer Correlated With MAPK Signaling. Front. Genet. 2021, 12, 672832. [Google Scholar] [CrossRef]
  104. Veenstra, V.L.; Damhofer, H.; Waasdorp, C.; van Rijssen, L.B.; van de Vijver, M.J.; Dijk, F.; Wilmink, H.W.; Besselink, M.G.; Busch, O.R.; Chang, D.K.; et al. ADAM12 Is a Circulating Marker for Stromal Activation in Pancreatic Cancer and Predicts Response to Chemotherapy. Oncogenesis 2018, 7, 87. [Google Scholar] [CrossRef]
  105. Gimotty, P.A.; Till, J.E.; Udgata, S.; Takenaka, N.; Yee, S.S.; LaRiviere, M.J.; O’Hara, M.H.; Reiss, K.A.; O’Dwyer, P.; Katona, B.W.; et al. THSB2 as a Prognostic Biomarker for Patients Diagnosed with Metastatic Panreatic Ductal Adenocarcinoma. Oncotarget 2021, 12, 2266–2272. [Google Scholar] [CrossRef] [PubMed]
  106. Seto, E.; Yoshida, M. Erasers of Histone Acetylation: The Histone Deacetylase Enzymes. Cold Spring Harb. Perspect. Biol. 2014, 6, a018713. [Google Scholar] [CrossRef] [PubMed]
  107. Bubna, A.K. Vorinostat-An Overview. Indian J. Dermatol. 2015, 60, 419. [Google Scholar] [CrossRef] [PubMed]
  108. Zhang, Y.; Adachi, M.; Kawamura, R.; Imai, K. Bmf Is a Possible Mediator in Histone Deacetylase Inhibitors FK228 and CBHA-Induced Apoptosis. Cell Death Differ. 2006, 13, 129–140. [Google Scholar] [CrossRef]
  109. Zhao, C.; Dong, H.; Xu, Q.; Zhang, Y. Histone Deacetylase (HDAC) Inhibitors in Cancer: A Patent Review (2017–Present). Expert Opin. Ther. Pat. 2020, 30, 263–274. [Google Scholar] [CrossRef]
  110. Bali, P.; Pranpat, M.; Bradner, J.; Balasis, M.; Fiskus, W.; Guo, F.; Rocha, K.; Kumaraswamy, S.; Boyapalle, S.; Atadja, P.; et al. Inhibition of Histone Deacetylase 6 Acetylates and Disrupts the Chaperone Function of Heat Shock Protein 90: A Novel Basis for Antileukemia Activity of Histone Deacetylase Inhibitors. J. Biol. Chem. 2005, 280, 26729–26734. [Google Scholar] [CrossRef]
  111. Richon, V.M.; Sandhoff, T.W.; Rifkind, R.A.; Marks, P.A. Histone Deacetylase Inhibitor Selectively Induces P21WAF1 Expression and Gene-Associated Histone Acetylation. Proc. Natl. Acad. Sci. USA 2000, 97, 10014–10019. [Google Scholar] [CrossRef]
  112. Rikiishi, H. Autophagic and Apoptotic Effects of HDAC Inhibitors on Cancer Cells. J. Biomed. Biotechnol. 2011, 2011, 830260. [Google Scholar] [CrossRef]
  113. Herrera-Martínez, M.; Orozco-Samperio, E.; Montaño, S.; Ariza-Ortega, J.A.; Flores-García, Y.; López-Contreras, L. Vorinostat as Potential Antiparasitic Drug. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 7412–7419. [Google Scholar] [CrossRef]
  114. Abdel-Ghany, S.; Raslan, S.; Tombuloglu, H.; Shamseddin, A.; Cevik, E.; Said, O.A.; Madyan, E.F.; Senel, M.; Bozkurt, A.; Rehman, S.; et al. Vorinostat-Loaded Titanium Oxide Nanoparticles (Anatase) Induce G2/M Cell Cycle Arrest in Breast Cancer Cells via PALB2 Upregulation. 3 Biotech 2020, 10, 407. [Google Scholar] [CrossRef]
  115. Modesitt, S.C.; Sill, M.; Hoffman, J.S.; Bender, D.P. A Phase II Study of Vorinostat in the Treatment of Persistent or Recurrent Epithelial Ovarian or Primary Peritoneal Carcinoma: A Gynecologic Oncology Group Study. Gynecol. Oncol. 2008, 109, 182–186. [Google Scholar] [CrossRef] [PubMed]
  116. You, B.R.; Park, W.H. Suberoylanilide Hydroxamic Acid Induces Thioredoxin1-Mediated Apoptosis in Lung Cancer Cells via up-Regulation of MiR-129-5p. Mol. Carcinog. 2017, 56, 2566–2577. [Google Scholar] [CrossRef] [PubMed]
  117. Huang, C.; Ida, H.; Ito, K.; Zhang, H.; Ito, Y. Contribution of Reactivated RUNX3 to Inhibition of Gastric Cancer Cell Growth Following Suberoylanilide Hydroxamic Acid (Vorinostat) Treatment. Biochem. Pharmacol. 2007, 73, 990–1000. [Google Scholar] [CrossRef] [PubMed]
  118. Beljanski, V. Enna, S.J., Bylund, D.B., Eds.; Trichostatin A. In xPharm: The Comprehensive Pharmacology Reference; Enna, S.J., Bylund, D.B., Eds.; Elsevier: New York, NY, USA. In xPharm: The Comprehensive Pharmacology Reference; Elsevier: New York, NY, USA, 2009; pp. 1–4. [Google Scholar] [CrossRef]
  119. Vigushin, D.M.; Ali, S.; Pace, P.E.; Mirsaidi, N.; Ito, K.; Adcock, I.; Coombes, R.C. Trichostatin A Is a Histone Deacetylase Inhibitor with Potent Antitumor Activity against Breast Cancer in Vivo1. Clin. Cancer Res. 2001, 7, 971–976. [Google Scholar]
  120. Platta, C.S.; Greenblatt, D.Y.; Kunnimalaiyaan, M.; Chen, H. The HDAC Inhibitor Trichostatin A Inhibits Growth of Small Cell Lung Cancer Cells. J. Surg. Res. 2007, 142, 219–226. [Google Scholar] [CrossRef]
  121. Donadelli, M.; Costanzo, C.; Faggioli, L.; Scupoli, M.T.; Moore, P.S.; Bassi, C.; Scarpa, A.; Palmieri, M. Trichostatin A, an Inhibitor of Histone Deacetylases, Strongly Suppresses Growth of Pancreatic Adenocarcinoma Cells. Mol. Carcinog. 2003, 38, 59–69. [Google Scholar] [CrossRef]
  122. Knutson, D.D.; Swinyer, L.J.; Smoot, W.H. Meclocycline Sulfosalicylate. Topical Antibiotic Agent for the Treatment of Acne Vulgaris. Cutis 1981, 27, 203–204. [Google Scholar]
  123. Shutter, M.C.; Akhondi, H. Tetracycline. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  124. Tribouillard-Tanvier, D.; Béringue, V.; Desban, N.; Gug, F.; Bach, S.; Voisset, C.; Galons, H.; Laude, H.; Vilette, D.; Blondel, M. Antihypertensive Drug Guanabenz Is Active in Vivo against Both Yeast and Mammalian Prions. PLoS ONE 2008, 3, e1981. [Google Scholar] [CrossRef]
  125. Kang, H.J.; Seol, H.S.; Lee, S.E.; Suh, Y.-A.; Kim, J.; Jang, S.J.; Yu, E. Guanabenz Acetate Induces Endoplasmic Reticulum Stress–Related Cell Death in Hepatocellular Carcinoma Cells. J. Pathol. Transl. Med. 2019, 53, 94–103. [Google Scholar] [CrossRef]
  126. Hamamura, K.; Minami, K.; Tanjung, N.; Wan, Q.; Koizumi, M.; Matsuura, N.; Na, S.; Yokota, H. Attenuation of Malignant Phenotypes of Breast Cancer Cells through EIF2α-Mediated Downregulation of Rac1 Signaling. Int. J. Oncol. 2014, 44, 1980–1988. [Google Scholar] [CrossRef]
  127. Li, X.; Liu, Y.; Zhao, Y.; Tian, W.; Zhai, L.; Pang, H.; Kang, J.; Hou, H.; Chen, Y.; Li, D. Rhein Derivative 4F Inhibits the Malignant Phenotype of Breast Cancer by Downregulating Rac1 Protein. Front. Pharmacol. 2020, 11, 754. [Google Scholar] [CrossRef] [PubMed]
  128. Birchmeier, C.; Birchmeier, W.; Gherardi, E.; Vande Woude, G.F. Met, Metastasis, Motility and More. Nat. Rev. Mol. Cell Biol. 2003, 4, 915–925. [Google Scholar] [CrossRef] [PubMed]
  129. Lamszus, K.; Laterra, J.; Westphal, M.; Rosen, E.M. Scatter Factor/Hepatocyte Growth Factor (SF/HGF) Content and Function in Human Gliomas. Int. J. Dev. Neurosci. 1999, 17, 517–530. [Google Scholar] [CrossRef] [PubMed]
  130. Trusolino, L.; Bertotti, A.; Comoglio, P.M. MET Signalling: Principles and Functions in Development, Organ Regeneration and Cancer. Nat. Rev. Mol. Cell Biol. 2010, 11, 834–848. [Google Scholar] [CrossRef]
  131. Sato, K.; Nakano, A. Mechanisms of COPII Vesicle Formation and Protein Sorting. FEBS Lett. 2007, 581, 2076–2082. [Google Scholar] [CrossRef]
  132. Boyadjiev, S.A.; Kim, S.-D.; Hata, A.; Haldeman-Englert, C.; Zackai, E.H.; Naydenov, C.; Hamamoto, S.; Schekman, R.W.; Kim, J. Cranio-Lenticulo-Sutural Dysplasia Associated with Defects in Collagen Secretion. Clin. Genet. 2011, 80, 169–176. [Google Scholar] [CrossRef]
  133. Zhu, M.; Tao, J.; Vasievich, M.P.; Wei, W.; Zhu, G.; Khoriaty, R.N.; Zhang, B. Neural Tube Opening and Abnormal Extraembryonic Membrane Development in SEC23A Deficient Mice. Sci. Rep. 2015, 5, 15471. [Google Scholar] [CrossRef]
Figure 1. The framework for data collection and analysis in this study.
Figure 1. The framework for data collection and analysis in this study.
Cimb 47 00496 g001
Figure 2. Identification of common (A) DEGs, (B) DEmiRNAs, and (C) DEcircRNAs between different datasets. To eliminate systemic bias, the expression values of all datasets were normalized with the quantile normalization method. The Benjamini–Hochberg method was used to control the false discovery rate. To identify the differentially expressed elements, the cut-off criterion was applied as |log2(fold change)| > 1 and p-value < 0.05 for all datasets.
Figure 2. Identification of common (A) DEGs, (B) DEmiRNAs, and (C) DEcircRNAs between different datasets. To eliminate systemic bias, the expression values of all datasets were normalized with the quantile normalization method. The Benjamini–Hochberg method was used to control the false discovery rate. To identify the differentially expressed elements, the cut-off criterion was applied as |log2(fold change)| > 1 and p-value < 0.05 for all datasets.
Cimb 47 00496 g002
Figure 3. Functional enrichment analyses of DEGs in (A) GO biological processes and (B) KEGG pathways. DEmiRNAs’ functional enrichment analyses in (C) GO biological processes and (D) KEGG pathways.
Figure 3. Functional enrichment analyses of DEGs in (A) GO biological processes and (B) KEGG pathways. DEmiRNAs’ functional enrichment analyses in (C) GO biological processes and (D) KEGG pathways.
Cimb 47 00496 g003
Figure 4. (A) PDAC-specific differential expression network, (B) ceRNA expression network, and (C) PDAC-specific core network.
Figure 4. (A) PDAC-specific differential expression network, (B) ceRNA expression network, and (C) PDAC-specific core network.
Cimb 47 00496 g004
Figure 5. The overall survival curves for hub genes based on gene expression in the ceRNA network. Among the 7 hub genes, 5 of them (ADAM12, MET, QKI, SEC23A, and ZEB2) demonstrated significant survival probabilities. Survival curves were generated using the KM-Plotter tool, incorporating 1640 samples, including 1435 tumor tissues and 205 control tissues for 20,433 genes, as described by Posta and Győrffy [38]. The patient cohorts were provided from publicly available GEO and ICGC datasets, and 94% of the tumors were identified as PDAC. The log-rank test was used to assess overall survival (OS), and hazard ratios (HRs) with 95% confidence intervals were calculated. Log-rank p-values < 0.05 were considered statistically significant.
Figure 5. The overall survival curves for hub genes based on gene expression in the ceRNA network. Among the 7 hub genes, 5 of them (ADAM12, MET, QKI, SEC23A, and ZEB2) demonstrated significant survival probabilities. Survival curves were generated using the KM-Plotter tool, incorporating 1640 samples, including 1435 tumor tissues and 205 control tissues for 20,433 genes, as described by Posta and Győrffy [38]. The patient cohorts were provided from publicly available GEO and ICGC datasets, and 94% of the tumors were identified as PDAC. The log-rank test was used to assess overall survival (OS), and hazard ratios (HRs) with 95% confidence intervals were calculated. Log-rank p-values < 0.05 were considered statistically significant.
Cimb 47 00496 g005
Figure 6. The binding affinities of hub genes with repurposed drug candidates and their specific inhibitors as revealed by molecular docking analysis.
Figure 6. The binding affinities of hub genes with repurposed drug candidates and their specific inhibitors as revealed by molecular docking analysis.
Cimb 47 00496 g006
Figure 7. The protein (A) and gene (B) expression values of proposed PDAC biomarkers, which were collected from the Human Protein Atlas, were demonstrated.
Figure 7. The protein (A) and gene (B) expression values of proposed PDAC biomarkers, which were collected from the Human Protein Atlas, were demonstrated.
Cimb 47 00496 g007
Table 1. The dataset selection for the normal and PDAC pairs-specific transcriptome analysis.
Table 1. The dataset selection for the normal and PDAC pairs-specific transcriptome analysis.
Detected RNA TypesGEO Reference SeriesPlatformSamplesStudy
Design
Ref.
Messenger RNA GSE15471Affymetrix Human Genome U133 Plus 2.0 Array7839 N/
39 PDAC
[20,21]
GSE41368Affymetrix Human Gene 1.0 ST Array126 N/
6 PDAC
[22]
GSE71989Affymetrix Human Genome U133 Plus 2.0 Array228 N/
14 PDAC
[23]
GSE62165Affymetrix Human Genome U219 Array13113 N/
118 PDAC
[24]
MicroRNAGSE60978Agilent-031181 Unrestricted Human miRNA V16.0 Microarray 030840576 N/
51 PDAC
[25]
GSE32678miRCURY LNA microRNA Array, v.11.0—hsa, mmu, and rno327 N/
25 PDAC
[26,27]
GSE41369NanoString nCounter Human miRNA assay V1189 N/
9 PDAC
[22]
Circular RNAGSE69362Agilent-069978 Arraystar Human CircRNA microarray V1126 N/
6 PDAC
[28,29]
GSE79634Agilent-069978 Arraystar Human CircRNA microarray V14020 N/
20 PDAC
[30]
N: normal tissue; PDAC: Pancreatic ductal adenocarcinoma.
Table 2. Significant hub genes identified from the ceRNA core network.
Table 2. Significant hub genes identified from the ceRNA core network.
Hub GeneNameFunctionUniprot IDInhibitorPubChem IDRef.
ADAM12Disintegrin and metalloproteinase domain-containing protein 12Functions in the development of preimplantation embryosO43184Abrine160511[50,51,52,53]
METMET Proto-Oncogene, Receptor Tyrosine KinaseAffects angiogenesis, wound healing, morphogenesis, cell proliferation, cancer invasion, and survivalP08581Acetaminophen1983[54,55,56]
QKIQuaking Homolog, KH Domain Containing RNA BindingControls mRNA stability, translation, export of mRNAs from the nucleus, and pre-mRNA splicingQ96PU8Aristolochic acid I2236[57,58]
SEC23ASEC23 Homolog A, COPII Coat Complex ComponentServing as a GTPase-activating proteinQ154361,2-Dimethylhydrazine1322[59,60]
ZEB2Zinc Finger E-Box-Binding Homeobox 2Activating Wnt/β-catenin signalingO60315Antimycin A14957[61,62,63]
Table 3. Repurposed drug candidates for potential PDAC treatment.
Table 3. Repurposed drug candidates for potential PDAC treatment.
DrugDrug ClassMechanism of ActionApproval StatusTrial Number/Trial StatusRef.
VorinostatHistone deacetylase (HDAC) inhibitorInhibits the enzymatic activity of HDACApprovedPAAD/NCT00948688/Phase I/II[64]
PA/NCT00983268/Phase I/II (Terminated)
PAAD/NCT02349867/Phase I
Trichostatin AAntifungal agentInduces terminal differentiation, cell cycle arrest, and apoptosis in various cancer cell linesInvestigationalHM/NCT03838926/Phase I[65,66]
Meclocycline sulfosalicylateAntimicrobial agentInhibits protein synthesis in bacteria by binding to the 30S ribosomal subunitApproved but discontinuedOM/NCT00385515/Phase II[67]
Guanabenz acetateAntihypertensive agentStimulates central alpha-2 adrenergic receptors, which reduces the release of norepinephrineInvestigationalMS/NCT02423083
/Phase I
[68]
PAAD: Pancreatic Adenocarcinoma, PA: Pancreatic Cancer, HM: Hematologic Malignancies, OM: Oral Mucositis, MS: Multiple Sclerosis.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Aydin, B.; Okutan, K.; Aydogan, O.O.; Sinha, R.; Turanli, B. The Construction of ceRNA Regulatory Network Unraveled Prognostic Biomarkers and Repositioned Drug Candidates for the Management of Pancreatic Ductal Adenocarcinoma. Curr. Issues Mol. Biol. 2025, 47, 496. https://doi.org/10.3390/cimb47070496

AMA Style

Aydin B, Okutan K, Aydogan OO, Sinha R, Turanli B. The Construction of ceRNA Regulatory Network Unraveled Prognostic Biomarkers and Repositioned Drug Candidates for the Management of Pancreatic Ductal Adenocarcinoma. Current Issues in Molecular Biology. 2025; 47(7):496. https://doi.org/10.3390/cimb47070496

Chicago/Turabian Style

Aydin, Busra, Keziban Okutan, Ozge Onluturk Aydogan, Raghu Sinha, and Beste Turanli. 2025. "The Construction of ceRNA Regulatory Network Unraveled Prognostic Biomarkers and Repositioned Drug Candidates for the Management of Pancreatic Ductal Adenocarcinoma" Current Issues in Molecular Biology 47, no. 7: 496. https://doi.org/10.3390/cimb47070496

APA Style

Aydin, B., Okutan, K., Aydogan, O. O., Sinha, R., & Turanli, B. (2025). The Construction of ceRNA Regulatory Network Unraveled Prognostic Biomarkers and Repositioned Drug Candidates for the Management of Pancreatic Ductal Adenocarcinoma. Current Issues in Molecular Biology, 47(7), 496. https://doi.org/10.3390/cimb47070496

Article Metrics

Back to TopTop