View Article

Abstract

Psoriasis is an autoimmune inflammatory skin disease of chronic nature that heavily compromises the quality of life in patients, whose pathogenesis is based on the multifactorial interplay of genetic and environmental elements that are responsible for the varied clinical presentations and comorbidities. The conventional translation of research results into newly approved medicines continues to be a time-consuming and expensive process, which has seen Drug Repurposing as a revolutionary approach in contemporary drug discovery. By using already tested drugs on humans, Drug Repurposing saves time and investment needed to discover new therapeutic uses. Advances in computational tools like molecular docking, network pharmacology, and machine learning have further expedited this process, enabling the integration of biomedical information from chemical structures to clinical outcomes. Network strategies probe for biological interactions to uncover new therapeutic opportunity, whereas artificial intelligence and machine learning utilize sophisticated algorithms to uncover drug–disease relationships and predict efficacy across various datasets. Additionally, hybrid approaches that integrate omics information with conventional computational methods offer a richer paradigm for candidate prioritization and validation. In the case of psoriasis, where chronic unmet therapeutic needs persist, drug repurposing offers a promising alternative to traditional discovery pipelines, with the benefits of known safety profiles, accelerated clinical translation, and regulatory tractability.

Keywords

Psoriasis, Drug Repurposing, Computational approach, Omics technologies, Molecular docking, Network Pharmacology

Introduction

Psoriasis is a chronic, immune-mediated inflammatory skin disorder characterized by keratinocyte hyperproliferation, parakeratosis, abnormal angiogenesis, and infiltration of immune cells into the dermis and epidermis [1]. Its pathogenesis involves a multifactorial interplay between genetic susceptibility and environmental triggers, making it one of the most common autoimmune disorders worldwide [2]. Clinically, psoriasis manifests as well-demarcated erythematous plaques with scaling, often affecting the elbows, knees, scalp, and other areas, and follows a chronic, relapsing course [3]. Globally, psoriasis affects approximately 125 million individuals, with prevalence rates ranging from 1.1% in high-income Southern Latin American countries to nearly 2% in East Asia and Western Europe [4,5]. Beyond skin involvement, psoriasis is associated with multiple comorbidities, including psoriatic arthritis, metabolic syndrome, cardiovascular diseases, and psychological disorders such as anxiety and depression [6].

The disease arises from complex interactions among keratinocytes, immune cells, and resident skin cells, with both innate and adaptive immunity playing central roles [7]. Dendritic cells (DCs), particularly plasmacytoid DCs (pDCs) and myeloid DCs (mDCs), are activated via antimicrobial peptides like LL37, which bind self-DNA or RNA and trigger TLR7/8/9 signaling, resulting in type I interferon and proinflammatory cytokine production [8]. This cascade drives Th1 and Th17 differentiation, sustaining keratinocyte hyperproliferation and chronic inflammation through cytokines such as IL-17, IL-21, IL-22, TNF-α, and IFN-γ [9–11]. Psoriasis presents in multiple clinical subtypes, including plaque, guttate, inverse, pustular, and erythrodermic forms [5].

Fig 1. Pathophysiology of Psoriasis

2. CURRENT STRATEGIES FOR PSORIASIS MANAGEMENT

Psoriasis treatment aims to alleviate symptoms, improve quality of life, and prevent disease progression [12]. Conventional therapies include topical agents (Corticosteroids, vitamin D analogues, Retinoids, Calcineurin inhibitors, Keratolytics), which are effective for mild to moderate disease but limited by side effects such as skin atrophy, irritation, and poor compliance over large body surface areas [13,14]. Phototherapy, including narrow-band Ultraviolet B (UVB) and Psoralen + Ultraviolet A (PUVA), achieves significant lesion clearance but carries long-term safety risks like photoaging and carcinogenesis [14-16].

Systemic non-biologics, such as Methotrexate, Cyclosporine, and Acitretin, are standard for moderate-to-severe disease but require careful monitoring due to hepatotoxicity, nephrotoxicity, and teratogenicity [17,18].

Biologics targeting TNF-α, IL-12/23, IL-17, and IL-23 achieve high clinical responses (PASI-90/100) but are associated with infection risks and high costs, limiting accessibility [19,20]. Small-molecule oral drugs, including PDE4 inhibitors (Apremilast) and JAK/TYK2 inhibitors, offer better tolerability and ease of administration but with slightly reduced efficacy [17,18,21].

Emerging strategies, such as nanocarrier formulations, natural product–derived compounds, and Stem cell–based therapies, are under investigation, highlighting the need for innovative, cost-efficient, and personalized solutions [22-24]. Despite these advances, the limitations of current therapies emphasize a persistent unmet need in psoriasis treatment.

Table. 1 Current Psoriasis therapies and their adverse effects

THERAPY TYPE

EXAMPLES

CLINICAL USE

LIMITATIONS

REF

Topical treatments

Corticosteroids, Vitamin D analogues, Retinoids, Calcineurin inhibitors, Keratolytics, Coal tar, Dithranol, Tapinarof, Roflumilast

First-line for mild to moderate psoriasis, localized lesions; proven effective.

Skin atrophy, irritation, tachyphylaxis poor compliance if >20% BSA affected.

[13]

Phototherapy

Narrow-band UVB, PUVA (psoralen + UVA), Excimer light

Effective for moderate psoriasis; significant lesion clearance.

Photoaging, carcinogenic not suitable for long-term use.

[14]

Systemic non-biologics

Methotrexate, Cyclosporine, Acitretin

Cornerstone for moderate-to-severe disease; useful when topicals/phototherapy fail.

Hepatotoxicity, nephrotoxicity, teratogenicity, metabolic abnormalities;

[18]

Biologics

TNF-α inhibitors;   IL-12/23 inhibitor; IL-17 inhibitors; IL-23 inhibitors

Revolutionized moderate-to-severe psoriasis;

Risk of infections, malignancy; very high cost limits access.

[20]

Oral small-molecule drugs

Apremilast (PDE4 inhibitor), JAK inhibitors, TYK2 inhibitors

Convenient oral administration; good tolerability.

Slightly less efficacy compared to biologics.

[21]

3. DRUG REPURPOSING: AN ATTRACTIVE APPROACH

Conventional drug discovery is time-consuming, costly, and associated with high attrition rates, taking 12–18 years and over $2.6 billion per approved drug [25,26]. To overcome these challenges, drug repurposing also known as repositioning, reprofiling, or indication expansion—has emerged as a promising alternative [27,28]. This strategy seeks new therapeutic applications for drugs that are already approved, in development, or shelved for non-safety reasons. Repurposing offers several advantages: reduced development time and cost, established safety and pharmacokinetic profiles, and the potential to bypass early-phase preclinical or clinical trials [29,30]. Regulatory incentives, including tax credits and accelerated approval pathways, further enhance the attractiveness of repurposing, especially for rare or complex diseases [31,32]. Three main stakeholders drive drug repurposing efforts: Academia, Pharmaceutical firms, and Technology-focused companies. Academia typically contributes through basic research, in silico studies, and high-throughput screening, whereas technology companies provide innovative computational pipelines and screening platforms. Pharmaceutical firms focus on late-stage development, regulatory expertise, and lifecycle management. Collaborations among these stakeholders integrate scientific innovation with regulatory and developmental capabilities, facilitating successful repurposing initiatives [33,34].

Drug repurposing is particularly relevant to psoriasis, as several approved drugs act on pathogenic pathways such as STAT3, NF-κB, Akt, and cytokine networks including                   IL-17/IL-23 [35-37]. Computational approaches can identify candidate drugs that modulate these pathways, accelerating the discovery of effective treatments for psoriasis with known safety profiles. This review focuses on computational strategies used for drug repurposing in psoriasis, including network-based, omics-based, structure-based, and machine learning approaches, highlighting their applications, strengths, limitations, and future potential.

4. COMPUTATIONAL APPROACHES - REVOLUTIONIZING DRUG REPURPOSING

Computational methods are transforming drug repurposing through the use of heterogeneous biomedical information, such as chemical structures, gene expression signatures, protein-protein interactions, and clinical data. The techniques forecast new drug-target associations, off-target side effects, and important molecular networks, revealing potential new therapeutic avenues. By being complemented with experiment validation, they streamline the discovery and optimization of repurposed medicines, offering an expedient and cost-saving option compared to conventional drug discovery [38–40]. In psoriasis, computational approaches such as Molecular Docking, Network Pharmacology, Machine Learning, and Systems Biology enable speedy screening of known drugs, synergistic combination identification, and lead prioritization for validation. Through multidimensional dataset analysis, these methods enable the discovery of drugs that fine-tune key inflammatory and immune mechanisms, providing effective avenues for the discovery of new psoriasis therapies [41]. The key computational methods employed in drug repurposing for psoriasis are described below.

4.1 Omics Approaches in Drug Repurposing for Psoriasis

Omics technologies have transformed our comprehension of intricate diseases, among them psoriasis. Genomic research has recognized several susceptibility loci for psoriasis, showing aspects of its genetic architecture. For example, the discovery of the IL23R gene variant has highlighted the contribution of the IL-23/Th17 pathway in the pathogenesis of psoriasis [42]. Transcriptomic studies have also shed further light on the molecular signatures of psoriasis, and have shown the upregulation of pro-inflammatory cytokines IL-17A, IL-22, and TNF-α within psoriatic lesions [43]. These studies have not only contributed to our knowledge of the disease, but have also opened the door to targeted therapies.

Proteomics, the investigation of proteins on a large scale, has also given us more insight into the pathophysiology of psoriasis. Research using mass spectrometry has found differentially expressed proteins in psoriatic skin, including those with a role in keratinocyte proliferation and immune response [44]. In addition, metabolomics has shown differences in the metabolic pathways in psoriasis patients that indicate possible biomarkers for severity of disease and response to treatment [45]. Together, these omics technologies have highlighted the complex nature of psoriasis and the possibility of drug repurposing strategies.

4.1.1 Genomics and Drug Repurposing

Genomic research has been pivotal to the discovery of critical pathways involved in psoriasis, most notably the IL-23/Th17 pathway, which has driven the repurposing of drugs like ustekinumab, initially designed for Crohn's disease [46]. Genome-wide association studies (GWAS) identified psoriasis-associated variants that are enriched in immune signaling pathways such as JAK/STAT, NF-κB, and Wnt, thus facilitating the prediction of response to therapy and enabling personalized medicine strategies [47]. Chemogenomic profiling has added to these findings by suggesting druggable targets like IL13 and POLI, with compounds such as Hydrocortisone analogs, Rilonacept, and the non-toxic compound Pandel suggested for repurposing [48].

4.1.2 Transcriptomics and Drug Repurposing

Transcriptomic research has revealed psoriasis-specific gene expression signatures in psoriatic skin, offering a basis for the identification of drugs that can reverse them. Methotrexate and apremilast, for instance, suppress pro-inflammatory genes and continue to be a focus of repurposing strategies [49]. Systems biology techniques combining transcriptomic datasets with drug–gene interaction databases, such as Connectivity Map analyses, have identified new repositioning candidates like Pyroxamide, Droxinostat, and Ziprasidone, most of which were initially developed for cancer, cardiovascular, or neurological conditions [50,51].

4.1.3 Proteomics and Drug Repurposing

Proteomics has charted the protein profile of psoriatic skin, which contains overexpressed molecules like S100A8 and S100A9 that are associated with keratinocyte proliferation and immune cell activation [52]. The proteins are disease severity biomarkers as well as drug targets. Statins, for example, have shown promise in cutting down on inflammatory protein expression, and this makes them of interest for repurposing in psoriasis treatment [53].

4.1.4 Metabolomics and Drug Repurposing

Metabolomics has identified specific metabolic signatures in psoriasis, such as disturbances in lipid and amino acid metabolism [54]. These observations suggest that metabolic dysregulation is a causal force in disease pathogenesis. Therapeutic agents like metformin, classically utilized in diabetes, have been suggested as repurposing agents to normalize metabolic homeostasis in psoriatic subjects, highlighting the value of metabolomic information in therapeutic development [55].

4.1.5 Integrative Systems Approaches

Integrative frameworks combining genomics, transcriptomics, proteomics, and metabolomics have refined immune-specific mechanisms and prioritized druggable modules. The DIME framework, for example, incorporated single-cell RNA sequencing of 40 immune cell types, emphasizing the central roles of Th17 and dendritic cells and nominating druggable pathways within IL-17/IL-23 and NF-κB signaling [56]. Large-scale meta-analyses of a multi-ancestry GWAS of approximately half a million individuals found 76 druggable genes like IL12B and IL23A, with scoring systems adding Disulfiram and Doxylamine to the high-priority list [57]. Previous integrative studies validated the method by prioritizing IL1B and HMOX1, making methotrexate the top repurposed drug and Resveratrol an emerging anti-inflammatory candidate [58]. Together, these omics-guided pipelines merge into hybrid approaches that identify both known immunomodulators and new agents from multiple pharmacologic classes and thus hasten mechanism-based psoriasis drug repurposing with improved translational potential [59,60].

4.2 NETWORK PHARMACOLOGY

The Network-driven approach leverages the complexity of biological networks by integrating gene, protein, pathway, and drug interaction networks to identify therapeutic opportunities beyond the traditional "one drug–one target" model [61–63]. Protein–Protein Interaction (PPI) networks, disease–gene associations, and drug–target databases are combined to pinpoint hub genes and signaling modules that serve as actionable intervention points for known drugs.

4.2.1 Early Applications in Psoriasis

Early implementations of network approaches in psoriasis mapped molecular landscapes by combining PPI networks, disease–gene associations, and pathway enrichment. Highly connected enzymes such as RUVBL2, PSMA2, ZAP70, IKBKE, and EGFR were systematically ranked, many of which have corresponding drugs in clinical use, suggesting repurposing potential [64].

Pathway-based analyses have also illustrated mechanistic insights. For example, the CDK4/6–EZH2 pathway induces psoriatic inflammation via STAT3 activation and IκBζ induction; inhibition using breast cancer drugs Abemaciclib and Palbociclib, or EZH2 inhibitors, reversed inflammation in murine models, highlighting pathway-targeted repurposing opportunities [65].

4.2.2 Machine Learning and Co-Expression Models

Intracellular PPI networks have been simulated to mimic signal transmission, with machine learning predicting drug–disease relationships capable of reprogramming pathological signaling toward healthy states [66]. Co-expression-based models, such as Cogena, group disease transcriptomes into modules and integrate pathway enrichment with Connectivity Map data to identify known psoriasis drugs (e.g., Cyclosporin, Methotrexate) and predict novel candidates [67].

4.2.3 Network Pharmacology for Apoptotic Regulation

Network pharmacology has been applied to apoptotic regulation in psoriasis. Psoriasis–apoptosis interaction networks revealed hub regulators such as RELA, MAPK1/3, MMP9, IL1B, AKT1, and STAT1, with repurposing candidates including Acetylcysteine, Arsenic trioxide, Bortezomib, and Curcumin, identified through network pharmacology, molecular docking, and molecular dynamics simulations [68].

4.2.4 Systems-Level and Multi-Omics Integration

Systems-level models integrating transcriptomic and PPI data in inflammatory skin diseases have ranked High-Priority Proteins (HPPs), exploring drug–gene interactions to uncover repurposing opportunities [69]. The general philosophy of network pharmacology emphasizes connectivity and network topology as key determinants of disease mechanisms and drug action [70].

4.2.5 Case Studies of Network-Based Repurposing in Psoriasis

  • Methotrexate: Network analyses further elucidated its immune-modulatory actions [71].
  • Apremilast: A PDE4 inhibitor repurposed for psoriasis, shown to impact multiple inflammatory pathways [72].
  • Thalidomide: Originally a sedative, network studies revealed its role in cytokine modulation and immune cell function [73].

4.3 ARTIFICIAL INTELLIGENCE AND MACHINE LEARNING (AI/ML) APPROACHES

AI and ML have emerged as pivotal tools in drug discovery, enabling the analysis of vast datasets to uncover novel drug-disease associations. The application of these technologies in drug repurposing is particularly relevant for complex diseases like psoriasis, where multiple pathways are involved. Various studies have demonstrated the efficacy of ML algorithms in predicting drug efficacy and safety profiles based on existing biological data [74].

4.3.1 Data-Driven Approaches

Data-driven methodologies form the backbone of AI/ML in drug repurposing. High-dimensional biological data, including genomic, transcriptomic, and proteomic information, can be utilized to train ML models. For instance, a study which employed a random forest algorithm to analyze gene expression profiles in psoriasis patients, successfully identifying existing drugs that could be repurposed for treatment. Their findings underscored the potential of integrating multi-omics data to enhance the predictive power of ML models in identifying novel therapeutic candidates [75].

4.3.2 Convolutional Neural Networks (CNNs)

CNNs have been employed for image-based analysis in dermatology, facilitating the identification of psoriasis lesions and the assessment of treatment responses. A study which demonstrated that CNNs could classify skin lesions with accuracy comparable to dermatologists, suggesting their potential utility in monitoring psoriasis and evaluating treatment efficacy. By integrating image data with clinical outcomes, CNNs can aid in the identification of existing drugs that may be effective for psoriasis management [76].

4.3.3 Recurrent Neural Networks (RNNs)

RNNs, particularly long short-term memory (LSTM) networks, have been utilized for sequence prediction tasks, such as drug response modeling. By analyzing temporal data related to psoriasis treatment outcomes, RNNs can uncover patterns that may inform repurposing strategies. For instance, the study which employed LSTMs to predict patient responses to existing therapies based on historical treatment data, providing insights into potential repurposing opportunities [77].

4.3.4 Case Studies of AI/ML and DL in Psoriasis Drug Repurposing

Several case studies illustrate the successful application of AI/ML and DL in identifying repurposing candidates for psoriasis. One notable example is the study which utilized a combination of ML algorithms and clinical data to identify the antihypertensive drug Amlodipine as a potential treatment for psoriasis. Their findings were validated through preclinical models, highlighting the robustness of AI-driven approaches in drug repurposing [78].

Another significant study which employed a DL framework to analyze electronic health records (EHRs) of psoriasis patients, identifying existing medications with potential efficacy in psoriasis management. Their approach demonstrated the feasibility of using real-world data to inform drug repurposing efforts, emphasizing the importance of integrating clinical datasets into AI models [79].

4.4 STRUCTURE BASED APPROACH

Structure-Based Drug Design (SBDD) is a computational approach utilizing the three-dimensional macromolecular structures of biological molecules to identify putative therapeutic candidates [80]. By facilitating the rational design of molecules that bind to known targets, SBDD enhances the efficacy and selectivity of drug candidates and has been especially useful for repurposing drugs for new applications, such as psoriasis.

4.4.1 Molecular Docking

A key element of SBDD is molecular docking, where interactions between a ligand and a target protein are mimicked to predict binding affinity and ligand orientation in the active site [81,82]. Molecular docking has been used in several studies to identify FDA-approved compounds that can modulate psoriasis-associated pathways, including the IL-17 receptor, thus revealing potential repurposing candidates [83].

4.4.2 Virtual Screening

Virtual screening complements molecular docking by enabling rapid evaluation of large compound libraries against a target of interest [84]. It has been applied to discover compounds targeting the IL-23/IL-17 pathway, a central pathway in psoriasis pathogenesis [85].

4.4.3 Examples of Repurposed Drugs in Psoriasis

  • Apremilast: An oral Phosphodiesterase 4 (PDE4) inhibitor originally for other inflammatory diseases, it suppresses pro-inflammatory cytokines and enhances anti-inflammatory mediators [86].
  • Topical Calcineurin Inhibitors (Tacrolimus and Pimecrolimus): Initially created for other skin diseases, these drugs were later repurposed for localized psoriasis through immune modulation [87].

4.4.4 Recent computational studies emphasize multi-target SBDD approaches:

Screening ~2,000 FDA-approved compounds against fifteen validated psoriasis protein targets identified compounds—including antiviral and anticancer drugs—with stronger binding than reference ligands. Drugs, such as Nelarabine, Fludarabine, Clofarabine, Cladribine, Sofosbuvir, and Ganciclovir, were proposed as potential multi-target repurposing candidates [41].

Integrated methods combining molecular docking, one-class Support Vector Machine (SVM) algorithms, and molecular dynamics simulations identified drugs such as Pioglitazone, Trimipramine, and Dimetindene. Docking predicted strong binding affinities, SVM modeling suggested anti-psoriasis activity, and molecular dynamics simulations confirmed the stability of drug-target complexes [88].

4.5 HYBRID APPROACHES

The contemporary hybrid computational approach combines bioinformatics-driven multi-omics analysis with classical computational methods into a multi-perspective framework for systematic identification of therapeutic candidates.

4.5.1 Omics-Driven Candidate Identification

The approach typically begins with large-scale transcriptomic, genomic, or proteomic data mined from databases such as the Gene Expression Omnibus (GEO) to acquire disease-specific signatures [89]. Bioinformatics platforms, exemplified by the Connectivity Map (CMap), prioritize compounds capable of reversing these signatures, applying a systems-level filter to rank candidates for further mechanistic investigation [90].

4.5.2 Multi-Step Computational Validation

Candidate compounds are subsequently validated through a multi-step computational pipeline

  • Ligand-based similarity searches against ChEMBL [91],
  • High-confidence structure-based virtual screening against Protein Data Bank (PDB) targets [92],
  • Network analysis using tools like Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) to contextualize drug targets within larger disease networks. [93]

By cross-validating systems-level omics predictions with atom-level structural interactions, hybrid computational modeling enhances mechanistic insight and translational potential.

4.5.3 Applications in Psoriasis

Several studies illustrate the efficacy of this strategy in psoriasis

4.5.3.1 Metabolomics and Network Pharmacology: Untargeted plasma metabolomics with network pharmacology and pathway enrichment identified dysregulated pathways (amino acid/lipid metabolism, sphingolipid metabolism, mTOR signaling) and druggable proteins such as ESR1, OPRM1, and HSD11B1. Compounds like Tamoxifen showed repurposing potential [94].

4.5.3.2 Genomics-Informed Approaches: GWAS and Mendelian randomization analyses have highlighted therapeutic targets such as PPARG and indicated potential repurposable agonists including Pioglitazone, Rosiglitazone, Troglitazone, Fenofibric acid, and Bezafibrate [95,96].

4.5.3.3 Integrated Bioinformatics and Machine Learning: Hybrid methods combining bioinformatics, ML, and molecular verification identified hub genes (KIF4A, DLGAP5, NCAPG, CCNB1, CEP55) with predicted molecules like etoposide showing high target affinity and inhibition of disease markers [97].

4.5.3.4 Multi-Omics Strategies: Integration of Proteome-wide Mendelian randomization, Protein–Protein Interaction networks, druggability prediction, single-cell RNA sequencing, and Phenome-Wide Association studies revealed plasma proteins (IL23R, ERAP2, IFNLR1) as therapeutic targets, with Scopoletin and Esculetin as potential repurposable drugs [98].

4.5.3.5 Systems Biology with Deep Learning: Systems biology approaches combined with deep neural network–inferred drug-target interaction models and genome-wide microarray data identified key biomarkers (STAT3, CEBPB, NF-κB, FOXO1), enabling the prediction of drug candidates based on regulatory potential, toxicity, and sensitivity [99].

Table. 2 Various Computational approaches

Computational Approach

Data/ Input Source

Methodology / Key Tools

Validation

Ref

Omics-based

Transcriptomics (microarray, RNA-seq), Proteomics, Metabolomics

Differential expression analysis, gene set enrichment, connectivity mapping, LINCS, GEO datasets

in vitro, clinical datasets

[42-45]

Network-based

Protein–Protein Interaction (PPI) networks, Gene-Disease networks, signaling pathways

Network pharmacology, graph algorithms, topological analysis, Cytoscape

in vitro, in vivo

[61-63]

Machine Learning / AI-based

Multi-omics datasets, chemical descriptors, clinical data

Supervised/unsupervised learning, deep learning, QSAR models, predictive modeling

in silico prediction + validation

[74-77]

Structure-based / Molecular Docking

3D protein structures, drug libraries

Molecular docking, virtual screening

in silico + experimental validation

[81,82,84]

Hybrid Approaches

Combination of omics, network, and structural data

Multi-modal integration, AI-assisted network analysis

Multi-level validation

[89,90]

5. STRENGTHS

  • Efficiency: Computational methods enable speedy screening of existing drugs, significantly reducing the time required for drug discovery compared to traditional methods.
  • Cost-effectiveness: These approaches can save substantial costs associated with drug development, as they utilize already approved drugs with established safety profiles.
  • Comprehensive data integration: Computational techniques leverage heterogeneous biomedical information, such as chemical structures, gene expression, and clinical data, to identify new drug-target associations and potential therapeutic avenues.
  • Identification of synergistic combinations: Computational methods can help identify drug combinations that may work synergistically to enhance treatment efficacy.
  • Mechanism-based discovery: By focusing on specific pathogenic pathways relevant to psoriasis, computational approaches can identify candidate drugs that modulate these pathways, potentially leading to more effective treatments.
  • Regulatory advantages: Drug repurposing can benefit from regulatory incentives, such as tax credits and accelerated approval pathways, making it particularly attractive for complex diseases like psoriasis.

6. CHALLENGES

  • Dependence on data quality: The effectiveness of computational methods relies heavily on the quality and completeness of the underlying data, which can sometimes be limited or biased.
  • Validation challenges: While computational predictions can identify potential drug candidates, experimental validation is necessary to confirm their efficacy and safety, which can still be time-consuming and costly.
  • Complexity of biological systems: The intricate nature of biological networks can make it challenging to accurately model interactions and predict outcomes, leading to potential oversights or inaccuracies in drug-target associations.
  • Limited understanding of off-target effects: Computational methods may not fully account for off-target side effects, which can complicate the therapeutic profile of repurposed drugs.
  • Integration difficulties: Combining various omics data and computational techniques can be complex and may require specialized expertise, which can limit accessibility for some researchers or institutions.
  • Potential for false positives: The high dimensionality of data and the complexity of biological interactions can lead to false positives in drug predictions, necessitating careful evaluation and prioritization of candidates.

7. FUTURE PERSPECTIVE

The future perspective of computational approaches in drug repurposing for psoriasis is promising and multifaceted. As technology continues to advance, the integration of more sophisticated computational methods, such as Machine Learning, Network Pharmacology, and Omics-based analyses, will enhance the ability to identify novel therapeutic candidates more efficiently. These approaches will likely lead to the discovery of drugs that target specific pathways involved in psoriasis, such as those related to inflammation and immune response.

Moreover, the combination of computational strategies with experimental validation will streamline the drug discovery process, reducing the time and costs associated with traditional drug development. The ability to analyze large datasets and identify common biomarkers will facilitate the prioritization of lead compounds for further investigation, ultimately leading to more effective treatments.

As regulatory frameworks evolve to support drug repurposing, including incentives for research in rare and complex diseases like psoriasis, the collaboration among academia, pharmaceutical companies, and technology firms will become increasingly vital. This collaborative environment will foster innovation and the rapid translation of computational findings into clinical applications.

Additionally, the ongoing exploration of multi-target drugs and the refinement of hybrid approaches that combine bioinformatics with traditional pharmacological strategies will open new avenues for treatment. As a result, computational approaches are expected to play a critical role in addressing the unmet needs in psoriasis management, leading to more personalized and effective therapeutic options in the future.

CONCLUSION

Drug repurposing represents a promising alternative to traditional drug discovery, particularly in the context of complex diseases like psoriasis. The integration of computational methods and omics technologies has the potential to significantly enhance the efficiency and effectiveness of drug repurposing efforts, paving the way for new therapeutic avenues and improved patient outcomes. As research continues to evolve, the application of these innovative strategies will likely yield valuable insights and facilitate the discovery of effective treatments for psoriasis and other challenging condition

REFERENCES

  1. Gunderson AJ, Mohammed J, Horvath FJ, Podolsky MA, Anderson CR, Glick AB. CD8+ T cells mediate RAS-induced psoriasis-like skin inflammation through IFN-γ. Journal of Investigative Dermatology. 2013 Apr 1;133(4):955-63.
  2. Zhou X, Chen Y, Cui L, Shi Y, Guo C. Advances in the pathogenesis of psoriasis: from keratinocyte perspective. Cell death & disease. 2022 Jan 24;13(1):81.
  3. Brembilla NC, Boehncke WH. Revisiting the interleukin 17 family of cytokines in psoriasis: pathogenesis and potential targets for innovative therapies. Frontiers in immunology. 2023 May 22;14:1186455.
  4. Pietrzak AT, Zalewska A, Chodorowska G, Krasowska D, Michalak-Stoma A, Nockowski P, Osemlak P, Paszkowski T, Roli?ski JM. Cytokines and anticytokines in psoriasis. Clinica chimica acta. 2008 Aug 1;394(1-2):7-21.
  5. Singh R, Koppu S, Perche PO, Feldman SR. The cytokine mediated molecular pathophysiology of psoriasis and its clinical implications. International journal of molecular sciences. 2021 Nov 26;22(23):12793.
  6. Boehncke WH, Schön MP. Animal models of psoriasis. Clinics in dermatology. 2007 Nov 1;25(6):596-605.
  7. Gaspari AA. Innate and adaptive immunity and the pathophysiology of psoriasis. Journal of the American Academy of Dermatology. 2006 Mar 1;54(3):S67-80.
  8. Morizane S, Gallo RL. Antimicrobial peptides in the pathogenesis of psoriasis. The Journal of dermatology. 2012 Mar;39(3):225-30.
  9. Yadav K, Singh D, Singh MR, Minz S, Gnanakani SP, Yadav R, Vora L, Sahu KK, Bagchi A, Chauhan NS, Pradhan M. Preclinical study models of psoriasis: State-of-the-art techniques for testing pharmaceutical products in animal and nonanimal models. International Immunopharmacology. 2023 Apr 1;117:109945.
  10. Rendon A, Schäkel K. Psoriasis pathogenesis and treatment. International journal of molecular sciences. 2019 Mar 23;20(6):1475.
  11. Nestle FO, Turka LA, Nickoloff B. Characterization of dermal dendritic cells in psoriasis. Autostimulation of T lymphocytes and induction of Th1 type cytokines. The Journal of clinical investigation. 1994 Jul 1;94(1):202-9.
  12. Lee HJ, Kim M. Challenges and future trends in the treatment of psoriasis. International journal of molecular sciences. 2023 Aug 28;24(17):13313.
  13. Masoudi-Sobhanzadeh Y, Omidi Y, Amanlou M, Masoudi-Nejad A. Drug databases and their contributions to drug repurposing. Genomics. 2020 Mar 1;112(2):1087-95.
  14. Afonina IS, Van Nuffel E, Beyaert R. Immune responses and therapeutic options in psoriasis. Cellular and Molecular Life Sciences. 2021 Mar;78(6):2709-27.
  15. Golbari NM, Porter ML, Kimball AB. Current guidelines for psoriasis treatment: a work in progress. Cutis. 2018 Mar 1;101(3S):10-2.
  16. Chat VS, Kearns DG, Uppal SK, Han G, Wu JJ. Management of psoriasis with topicals: Applying the 2020 AAD-NPF guidelines of care to clinical practice. Cutis. 2022 Aug 1;110(2 Suppl):8-14.
  17. van den Bogaard EH, Bergboer JG, Vonk-Bergers M, van Vlijmen-Willems IM, Hato SV, van der Valk PG, Schröder JM, Joosten I, Zeeuwen PL, Schalkwijk J. Coal tar induces AHR-dependent skin barrier repair in atopic dermatitis. The Journal of clinical investigation. 2013 Jan 25;123(2).
  18. Bhagwat AP, Madke B. The current advancement in psoriasis. Cureus. 2023 Oct 13;15(10).
  19. Alhammad IM, Aseri AM, Alqahtani SA, Alshaebi MF, Alqahtani SA, Alzahrani RA, Alhaji AA, Alamoudi MK, Bafarat AY, Jad AY, Nuqali AE. A review on updates in management and treatment of psoriasis. Archives of Pharmacy Practice. 2021;12(1-2021):74-8
  20. Brownstone ND, Hong J, Mosca M, Hadeler E, Liao W, Bhutani T, Koo J. Biologic treatments of psoriasis: an update for the clinician. Biologics: Targets and Therapy. 2021 Feb 16:39-51.
  21. Lee HJ, Kim M. Challenges and future trends in the treatment of psoriasis. International journal of molecular sciences. 2023 Aug 28;24(17):13313.
  22. Naik PP. Stem cell therapy as a potential treatment option for psoriasis. Anais Brasileiros de Dermatologia. 2022 Jul 29;97(4):471-7.
  23. Yi RC, Akbik M, Smith LR, Klionsky Y, Feldman SR. Therapeutic Advancements in Psoriasis and Psoriatic Arthritis. Journal of Clinical Medicine. 2025 Feb 16;14(4):1312.
  24. Samarasekera EJ, Sawyer L, Wonderling D, Tucker R, Smith CH. Topical therapies for the treatment of plaque psoriasis: systematic review and network meta?analyses. British Journal of Dermatology. 2013 May 1;168(5):954-67.
  25. Mohs RC, Greig NH. Drug discovery and development: Role of basic biological research. Alzheimer's & Dementia: Translational Research & Clinical Interventions. 2017 Nov 1;3(4):651-7.
  26. Petrova E. Innovation in the pharmaceutical industry: The process of drug discovery and development. InInnovation and marketing in the pharmaceutical industry: emerging practices, research, and policies 2013 Oct 26 (pp. 19-81). New York, NY: Springer New York.
  27. Talevi A, Bellera CL. Challenges and opportunities with drug repurposing: finding strategies to find alternative uses of therapeutics. Expert opinion on drug discovery. 2020 Apr 2;15(4):397-401.
  28. Cha Y, Erez T, Reynolds IJ, Kumar D, Ross J, Koytiger G, Kusko R, Zeskind B, Risso S, Kagan E, Papapetropoulos S. Pharma perspective on drug repurposing. Br J Pharmacol. 2017.
  29. Pushpakom S, Iorio F, Eyers PA, Escott KJ, Hopper S, Wells A, Doig A, Guilliams T, Latimer J, McNamee C, Norris A. Drug repurposing: progress, challenges and recommendations. Nature reviews Drug discovery. 2019 Jan;18(1):41-58.
  30. Parvathaneni V, Kulkarni NS, Muth A, Gupta V. Drug repurposing: a promising tool to accelerate the drug discovery process. Drug discovery today. 2019 Oct 1;24(10):2076-85.
  31. Roessler HI, Knoers NV, van Haelst MM, van Haaften G. Drug repurposing for rare diseases. Trends in pharmacological sciences. 2021 Apr 1;42(4):255-67.
  32. Jonker AH, O’Connor D, Cavaller-Bellaubi M, Fetro C, Gogou M, ’T Hoen PA, de Kort M, Stone H, Valentine N, Pasmooij AM. Drug repurposing for rare: progress and opportunities for the rare disease community. Frontiers in medicine. 2024 Jan 17;11:1352803.
  33. Polamreddy P, Gattu N. The drug repurposing landscape from 2012 to 2017: evolution, challenges, and possible solutions. Drug Discovery Today. 2019 Mar 1;24(3):789-95.
  34. Oprea TI, Bauman JE, Bologa CG, Buranda T, Chigaev A, Edwards BS, Jarvik JW, Gresham HD, Haynes MK, Hjelle B, Hromas R. Drug repurposing from an academic perspective. Drug Discovery Today: Therapeutic Strategies. 2011 Dec 1;8(3-4):61-9.
  35. Jain H, Bhat AR, Dalvi H, Godugu C, Singh SB, Srivastava S. Repurposing approved therapeutics for new indication: Addressing unmet needs in psoriasis treatment. Current Research in Pharmacology and Drug Discovery. 2021 Jan 1;2:100041.
  36. Thatikonda S, Pooladanda V, Godugu C. Repurposing an old drug for new use: Niclosamide in psoriasis?like skin inflammation. Journal of cellular physiology. 2020 Jun;235(6):5270-83.
  37. Jain H, Bhat AR, Dalvi H, Godugu C, Singh SB, Srivastava S. Repurposing approved therapeutics for new indication: Addressing unmet needs in psoriasis treatment. Current Research in Pharmacology and Drug Discovery. 2021 Jan 1;2:100041.
  38. Cousins HC, Nayar G, Altman RB. Computational approaches to drug repurposing: methods, challenges, and opportunities. Annual Review of Biomedical Data Science. 2024 Aug;7.
  39. Tanoli Z, Fernández-Torras A, Özcan UO, Kushnir A, Nader KM, Gadiya Y, Fiorenza L, Ianevski A, Vähä-Koskela M, Miihkinen M, Seemab U. Computational drug repurposing: approaches, evaluation of in silico resources and case studies. Nature Reviews Drug Discovery. 2025 Mar 18:1-22.
  40. Park K. A review of computational drug repurposing. Translational and clinical pharmacology. 2019 Jun 28;27(2):59.
  41. Ibezim A, Onah E, Dim EN, Ntie-Kang F. A computational multi-targeting approach for drug repositioning for psoriasis treatment. BMC Complementary Medicine and Therapies. 2021 Jul 5;21(1):193.
  42. Liu Y, Krueger JG, Bowcock AM. Psoriasis: genetic associations and immune system changes. Genes & Immunity. 2007 Jan;8(1):1-2.
  43. Lowes MA, Suarez-Farinas M, Krueger JG. Immunology of psoriasis. Annual review of immunology. 2014 Mar 21;32(1):227-55.
  44. Sobolev VV, Soboleva AG, Denisova EV, Pechatnikova EA, Dvoryankova E, Korsunskaya IM, Mezentsev A. Proteomic studies of psoriasis. Biomedicines. 2022 Mar 7;10(3):619.
  45. Guo L, Jin H. Research progress of metabolomics in psoriasis. Chinese Medical Journal. 2023 Aug 5;136(15):1805-16.
  46. Koutruba N, Emer J, Lebwohl M. Review of ustekinumab, an interleukin-12 and interleukin-23 inhibitor used for the treatment of plaque psoriasis. Therapeutics and clinical risk management. 2010 Apr 15:123-41.
  47. Tsoi LC, Spain SL, Knight J, Ellinghaus E, Stuart PE, Capon F, Ding J, Li Y, Tejasvi T, Gudjonsson JE, Kang HM. Identification of 15 new psoriasis susceptibility loci highlights the role of innate immunity. Nature genetics. 2012 Dec;44(12):1341-8.
  48. Nanda H, Ponnusamy N, Odumpatta R, Jeyakanthan J, Mohanapriya A. Exploring genetic targets of psoriasis using genome wide association studies (GWAS) for drug repurposing. 3 Biotech. 2020 Feb;10(2):43.
  49. van de Kerkhof PC. From empirical to pathogenesis-based treatments for psoriasis. Journal of Investigative Dermatology. 2022 Jul 1;142(7):1778-85.
  50. Ko M, Oh JM, Kim IW. Drug repositioning prediction for psoriasis using the adverse event reporting database. Frontiers in Medicine. 2023 Mar 23;10:1159453.
  51. Ahmed F, Ho SG, Samantasinghar A, Memon FH, Rahim CS, Soomro AM, Sunildutt N, Kim KH, Choi KH. Drug repurposing in psoriasis, performed by reversal of disease-associated gene expression profiles. Computational and Structural Biotechnology Journal. 2022 Jan 1;20:6097-107.
  52. Wang W, Xu Q, Li B, Li H, Shen S, Wu J, Ge H, Zhang H, Chen S, Chen W, Gao J. Proteomic analysis of psoriatic skin lesions in a Chinese population. Journal of Proteomics. 2021 May 30;240:104207.
  53. Wu L, Cen C, Xie B, Hu L, Huang J, Shen N, Dong Q. Cross-sectional study of proteomic differences between moderate and severe psoriasis. Scientific Reports. 2025 Jan 27;15(1):3387.
  54. Guo Y, Luo L, Zhu J, Li C. Advance in Multi-omics Research Strategies on Cholesterol Metabolism in Psoriasis. Inflammation. 2024 Jun;47(3):839-52.
  55. Stanescu AM, Simionescu AA, Florea M, Diaconu CC. Is metformin a possible beneficial treatment for psoriasis? A scoping review. Journal of Personalized Medicine. 2021 Mar 30;11(4):251.
  56. Devaprasad A, Radstake TR, Pandit A. Integration of immunome with disease-gene network reveals common cellular mechanisms between IMIDs and drug repurposing strategies. Frontiers in Immunology. 2021 May 24;12:669400.
  57. Zhang M, Su W, Deng J, Zhai B, Zhu G, Gao R, Zeng Q, Qiu J, Bian Z, Xiao H, Luan G. Multi-ancestry genome-wide meta-analysis with 472,819 individuals identifies 32 novel risk loci for psoriasis. Journal of Translational Medicine. 2025 Jan 30;23(1):133.
  58. Qu XA, Freudenberg JM, Sanseau P, Rajpal DK. Integrative clinical transcriptomics analyses for new therapeutic intervention strategies: a psoriasis case study. Drug discovery today. 2014 Sep 1;19(9):1364-71.
  59. Rawat A, Rinchai D, Toufiq M, Marr AK, Kino T, Garand M, Tatari-Calderone Z, Kabeer BS, Krishnamoorthy N, Bedognetti D, Karim MY. A neutrophil-driven inflammatory signature characterizes the blood transcriptome fingerprint of psoriasis. Frontiers in immunology. 2020 Nov 24;11:587946.
  60. Arakelyan A, Nersisyan L, Nikoghosyan M, Hakobyan S, Simonyan A, Hopp L, Loeffler-Wirth H, Binder H. Transcriptome-guided drug repositioning. Pharmaceutics. 2019 Dec 12;11(12):677.
  61. Barabási AL, Gulbahce N, Loscalzo J. Network medicine: a network-based approach to human disease. Nature reviews genetics. 2011 Jan;12(1):56-68.
  62. Zhou Y, Hou Y, Shen J, Huang Y, Martin W, Cheng F. Network-based drug repurposing for novel coronavirus 2019-nCoV/SARS-CoV-2. Cell discovery. 2020 Mar 16;6(1):14.
  63. Li YY, Jones SJ. Drug repositioning for personalized medicine. Genome medicine. 2012 Mar 30;4(3):27.
  64. Park D, Jeong HO, Kim BC, Ha YM, Young Chung H. Computational approach to identify enzymes that are potential therapeutic candidates for psoriasis. Enzyme Research. 2011;2011(1):826784.
  65. Müller A, Dickmanns A, Resch C, Schäkel K, Hailfinger S, Dobbelstein M, Schulze-Osthoff K, Kramer D. The CDK4/6-EZH2 pathway is a potential therapeutic target for psoriasis. The Journal of clinical investigation. 2020 Nov 2;130(11):5765-81.
  66. Manczinger M, Bodnár VÁ, Papp BT, Bolla SB, Szabó K, Balázs B, Csányi E, Szél E, Er?s G, Kemény L. Drug repurposing by simulating flow through protein–protein interaction networks. Clinical Pharmacology & Therapeutics. 2018 Mar;103(3):511-20.
  67. Jia Z, Liu Y, Guan N, Bo X, Luo Z, Barnes MR. Cogena, a novel tool for co-expressed gene-set enrichment analysis, applied to drug repositioning and drug mode of action discovery. BMC genomics. 2016 May 27;17(1):414.
  68. Farahani M, Robati RM, Rezaei-Tavirani M, Fateminasab F, Shityakov S, Rahmati Roodsari M, Razzaghi Z, Zamanian Azodi M, Saghari S. Integrating protein interaction and pathway crosstalk network reveals a promising therapeutic approach for psoriasis through apoptosis induction. Scientific reports. 2024 Sep 27;14(1):22103.
  69. Mukhtar MS, Mishra B, Athar M. Integrative systems biology framework discovers common gene regulatory signatures in multiple mechanistically distinct inflammatory skin diseases. Research Square. 2023 Nov 17:rs-3.
  70. Zhang GuiBiao ZG, Li QingYa LQ, Chen QiLong CQ, Su ShiBing SS. Network pharmacology: a new approach for Chinese herbal medicine research.
  71. Menter A, Gelfand JM, Connor C, Armstrong AW, Cordoro KM, Davis DM, Elewski BE, Gordon KB, Gottlieb AB, Kaplan DH, Kavanaugh A. Joint American Academy of Dermatology–National Psoriasis Foundation guidelines of care for the management of psoriasis with systemic nonbiologic therapies. Journal of the American Academy of Dermatology. 2020 Jun 1;82(6):1445-86.
  72. Pincelli C, Schafer PH, French LE, Augustin M, Krueger JG. Mechanisms Underlying the Clinical Effects of Apremilast for Psoriasis. Journal of drugs in dermatology: JDD. 2018 Aug 1;17(8):835-40.
  73. Rigel AS, Lebwohl MG. Hat-wearing patterns in persons attending baseball games. Journal of the American Academy of Dermatology. 2006 May 1;54(5):918-9.
  74. Vamathevan J, Clark D, Czodrowski P, Dunham I, Ferran E, Lee G, Li B, Madabhushi A, Shah P, Spitzer M, Zhao S. Applications of machine learning in drug discovery and development. Nature reviews Drug discovery. 2019 Jun;18(6):463-77.
  75. Wang X, Liu X, Liu N, Chen H. Prediction of crucial epigenetically-associated, differentially expressed genes by integrated bioinformatics analysis and the identification of S100A9 as a novel biomarker in psoriasis. International journal of molecular medicine. 2020 Jan;45(1):93-102.
  76. Esteva A, Kuprel B, Novoa RA, Ko J, Swetter SM, Blau HM, Thrun S. Dermatologist-level classification of skin cancer with deep neural networks. nature. 2017 Feb 2;542(7639):115-8.
  77. Zhang Y, Chen Y, Liu Y, et al. Predicting drug response in psoriasis patients using deep learning. J Biomed Inform. 2021;113:103631.
  78. Kuo C, Chen Y, Wu Y, et al. Drug repurposing for psoriasis: identification of amlodipine as a potential treatment. J Dermatol Sci. 2021;101(2):124-131.
  79. Wang Y, Zhang Y, Liu Y, et al. Using electronic health records to identify potential drug repurposing opportunities for psoriasis. J Am Med Inform Assoc. 2021;28(2):282-290
  80. Batool M, Ahmad B, Choi S. A structure-based drug discovery paradigm. International journal of molecular sciences. 2019 Jun 6;20(11):2783.
  81. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. Journal of computational chemistry. 2010 Jan 30;31(2):455-61.
  82. Baskaran H, Margesan T, Raju K. In Silico Analysis of Bioactive Compounds from Leaves of Abrus precatorius L.(Rosary Pea) Against Breast Cancer. Cureus. 2024 Aug 26;16(8).
  83. Alvarez-Coiradas E, Munteanu CR, Diaz-Saez L, Pazos A, Huber KV, Loza MI, Dominguez E. Discovery of novel immunopharmacological ligands targeting the IL-17 inflammatory pathway. International Immunopharmacology. 2020 Dec 1;89:107026.
  84. Cosconati S, Forli S, Perryman AL, Harris R, Goodsell DS, Olson AJ. Virtual screening with AutoDock: theory and practice. Expert opinion on drug discovery. 2010 Jun 1;5(6):597-607.
  85. Boutet MA, Nerviani A, Gallo Afflitto G, Pitzalis C. Role of the IL-23/IL-17 axis in psoriasis and psoriatic arthritis: the clinical importance of its divergence in skin and joints. International journal of molecular sciences. 2018 Feb 9;19(2):530.
  86. Torres T, Puig L. Apremilast: a novel oral treatment for psoriasis and psoriatic arthritis. American Journal of clinical dermatology. 2018 Feb;19(1):23-32.
  87. Wohlrab J. Calcineurininhibitoren zur topischen Therapie der Psoriasis. Der Hautarzt. 2006 Aug;57(8):685-9.
  88. Choudhary S, Khan NS, Saxena P, Sahu P, Pradhan D, Singh H, Thomas G, Kumar N. Repurposing FDA approved drugs for psoriasis indications through integrated molecular docking, one-SVM algorithm, and molecular dynamics simulation approaches. Scientific Reports. 2025 Jul 1;15(1):21211.
  89. Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, Yefanov A. NCBI GEO: archive for functional genomics data sets—update. Nucleic acids research. 2012 Nov 26;41(D1):D991-5.
  90. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, Reich M. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. science. 2006 Sep 29;313(5795):1929-35.
  91. Gaulton A, Hersey A, Nowotka M, Bento AP, Chambers J, Mendez D, Mutowo P, Atkinson F, Bellis LJ, Cibrián-Uhalte E, Davies M. The ChEMBL database in 2017. Nucleic acids research. 2017 Jan 4;45(D1):D945-54.
  92. Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H, Shindyalov IN, Bourne PE. The protein data bank. Nucleic acids research. 2000 Jan 1;28(1):235-42.
  93. Szklarczyk D, Kirsch R, Koutrouli M, Nastou K, Mehryary F, Hachilif R, Gable AL, Fang T, Doncheva NT, Pyysalo S, Bork P. The STRING database in 2023: protein–protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic acids research. 2023 Jan 6;51(D1):D638-46.
  94. Song Q, Chen Y, Ma J, Zhou W, Song J, Wu C, Liu J. Metabolomics reveals molecular signatures for psoriasis biomarkers and drug targets discovery. Clinical, Cosmetic and Investigational Dermatology. 2023 Dec 31:3181-91.
  95. Liu S, Li L, Liang Y, Tan Y, Wang X, Feng Y, Chen N, Lei X. Novel genetic insight for psoriasis: integrative genome-wide analyses in 863 080 individuals and proteome-wide Mendelian randomization. Briefings in Bioinformatics. 2025 Jan;26(1):bbaf032.
  96. Xue Y, Xia Y, Cheng D, Shi T, Mei P, Hong S. Association between genetically proxied PPARG activation and psoriasis vulgaris: a Mendelian randomization study. Journal of Dermatological treatment. 2024 Dec 31;35(1):2381763.
  97. Li X, Cao H, Niu M, Liu Q, Liang B, Hou J, Tu J, Gao J. Identification and validation of shared biomarkers and drug repurposing in psoriasis and Crohn’s disease: integrating bioinformatics, machine learning, and experimental approaches. Frontiers in Immunology. 2025 May 8;16:1587705.
  98. Cai YX, Zheng DS, Chen XL, Bai ZP, Zhang J, Deng W, Huang XF. An integrated multi-omics analysis identifies protein biomarkers and potential drug targets for psoriatic arthritis. Communications Biology. 2025 Feb 14;8(1):240.
  99. Zhan YP, Chen BS. Drug target identification and drug repurposing in psoriasis through systems biology approach, DNN-based DTI model and genome-wide microarray data. International Journal of Molecular Sciences. 2023 Jun 12;24(12):10033.

Reference

  1. Gunderson AJ, Mohammed J, Horvath FJ, Podolsky MA, Anderson CR, Glick AB. CD8+ T cells mediate RAS-induced psoriasis-like skin inflammation through IFN-γ. Journal of Investigative Dermatology. 2013 Apr 1;133(4):955-63.
  2. Zhou X, Chen Y, Cui L, Shi Y, Guo C. Advances in the pathogenesis of psoriasis: from keratinocyte perspective. Cell death & disease. 2022 Jan 24;13(1):81.
  3. Brembilla NC, Boehncke WH. Revisiting the interleukin 17 family of cytokines in psoriasis: pathogenesis and potential targets for innovative therapies. Frontiers in immunology. 2023 May 22;14:1186455.
  4. Pietrzak AT, Zalewska A, Chodorowska G, Krasowska D, Michalak-Stoma A, Nockowski P, Osemlak P, Paszkowski T, Roli?ski JM. Cytokines and anticytokines in psoriasis. Clinica chimica acta. 2008 Aug 1;394(1-2):7-21.
  5. Singh R, Koppu S, Perche PO, Feldman SR. The cytokine mediated molecular pathophysiology of psoriasis and its clinical implications. International journal of molecular sciences. 2021 Nov 26;22(23):12793.
  6. Boehncke WH, Schön MP. Animal models of psoriasis. Clinics in dermatology. 2007 Nov 1;25(6):596-605.
  7. Gaspari AA. Innate and adaptive immunity and the pathophysiology of psoriasis. Journal of the American Academy of Dermatology. 2006 Mar 1;54(3):S67-80.
  8. Morizane S, Gallo RL. Antimicrobial peptides in the pathogenesis of psoriasis. The Journal of dermatology. 2012 Mar;39(3):225-30.
  9. Yadav K, Singh D, Singh MR, Minz S, Gnanakani SP, Yadav R, Vora L, Sahu KK, Bagchi A, Chauhan NS, Pradhan M. Preclinical study models of psoriasis: State-of-the-art techniques for testing pharmaceutical products in animal and nonanimal models. International Immunopharmacology. 2023 Apr 1;117:109945.
  10. Rendon A, Schäkel K. Psoriasis pathogenesis and treatment. International journal of molecular sciences. 2019 Mar 23;20(6):1475.
  11. Nestle FO, Turka LA, Nickoloff B. Characterization of dermal dendritic cells in psoriasis. Autostimulation of T lymphocytes and induction of Th1 type cytokines. The Journal of clinical investigation. 1994 Jul 1;94(1):202-9.
  12. Lee HJ, Kim M. Challenges and future trends in the treatment of psoriasis. International journal of molecular sciences. 2023 Aug 28;24(17):13313.
  13. Masoudi-Sobhanzadeh Y, Omidi Y, Amanlou M, Masoudi-Nejad A. Drug databases and their contributions to drug repurposing. Genomics. 2020 Mar 1;112(2):1087-95.
  14. Afonina IS, Van Nuffel E, Beyaert R. Immune responses and therapeutic options in psoriasis. Cellular and Molecular Life Sciences. 2021 Mar;78(6):2709-27.
  15. Golbari NM, Porter ML, Kimball AB. Current guidelines for psoriasis treatment: a work in progress. Cutis. 2018 Mar 1;101(3S):10-2.
  16. Chat VS, Kearns DG, Uppal SK, Han G, Wu JJ. Management of psoriasis with topicals: Applying the 2020 AAD-NPF guidelines of care to clinical practice. Cutis. 2022 Aug 1;110(2 Suppl):8-14.
  17. van den Bogaard EH, Bergboer JG, Vonk-Bergers M, van Vlijmen-Willems IM, Hato SV, van der Valk PG, Schröder JM, Joosten I, Zeeuwen PL, Schalkwijk J. Coal tar induces AHR-dependent skin barrier repair in atopic dermatitis. The Journal of clinical investigation. 2013 Jan 25;123(2).
  18. Bhagwat AP, Madke B. The current advancement in psoriasis. Cureus. 2023 Oct 13;15(10).
  19. Alhammad IM, Aseri AM, Alqahtani SA, Alshaebi MF, Alqahtani SA, Alzahrani RA, Alhaji AA, Alamoudi MK, Bafarat AY, Jad AY, Nuqali AE. A review on updates in management and treatment of psoriasis. Archives of Pharmacy Practice. 2021;12(1-2021):74-8
  20. Brownstone ND, Hong J, Mosca M, Hadeler E, Liao W, Bhutani T, Koo J. Biologic treatments of psoriasis: an update for the clinician. Biologics: Targets and Therapy. 2021 Feb 16:39-51.
  21. Lee HJ, Kim M. Challenges and future trends in the treatment of psoriasis. International journal of molecular sciences. 2023 Aug 28;24(17):13313.
  22. Naik PP. Stem cell therapy as a potential treatment option for psoriasis. Anais Brasileiros de Dermatologia. 2022 Jul 29;97(4):471-7.
  23. Yi RC, Akbik M, Smith LR, Klionsky Y, Feldman SR. Therapeutic Advancements in Psoriasis and Psoriatic Arthritis. Journal of Clinical Medicine. 2025 Feb 16;14(4):1312.
  24. Samarasekera EJ, Sawyer L, Wonderling D, Tucker R, Smith CH. Topical therapies for the treatment of plaque psoriasis: systematic review and network meta?analyses. British Journal of Dermatology. 2013 May 1;168(5):954-67.
  25. Mohs RC, Greig NH. Drug discovery and development: Role of basic biological research. Alzheimer's & Dementia: Translational Research & Clinical Interventions. 2017 Nov 1;3(4):651-7.
  26. Petrova E. Innovation in the pharmaceutical industry: The process of drug discovery and development. InInnovation and marketing in the pharmaceutical industry: emerging practices, research, and policies 2013 Oct 26 (pp. 19-81). New York, NY: Springer New York.
  27. Talevi A, Bellera CL. Challenges and opportunities with drug repurposing: finding strategies to find alternative uses of therapeutics. Expert opinion on drug discovery. 2020 Apr 2;15(4):397-401.
  28. Cha Y, Erez T, Reynolds IJ, Kumar D, Ross J, Koytiger G, Kusko R, Zeskind B, Risso S, Kagan E, Papapetropoulos S. Pharma perspective on drug repurposing. Br J Pharmacol. 2017.
  29. Pushpakom S, Iorio F, Eyers PA, Escott KJ, Hopper S, Wells A, Doig A, Guilliams T, Latimer J, McNamee C, Norris A. Drug repurposing: progress, challenges and recommendations. Nature reviews Drug discovery. 2019 Jan;18(1):41-58.
  30. Parvathaneni V, Kulkarni NS, Muth A, Gupta V. Drug repurposing: a promising tool to accelerate the drug discovery process. Drug discovery today. 2019 Oct 1;24(10):2076-85.
  31. Roessler HI, Knoers NV, van Haelst MM, van Haaften G. Drug repurposing for rare diseases. Trends in pharmacological sciences. 2021 Apr 1;42(4):255-67.
  32. Jonker AH, O’Connor D, Cavaller-Bellaubi M, Fetro C, Gogou M, ’T Hoen PA, de Kort M, Stone H, Valentine N, Pasmooij AM. Drug repurposing for rare: progress and opportunities for the rare disease community. Frontiers in medicine. 2024 Jan 17;11:1352803.
  33. Polamreddy P, Gattu N. The drug repurposing landscape from 2012 to 2017: evolution, challenges, and possible solutions. Drug Discovery Today. 2019 Mar 1;24(3):789-95.
  34. Oprea TI, Bauman JE, Bologa CG, Buranda T, Chigaev A, Edwards BS, Jarvik JW, Gresham HD, Haynes MK, Hjelle B, Hromas R. Drug repurposing from an academic perspective. Drug Discovery Today: Therapeutic Strategies. 2011 Dec 1;8(3-4):61-9.
  35. Jain H, Bhat AR, Dalvi H, Godugu C, Singh SB, Srivastava S. Repurposing approved therapeutics for new indication: Addressing unmet needs in psoriasis treatment. Current Research in Pharmacology and Drug Discovery. 2021 Jan 1;2:100041.
  36. Thatikonda S, Pooladanda V, Godugu C. Repurposing an old drug for new use: Niclosamide in psoriasis?like skin inflammation. Journal of cellular physiology. 2020 Jun;235(6):5270-83.
  37. Jain H, Bhat AR, Dalvi H, Godugu C, Singh SB, Srivastava S. Repurposing approved therapeutics for new indication: Addressing unmet needs in psoriasis treatment. Current Research in Pharmacology and Drug Discovery. 2021 Jan 1;2:100041.
  38. Cousins HC, Nayar G, Altman RB. Computational approaches to drug repurposing: methods, challenges, and opportunities. Annual Review of Biomedical Data Science. 2024 Aug;7.
  39. Tanoli Z, Fernández-Torras A, Özcan UO, Kushnir A, Nader KM, Gadiya Y, Fiorenza L, Ianevski A, Vähä-Koskela M, Miihkinen M, Seemab U. Computational drug repurposing: approaches, evaluation of in silico resources and case studies. Nature Reviews Drug Discovery. 2025 Mar 18:1-22.
  40. Park K. A review of computational drug repurposing. Translational and clinical pharmacology. 2019 Jun 28;27(2):59.
  41. Ibezim A, Onah E, Dim EN, Ntie-Kang F. A computational multi-targeting approach for drug repositioning for psoriasis treatment. BMC Complementary Medicine and Therapies. 2021 Jul 5;21(1):193.
  42. Liu Y, Krueger JG, Bowcock AM. Psoriasis: genetic associations and immune system changes. Genes & Immunity. 2007 Jan;8(1):1-2.
  43. Lowes MA, Suarez-Farinas M, Krueger JG. Immunology of psoriasis. Annual review of immunology. 2014 Mar 21;32(1):227-55.
  44. Sobolev VV, Soboleva AG, Denisova EV, Pechatnikova EA, Dvoryankova E, Korsunskaya IM, Mezentsev A. Proteomic studies of psoriasis. Biomedicines. 2022 Mar 7;10(3):619.
  45. Guo L, Jin H. Research progress of metabolomics in psoriasis. Chinese Medical Journal. 2023 Aug 5;136(15):1805-16.
  46. Koutruba N, Emer J, Lebwohl M. Review of ustekinumab, an interleukin-12 and interleukin-23 inhibitor used for the treatment of plaque psoriasis. Therapeutics and clinical risk management. 2010 Apr 15:123-41.
  47. Tsoi LC, Spain SL, Knight J, Ellinghaus E, Stuart PE, Capon F, Ding J, Li Y, Tejasvi T, Gudjonsson JE, Kang HM. Identification of 15 new psoriasis susceptibility loci highlights the role of innate immunity. Nature genetics. 2012 Dec;44(12):1341-8.
  48. Nanda H, Ponnusamy N, Odumpatta R, Jeyakanthan J, Mohanapriya A. Exploring genetic targets of psoriasis using genome wide association studies (GWAS) for drug repurposing. 3 Biotech. 2020 Feb;10(2):43.
  49. van de Kerkhof PC. From empirical to pathogenesis-based treatments for psoriasis. Journal of Investigative Dermatology. 2022 Jul 1;142(7):1778-85.
  50. Ko M, Oh JM, Kim IW. Drug repositioning prediction for psoriasis using the adverse event reporting database. Frontiers in Medicine. 2023 Mar 23;10:1159453.
  51. Ahmed F, Ho SG, Samantasinghar A, Memon FH, Rahim CS, Soomro AM, Sunildutt N, Kim KH, Choi KH. Drug repurposing in psoriasis, performed by reversal of disease-associated gene expression profiles. Computational and Structural Biotechnology Journal. 2022 Jan 1;20:6097-107.
  52. Wang W, Xu Q, Li B, Li H, Shen S, Wu J, Ge H, Zhang H, Chen S, Chen W, Gao J. Proteomic analysis of psoriatic skin lesions in a Chinese population. Journal of Proteomics. 2021 May 30;240:104207.
  53. Wu L, Cen C, Xie B, Hu L, Huang J, Shen N, Dong Q. Cross-sectional study of proteomic differences between moderate and severe psoriasis. Scientific Reports. 2025 Jan 27;15(1):3387.
  54. Guo Y, Luo L, Zhu J, Li C. Advance in Multi-omics Research Strategies on Cholesterol Metabolism in Psoriasis. Inflammation. 2024 Jun;47(3):839-52.
  55. Stanescu AM, Simionescu AA, Florea M, Diaconu CC. Is metformin a possible beneficial treatment for psoriasis? A scoping review. Journal of Personalized Medicine. 2021 Mar 30;11(4):251.
  56. Devaprasad A, Radstake TR, Pandit A. Integration of immunome with disease-gene network reveals common cellular mechanisms between IMIDs and drug repurposing strategies. Frontiers in Immunology. 2021 May 24;12:669400.
  57. Zhang M, Su W, Deng J, Zhai B, Zhu G, Gao R, Zeng Q, Qiu J, Bian Z, Xiao H, Luan G. Multi-ancestry genome-wide meta-analysis with 472,819 individuals identifies 32 novel risk loci for psoriasis. Journal of Translational Medicine. 2025 Jan 30;23(1):133.
  58. Qu XA, Freudenberg JM, Sanseau P, Rajpal DK. Integrative clinical transcriptomics analyses for new therapeutic intervention strategies: a psoriasis case study. Drug discovery today. 2014 Sep 1;19(9):1364-71.
  59. Rawat A, Rinchai D, Toufiq M, Marr AK, Kino T, Garand M, Tatari-Calderone Z, Kabeer BS, Krishnamoorthy N, Bedognetti D, Karim MY. A neutrophil-driven inflammatory signature characterizes the blood transcriptome fingerprint of psoriasis. Frontiers in immunology. 2020 Nov 24;11:587946.
  60. Arakelyan A, Nersisyan L, Nikoghosyan M, Hakobyan S, Simonyan A, Hopp L, Loeffler-Wirth H, Binder H. Transcriptome-guided drug repositioning. Pharmaceutics. 2019 Dec 12;11(12):677.
  61. Barabási AL, Gulbahce N, Loscalzo J. Network medicine: a network-based approach to human disease. Nature reviews genetics. 2011 Jan;12(1):56-68.
  62. Zhou Y, Hou Y, Shen J, Huang Y, Martin W, Cheng F. Network-based drug repurposing for novel coronavirus 2019-nCoV/SARS-CoV-2. Cell discovery. 2020 Mar 16;6(1):14.
  63. Li YY, Jones SJ. Drug repositioning for personalized medicine. Genome medicine. 2012 Mar 30;4(3):27.
  64. Park D, Jeong HO, Kim BC, Ha YM, Young Chung H. Computational approach to identify enzymes that are potential therapeutic candidates for psoriasis. Enzyme Research. 2011;2011(1):826784.
  65. Müller A, Dickmanns A, Resch C, Schäkel K, Hailfinger S, Dobbelstein M, Schulze-Osthoff K, Kramer D. The CDK4/6-EZH2 pathway is a potential therapeutic target for psoriasis. The Journal of clinical investigation. 2020 Nov 2;130(11):5765-81.
  66. Manczinger M, Bodnár VÁ, Papp BT, Bolla SB, Szabó K, Balázs B, Csányi E, Szél E, Er?s G, Kemény L. Drug repurposing by simulating flow through protein–protein interaction networks. Clinical Pharmacology & Therapeutics. 2018 Mar;103(3):511-20.
  67. Jia Z, Liu Y, Guan N, Bo X, Luo Z, Barnes MR. Cogena, a novel tool for co-expressed gene-set enrichment analysis, applied to drug repositioning and drug mode of action discovery. BMC genomics. 2016 May 27;17(1):414.
  68. Farahani M, Robati RM, Rezaei-Tavirani M, Fateminasab F, Shityakov S, Rahmati Roodsari M, Razzaghi Z, Zamanian Azodi M, Saghari S. Integrating protein interaction and pathway crosstalk network reveals a promising therapeutic approach for psoriasis through apoptosis induction. Scientific reports. 2024 Sep 27;14(1):22103.
  69. Mukhtar MS, Mishra B, Athar M. Integrative systems biology framework discovers common gene regulatory signatures in multiple mechanistically distinct inflammatory skin diseases. Research Square. 2023 Nov 17:rs-3.
  70. Zhang GuiBiao ZG, Li QingYa LQ, Chen QiLong CQ, Su ShiBing SS. Network pharmacology: a new approach for Chinese herbal medicine research.
  71. Menter A, Gelfand JM, Connor C, Armstrong AW, Cordoro KM, Davis DM, Elewski BE, Gordon KB, Gottlieb AB, Kaplan DH, Kavanaugh A. Joint American Academy of Dermatology–National Psoriasis Foundation guidelines of care for the management of psoriasis with systemic nonbiologic therapies. Journal of the American Academy of Dermatology. 2020 Jun 1;82(6):1445-86.
  72. Pincelli C, Schafer PH, French LE, Augustin M, Krueger JG. Mechanisms Underlying the Clinical Effects of Apremilast for Psoriasis. Journal of drugs in dermatology: JDD. 2018 Aug 1;17(8):835-40.
  73. Rigel AS, Lebwohl MG. Hat-wearing patterns in persons attending baseball games. Journal of the American Academy of Dermatology. 2006 May 1;54(5):918-9.
  74. Vamathevan J, Clark D, Czodrowski P, Dunham I, Ferran E, Lee G, Li B, Madabhushi A, Shah P, Spitzer M, Zhao S. Applications of machine learning in drug discovery and development. Nature reviews Drug discovery. 2019 Jun;18(6):463-77.
  75. Wang X, Liu X, Liu N, Chen H. Prediction of crucial epigenetically-associated, differentially expressed genes by integrated bioinformatics analysis and the identification of S100A9 as a novel biomarker in psoriasis. International journal of molecular medicine. 2020 Jan;45(1):93-102.
  76. Esteva A, Kuprel B, Novoa RA, Ko J, Swetter SM, Blau HM, Thrun S. Dermatologist-level classification of skin cancer with deep neural networks. nature. 2017 Feb 2;542(7639):115-8.
  77. Zhang Y, Chen Y, Liu Y, et al. Predicting drug response in psoriasis patients using deep learning. J Biomed Inform. 2021;113:103631.
  78. Kuo C, Chen Y, Wu Y, et al. Drug repurposing for psoriasis: identification of amlodipine as a potential treatment. J Dermatol Sci. 2021;101(2):124-131.
  79. Wang Y, Zhang Y, Liu Y, et al. Using electronic health records to identify potential drug repurposing opportunities for psoriasis. J Am Med Inform Assoc. 2021;28(2):282-290
  80. Batool M, Ahmad B, Choi S. A structure-based drug discovery paradigm. International journal of molecular sciences. 2019 Jun 6;20(11):2783.
  81. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. Journal of computational chemistry. 2010 Jan 30;31(2):455-61.
  82. Baskaran H, Margesan T, Raju K. In Silico Analysis of Bioactive Compounds from Leaves of Abrus precatorius L.(Rosary Pea) Against Breast Cancer. Cureus. 2024 Aug 26;16(8).
  83. Alvarez-Coiradas E, Munteanu CR, Diaz-Saez L, Pazos A, Huber KV, Loza MI, Dominguez E. Discovery of novel immunopharmacological ligands targeting the IL-17 inflammatory pathway. International Immunopharmacology. 2020 Dec 1;89:107026.
  84. Cosconati S, Forli S, Perryman AL, Harris R, Goodsell DS, Olson AJ. Virtual screening with AutoDock: theory and practice. Expert opinion on drug discovery. 2010 Jun 1;5(6):597-607.
  85. Boutet MA, Nerviani A, Gallo Afflitto G, Pitzalis C. Role of the IL-23/IL-17 axis in psoriasis and psoriatic arthritis: the clinical importance of its divergence in skin and joints. International journal of molecular sciences. 2018 Feb 9;19(2):530.
  86. Torres T, Puig L. Apremilast: a novel oral treatment for psoriasis and psoriatic arthritis. American Journal of clinical dermatology. 2018 Feb;19(1):23-32.
  87. Wohlrab J. Calcineurininhibitoren zur topischen Therapie der Psoriasis. Der Hautarzt. 2006 Aug;57(8):685-9.
  88. Choudhary S, Khan NS, Saxena P, Sahu P, Pradhan D, Singh H, Thomas G, Kumar N. Repurposing FDA approved drugs for psoriasis indications through integrated molecular docking, one-SVM algorithm, and molecular dynamics simulation approaches. Scientific Reports. 2025 Jul 1;15(1):21211.
  89. Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, Yefanov A. NCBI GEO: archive for functional genomics data sets—update. Nucleic acids research. 2012 Nov 26;41(D1):D991-5.
  90. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, Reich M. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. science. 2006 Sep 29;313(5795):1929-35.
  91. Gaulton A, Hersey A, Nowotka M, Bento AP, Chambers J, Mendez D, Mutowo P, Atkinson F, Bellis LJ, Cibrián-Uhalte E, Davies M. The ChEMBL database in 2017. Nucleic acids research. 2017 Jan 4;45(D1):D945-54.
  92. Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H, Shindyalov IN, Bourne PE. The protein data bank. Nucleic acids research. 2000 Jan 1;28(1):235-42.
  93. Szklarczyk D, Kirsch R, Koutrouli M, Nastou K, Mehryary F, Hachilif R, Gable AL, Fang T, Doncheva NT, Pyysalo S, Bork P. The STRING database in 2023: protein–protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic acids research. 2023 Jan 6;51(D1):D638-46.
  94. Song Q, Chen Y, Ma J, Zhou W, Song J, Wu C, Liu J. Metabolomics reveals molecular signatures for psoriasis biomarkers and drug targets discovery. Clinical, Cosmetic and Investigational Dermatology. 2023 Dec 31:3181-91.
  95. Liu S, Li L, Liang Y, Tan Y, Wang X, Feng Y, Chen N, Lei X. Novel genetic insight for psoriasis: integrative genome-wide analyses in 863 080 individuals and proteome-wide Mendelian randomization. Briefings in Bioinformatics. 2025 Jan;26(1):bbaf032.
  96. Xue Y, Xia Y, Cheng D, Shi T, Mei P, Hong S. Association between genetically proxied PPARG activation and psoriasis vulgaris: a Mendelian randomization study. Journal of Dermatological treatment. 2024 Dec 31;35(1):2381763.
  97. Li X, Cao H, Niu M, Liu Q, Liang B, Hou J, Tu J, Gao J. Identification and validation of shared biomarkers and drug repurposing in psoriasis and Crohn’s disease: integrating bioinformatics, machine learning, and experimental approaches. Frontiers in Immunology. 2025 May 8;16:1587705.
  98. Cai YX, Zheng DS, Chen XL, Bai ZP, Zhang J, Deng W, Huang XF. An integrated multi-omics analysis identifies protein biomarkers and potential drug targets for psoriatic arthritis. Communications Biology. 2025 Feb 14;8(1):240.
  99. Zhan YP, Chen BS. Drug target identification and drug repurposing in psoriasis through systems biology approach, DNN-based DTI model and genome-wide microarray data. International Journal of Molecular Sciences. 2023 Jun 12;24(12):10033.

Photo
R. Abirami
Corresponding author

College of Pharmacy, Madras Medical College, Chennai-03, Tamil Nadu, India

Photo
R. Indumathy
Co-author

College of Pharmacy, Madras Medical College, Chennai-03, Tamil Nadu, India

Photo
P. Vinodhini
Co-author

College of Pharmacy, Madras Medical College, Chennai-03, Tamil Nadu, India

Photo
V. Soundarya
Co-author

College of Pharmacy, Madras Medical College, Chennai-03, Tamil Nadu, India

R. Abirami, R. Indumathy, P. Vinodhini, V. Soundarya, Drug Repurposing in Psoriasis: Unlocking Therapeutic Insights Through Computational Methods, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 10, 1470-1487. https://doi.org/10.5281/zenodo.17352052

More related articles
Intrauterine Devices-Intrauterine Drug Delivery Sy...
Aamena Simeem, Payal Shelke, Nandini Gire, G. K. Brahma, Diya Gaw...
Inhalation-Based Novel Drug Delivery Systems: Adva...
Omkar Kokane, Shraddha Chavan, Vaishnavi Pawar, Apurva Kamble, G....
Nanostructured Lipid Carriers as Brain-Targeted Drug Delivery Vehicles in Neurod...
Madhuri Sonawane, Vidhya Thorat, Pooja Paliwal, Yashashri Deore, Rohan Sawant, Dr. P. N. Sable, ...
Related Articles
Nose-to-Brain Drug Delivery: A Novel Approach Through Solid Lipid Nanoparticles...
Yashashri Deore , Vidhya Thorat , Dr. Pravinkumar Sable , Madhuri Sonawane , Pooja Paliwal , Rohan S...
A Comprehensive Review of Transdermal Drug Delivery Systems for Antihypertensive...
Vikram T Choudhary, Priya Rajkumar, Dr. Gururaj S Kulkarni, ...
More related articles
Intrauterine Devices-Intrauterine Drug Delivery System...
Aamena Simeem, Payal Shelke, Nandini Gire, G. K. Brahma, Diya Gawade, Sneha Bhalerao, ...
Inhalation-Based Novel Drug Delivery Systems: Advances and Applications...
Omkar Kokane, Shraddha Chavan, Vaishnavi Pawar, Apurva Kamble, G. K. Brahma, ...
Intrauterine Devices-Intrauterine Drug Delivery System...
Aamena Simeem, Payal Shelke, Nandini Gire, G. K. Brahma, Diya Gawade, Sneha Bhalerao, ...
Inhalation-Based Novel Drug Delivery Systems: Advances and Applications...
Omkar Kokane, Shraddha Chavan, Vaishnavi Pawar, Apurva Kamble, G. K. Brahma, ...