Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (706)

Search Parameters:
Keywords = novel antiviral drugs

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 1024 KiB  
Review
SARS-CoV-2 Infection and Antiviral Strategies: Advances and Limitations
by Vinicius Cardoso Soares, Isabela Batista Gonçalves Moreira and Suelen Silva Gomes Dias
Viruses 2025, 17(8), 1064; https://doi.org/10.3390/v17081064 - 30 Jul 2025
Viewed by 510
Abstract
Since the onset of the COVID-19 pandemic, remarkable progress has been made in the development of antiviral therapies for SARS-CoV-2. Several direct-acting antivirals, such as remdesivir, molnupiravir, and nirmatrelvir/ritonavir, offer clinical benefits. These agents have significantly contributed to reducing the viral loads and [...] Read more.
Since the onset of the COVID-19 pandemic, remarkable progress has been made in the development of antiviral therapies for SARS-CoV-2. Several direct-acting antivirals, such as remdesivir, molnupiravir, and nirmatrelvir/ritonavir, offer clinical benefits. These agents have significantly contributed to reducing the viral loads and duration of the illness, as well as the disease’s severity and mortality. However, despite these advances, important limitations remain. The continued emergence of resistant SARS-CoV-2 variants highlights the urgent need for adaptable and durable therapeutic strategies. Therefore, this review aims to provide an updated overview of the main antiviral strategies that are used and the discovery of new drugs against SARS-CoV-2, as well as the therapeutic limitations that have shaped clinical management in recent years. The major challenges include resistance associated with viral mutations, limited treatment windows, and unequal access to treatment. Moreover, there is an ongoing need to identify novel compounds with broad-spectrum activity, improved pharmacokinetics, and suitable safety profiles. Combination treatment regimens represent a promising strategy to increase the efficacy of treating COVID-19 while minimizing the potential for resistance. Ideally, these interventions should be safe, affordable, and easy to administer, which would ensure broad global access and equitable treatment and enable control of COVID-19 cases and preparedness for future threats. Full article
Show Figures

Figure 1

16 pages, 14493 KiB  
Article
Identification of Drug Repurposing Candidates for Coxsackievirus B3 Infection in iPSC-Derived Brain-like Endothelial Cells
by Jacob F. Wood, John M. Vergis, Ali S. Imami, William G. Ryan, Jon J. Sin, Brandon J. Kim, Isaac T. Schiefer and Robert E. McCullumsmith
Int. J. Mol. Sci. 2025, 26(15), 7041; https://doi.org/10.3390/ijms26157041 - 22 Jul 2025
Viewed by 208
Abstract
The enterovirus Coxsackievirus B3 causes a range of serious health problems, including aseptic meningitis, myocarditis, and pancreatitis. Currently, Coxsackievirus B3 has no targeted antiviral treatments or vaccines, leaving supportive care as the primary management option. Understanding how Coxsackievirus B3 interacts with and alters [...] Read more.
The enterovirus Coxsackievirus B3 causes a range of serious health problems, including aseptic meningitis, myocarditis, and pancreatitis. Currently, Coxsackievirus B3 has no targeted antiviral treatments or vaccines, leaving supportive care as the primary management option. Understanding how Coxsackievirus B3 interacts with and alters the blood–brain barrier may help identify new therapies to combat this often-devastating infection. We reanalyzed a previously published RNA sequencing dataset for Coxsackievirus B3-infected human-induced pluripotent stem-cell-derived brain endothelial cells (iBECs) to examine how Coxsackievirus B3 altered mRNA expression. By integrating GSEA, EnrichR, and iLINCs-based perturbagen analysis, we present a novel, systems-level approach to uncover potential drug repurposing candidates for CVB3 infection. We found dynamic changes in host transcriptomic response to Coxsackievirus B3 infection at 2- and 5-day infection time points. Downregulated pathways included ribosomal biogenesis and protein synthesis, while upregulated pathways included a defense response to viruses, and interferon production. Using iLINCs transcriptomic analysis, MEK, PDGFR, and VEGF inhibitors were identified as possible novel antiviral therapeutics. Our findings further elucidate Coxsackievirus B3-associated pathways in (iBECs) and highlight potential drug repurposing candidates, including pelitinib and neratinib, which may disrupt Coxsackievirus B3 pathology at the blood–brain barrier (BBB). Full article
Show Figures

Figure 1

29 pages, 2729 KiB  
Article
Computational Evaluation and Multi-Criteria Optimization of Natural Compound Analogs Targeting SARS-CoV-2 Proteases
by Paul Andrei Negru, Andrei-Flavius Radu, Ada Radu, Delia Mirela Tit and Gabriela Bungau
Curr. Issues Mol. Biol. 2025, 47(7), 577; https://doi.org/10.3390/cimb47070577 - 21 Jul 2025
Viewed by 388
Abstract
The global impact of the COVID-19 crisis has underscored the need for novel therapeutic candidates capable of efficiently targeting essential viral proteins. Existing therapeutic strategies continue to encounter limitations such as reduced efficacy against emerging variants, safety concerns, and suboptimal pharmacodynamics, which emphasize [...] Read more.
The global impact of the COVID-19 crisis has underscored the need for novel therapeutic candidates capable of efficiently targeting essential viral proteins. Existing therapeutic strategies continue to encounter limitations such as reduced efficacy against emerging variants, safety concerns, and suboptimal pharmacodynamics, which emphasize the potential of natural-origin compounds as supportive agents with immunomodulatory, anti-inflammatory, and antioxidant benefits. The present study significantly advances prior molecular docking research through comprehensive virtual screening of structurally related analogs derived from antiviral phytochemicals. These compounds were evaluated specifically against the SARS-CoV-2 main protease (3CLpro) and papain-like protease (PLpro). Utilizing chemical similarity algorithms via the ChEMBL database, over 600 candidate molecules were retrieved and subjected to automated docking, interaction pattern analysis, and comprehensive ADMET profiling. Several analogs showed enhanced binding scores relative to their parent scaffolds, with CHEMBL1720210 (a shogaol-derived analog) demonstrating strong interaction with PLpro (−9.34 kcal/mol), and CHEMBL1495225 (a 6-gingerol derivative) showing high affinity for 3CLpro (−8.04 kcal/mol). Molecular interaction analysis revealed that CHEMBL1720210 forms hydrogen bonds with key PLpro residues including GLY163, LEU162, GLN269, TYR265, and TYR273, complemented by hydrophobic interactions with TYR268 and PRO248. CHEMBL1495225 establishes multiple hydrogen bonds with the 3CLpro residues ASP197, ARG131, TYR239, LEU272, and GLY195, along with hydrophobic contacts with LEU287. Gene expression predictions via DIGEP-Pred indicated that the top-ranked compounds could influence biological pathways linked to inflammation and oxidative stress, processes implicated in COVID-19’s pathology. Notably, CHEMBL4069090 emerged as a lead compound with favorable drug-likeness and predicted binding to PLpro. Overall, the applied in silico framework facilitated the rational prioritization of bioactive analogs with promising pharmacological profiles, supporting their advancement toward experimental validation and therapeutic exploration against SARS-CoV-2. Full article
(This article belongs to the Special Issue Novel Drugs and Natural Products Discovery)
Show Figures

Figure 1

14 pages, 2021 KiB  
Review
New Advances in Small Molecules Targeted at Viral Capsid–Genome Binding
by Jiamei Li, Chengfeng Zhang, Benteng Li and Yuqing Wu
Int. J. Mol. Sci. 2025, 26(14), 6979; https://doi.org/10.3390/ijms26146979 - 20 Jul 2025
Viewed by 362
Abstract
The capsid protein plays a crucial role in the viral life cycle. By interacting with the viral genome, it facilitates the assembly of the nucleocapsid, ultimately leading to the formation of the viral particle. Therefore, interfering with or disrupting the interaction between the [...] Read more.
The capsid protein plays a crucial role in the viral life cycle. By interacting with the viral genome, it facilitates the assembly of the nucleocapsid, ultimately leading to the formation of the viral particle. Therefore, interfering with or disrupting the interaction between the capsid protein and viral genome can effectively inhibit viral replication and infection. This review focuses on elucidating the binding mechanisms between the capsid protein and the viral genome, as well as their potential applications as therapeutic targets. In particular, it summarizes the research progress on small-molecule drugs targeting the capsid–genome binding sites of dengue virus, HBV, and SARS-CoV-2. Notably, this review provides a detailed discussion on the mechanisms by which these small-molecule inhibitors interfere with the capsid–genome interaction, aiming to offer inspiration for the future development of novel antiviral drugs targeting the capsid–genome binding. Full article
(This article belongs to the Special Issue Latest Review Papers in Macromolecules 2025)
Show Figures

Figure 1

15 pages, 2357 KiB  
Article
Development of a Novel, Highly Sensitive System for Evaluating Ebola Virus Particle Formation
by Wakako Furuyama, Miako Sakaguchi, Hanako Ariyoshi and Asuka Nanbo
Viruses 2025, 17(7), 1016; https://doi.org/10.3390/v17071016 - 19 Jul 2025
Viewed by 476
Abstract
Ebola virus (EBOV) causes severe hemorrhagic fevers in humans, and effective countermeasures remain limited. The EBOV-encoded major matrix protein VP40 is essential for viral assembly, budding, and particle release, making it a promising target for antiviral drug development. However, no approved drugs currently [...] Read more.
Ebola virus (EBOV) causes severe hemorrhagic fevers in humans, and effective countermeasures remain limited. The EBOV-encoded major matrix protein VP40 is essential for viral assembly, budding, and particle release, making it a promising target for antiviral drug development. However, no approved drugs currently target the viral particle formation process. In this study, we established a simple and highly sensitive screening system to evaluate VP40-mediated virus-like particle (VLP) formation under biosafety level −2 conditions. The system uses the HiBiT luminescence-based reporter fused to VP40, allowing for the detection of VP40 release. Our results demonstrate that the HiBiT sequence fused at the N-terminus [HiBiT-VP40 (N)] retains VP40′s ability to form VLPs, supporting its use as a functional reporter. Furthermore, we validated the system by assessing the role of Rab11-dependent trafficking in VP40-mediated budding and by evaluating the effect of nocodazole, a microtubule depolymerizer, on VLP release. This novel screening system provides a convenient and reliable platform for screening potential inhibitors targeting the late stages of EBOV infection, including viral particle formation and release. Additionally, its potential adaptability to other filoviruses suggests wide applicability in the discovery and development of additional novel therapeutic agents. Full article
Show Figures

Figure 1

23 pages, 846 KiB  
Review
Multifaceted Marine Peptides and Their Therapeutic Potential
by Svetlana V. Guryanova and Tatiana V. Ovchinnikova
Mar. Drugs 2025, 23(7), 288; https://doi.org/10.3390/md23070288 - 15 Jul 2025
Viewed by 664
Abstract
Marine peptides, derived from a great number of aquatic organisms, exhibit a broad spectrum of biological activities that hold a significant therapeutic potential. This article reviews the multifaceted roles of marine peptides, focusing on their antibacterial, antibiofilm, antifungal, antiviral, antiparasitic, cytotoxic, anticancer, immunomodulatory, [...] Read more.
Marine peptides, derived from a great number of aquatic organisms, exhibit a broad spectrum of biological activities that hold a significant therapeutic potential. This article reviews the multifaceted roles of marine peptides, focusing on their antibacterial, antibiofilm, antifungal, antiviral, antiparasitic, cytotoxic, anticancer, immunomodulatory, chemotactic, opsonizing, anti-inflammatory, antiaging, skin-protective, and wound-healing properties. By elucidating mechanisms of their action and highlighting key research findings, this review aims to provide a comprehensive understanding of possible therapeutic applications of marine peptides, underscoring their importance in developing novel drugs as well as in cosmetology, food industry, aquatic and agriculture biotechnology. Further investigations are essential to harness their therapeutic potential and should focus on detailed mechanism studies, large-scale production, and clinical evaluations with a view to confirm their efficacy and safety and translate these findings into practical applications. It is also important to investigate the potential synergistic effects of marine peptide combinations with existing medicines to enhance their efficacy. Challenges include the sustainable sourcing of marine peptides, and therefore an environmental impact of harvesting marine organisms must be considered as well. Full article
Show Figures

Figure 1

29 pages, 3105 KiB  
Review
Uncaria tomentosa as a Promising Natural Source of Molecules with Multiple Activities: Review of Its Ethnomedicinal Uses, Phytochemistry and Pharmacology
by Olinda Marques, Artur Figueirinha, Maria Eugénia Pina and Maria Teresa Batista
Int. J. Mol. Sci. 2025, 26(14), 6758; https://doi.org/10.3390/ijms26146758 - 15 Jul 2025
Viewed by 487
Abstract
Uncaria tomentosa (Ut) is a Rubiaceae widely used in Peru’s traditional medicine. It is mainly known by the vernacular name of Cat’s claw due to its morphological aspects and is found in tropical low mountain forests of Central and South America. [...] Read more.
Uncaria tomentosa (Ut) is a Rubiaceae widely used in Peru’s traditional medicine. It is mainly known by the vernacular name of Cat’s claw due to its morphological aspects and is found in tropical low mountain forests of Central and South America. A decoction of Ut bark, root and leaves is used traditionally for different health problems, including arthritis, weakness, viral infections, skin disorders, abscesses, allergies, asthma, cancer, fevers, gastric ulcers, haemorrhages, inflammations, menstrual irregularity, rheumatism, urinary tract inflammation and wounds, among others, which gave rise to scientific and commercial interest. The present paper reviews research progress relating to the ethnobotany, phytochemistry and pharmacology of Ut, and some promising research routes are also discussed. We highlight the centrality of its different biological activities, such as antioxidant, anti-inflammatory, antiproliferative, antiviral, and antinociceptive, among others. Recently, studies of the health effects of this plant suggest that novel nutraceuticals can be obtained from it and applied as a preventive or prophylaxis strategy before the start of conventional drug therapy, especially for patients who are not prone to conventional pharmacological approaches to diseases. The present work emphasizes the current pharmacological properties of Uncaria tomentosa, evidencing its therapeutic benefits and encouraging further research on this medicinal plant. Full article
(This article belongs to the Special Issue Current Research in Pharmacognosy: A Focus on Biological Activities)
Show Figures

Figure 1

25 pages, 3522 KiB  
Article
Repurposing of Some Nucleoside Analogs Targeting Some Key Proteins of the Avian H5N1 Clade 2.3.4.4b to Combat the Circulating HPAI in Birds: An In Silico Approach
by Mohd Yasir Khan, Abid Ullah Shah, Nithyadevi Duraisamy, Mohammed Cherkaoui and Maged Gomaa Hemida
Viruses 2025, 17(7), 972; https://doi.org/10.3390/v17070972 - 10 Jul 2025
Viewed by 493
Abstract
(1) Background: The highly pathogenic avian influenza virus H5N1 clade 2.3.4.4b is an emerging threat that poses a great risk to the poultry industry. A few human cases have been linked to the infection with this clade in many parts of the world, [...] Read more.
(1) Background: The highly pathogenic avian influenza virus H5N1 clade 2.3.4.4b is an emerging threat that poses a great risk to the poultry industry. A few human cases have been linked to the infection with this clade in many parts of the world, including the USA. Unfortunately, there are no specific vaccines or antiviral drugs that could help prevent and treat the infection caused by this virus in birds. Our major objective is to identify/repurpose some (novel/known) antiviral compounds that may inhibit viral replication by targeting some key viral proteins. (2) Methods: We used state-of-the-art machine learning tools such as molecular docking and MD-simulation methods from Biovia Discovery Studio (v24.1.0.321712). The key target proteins such as hemagglutinin (HA), neuraminidase (NA), Matrix-2 protein (M2), and the cap-binding domain of PB2 (PB2/CBD) homology models were validated through structural assessment via DOPE scores, Ramachandran plots, and Verify-3D metrics, ensuring reliable structural representations, confirming their reliability for subsequent in silico approaches. These approaches include molecular docking followed by molecular dynamics simulation for 50 nanoseconds (ns), highlighting the structural stability and compactness of the docked complexes. (3) Results: Molecular docking revealed strong binding affinities for both sofosbuvir and GS441524, particularly with the NA and PB2/CBD protein targets. Among them, GS441524 exhibited superior interaction scores and a greater number of hydrogen bonds with key functional residues of NA and PB2/CBD. The MM-GBSA binding free energy calculations further supported these findings, as GS441524 displayed more favorable binding energies compared to several known standard inhibitors, including F0045S for HA, Zanamivir for NA, Rimantadine and Amantadine for M2, and PB2-39 for PB2/CBD. Additionally, 50 ns molecular dynamics simulations highlighted the structural stability and compactness of the GS441524-PB2/CBD complex, further supporting its potential as a promising antiviral candidate. Furthermore, hydrogen bond monitor analysis over the 50 ns simulation confirmed persistent and specific interactions between the ligand and proteins, suggesting that GS441524 may effectively inhibit the NA, and PB2/CBD might potentially disrupt PB2-mediated RNA synthesis. (4) Conclusions: Our findings are consistent with previous evidence supporting the antiviral activity of certain nucleoside analog inhibitors, including GS441524, against various coronaviruses. These results further support the potential repurposing of GS441524 as a promising therapeutic candidate against H5N1 avian influenza clade 2.3.4.4b. However, further functional studies are required to validate these in silico predictions and support the inhibitory action of GS441524 against the targeted proteins of H5N1, specifically clade 2.3.4.4b. Full article
(This article belongs to the Special Issue Interplay Between Influenza Virus and Host Factors)
Show Figures

Figure 1

14 pages, 694 KiB  
Article
In Vitro Antiviral Activity of the Fungal Metabolite 6-Pentyl-α-Pyrone Against Bovine Coronavirus: A Translational Study to SARS-CoV-2
by Violetta Iris Vasinioti, Amienwanlen Eugene Odigie, Maria Stella Lucente, Luca Del Sorbo, Cristiana Catella, Elisabetta Casalino, Maria Michela Salvatore, Alessia Staropoli, Francesco Vinale, Maria Tempesta, Filomena Fiorito, Anna Andolfi, Alessio Buonavoglia, Annamaria Pratelli and Paolo Capozza
Vet. Sci. 2025, 12(7), 634; https://doi.org/10.3390/vetsci12070634 - 2 Jul 2025
Viewed by 740
Abstract
The recent COVID-19 pandemic has prompted the scientific community to prioritize the discovery of preventive methods and new therapeutics, including the investigation of natural compounds with antiviral potential. Fungal secondary metabolites (SMs) represent a promising source of antiviral drugs due to their structural [...] Read more.
The recent COVID-19 pandemic has prompted the scientific community to prioritize the discovery of preventive methods and new therapeutics, including the investigation of natural compounds with antiviral potential. Fungal secondary metabolites (SMs) represent a promising source of antiviral drugs due to their structural diversity and intrinsic biocompatibility. Herein, the antiviral activity of 6-pentyl-α-pyrone (6PP) against bovine coronavirus (BCoV) has been evaluated in vitro. Considering that BCoV and SARS-CoV-2 are both members of the Betacoronavirus genus and share several key features, BCoV represents a valuable reference model for human coronavirus research. A non-cytotoxic dose of 6PP was used on MDBK cells to evaluate its antiviral activity against BCoV. Different experimental conditions were employed to examine cell monolayer protection both pre- and post-infection, as well as the potential inhibition of viral internalization. Overall, post-infection 6PP treatment reduced viral load and decreased viral internalization. Results were analyzed using viral titration and quantitative PCR, while data interpretation was performed by statistical software tools. This study presents a novel fluorescence quantification approach with high confidence demonstrated by its significant concordance with RT-qPCR results. These data suggest that 6PP could be an effective antiviral agent for BCoV, warranting further investigation of its role in coronavirus inhibition. Full article
(This article belongs to the Section Veterinary Microbiology, Parasitology and Immunology)
Show Figures

Graphical abstract

17 pages, 5007 KiB  
Review
PROTAC-Based Antivirals for Respiratory Viruses: A Novel Approach for Targeted Therapy and Vaccine Development
by Amith Anugu, Pankaj Singh, Dharambir Kashyap, Jillwin Joseph, Sheetal Naik, Subhabrata Sarkar, Kamran Zaman, Manpreet Dhaliwal, Shubham Nagar, Tanishq Gupta and Prasanna Honnavar
Microorganisms 2025, 13(7), 1557; https://doi.org/10.3390/microorganisms13071557 - 2 Jul 2025
Viewed by 540
Abstract
The global burden of respiratory viral infections is notable, which is attributed to their higher transmissibility compared to other viral diseases. Respiratory viruses are seen to have evolved resistance to available treatment options. Although vaccines and antiviral drugs control some respiratory viruses, this [...] Read more.
The global burden of respiratory viral infections is notable, which is attributed to their higher transmissibility compared to other viral diseases. Respiratory viruses are seen to have evolved resistance to available treatment options. Although vaccines and antiviral drugs control some respiratory viruses, this control is limited due to unexpected events, such as mutations and the development of antiviral resistance. The technology of proteolysis-targeting chimeras (PROTACs) has been emerging as a novel technology in viral therapeutics. These are small molecules that can selectively degrade target proteins via the ubiquitin–proteasome pathway. PROTACs as a therapy were initially developed against cancer, but they have recently shown promising results in their antiviral mechanisms by targeting viral and/or host proteins involved in the pathogenesis of viral infections. In this review, we elaborate on the antiviral potential of PROTACs as therapeutic agents and their potential as vaccine components against important respiratory viral pathogens, including influenza viruses, coronaviruses (SARS-CoV-2), and respiratory syncytial virus. Advanced applications of PROTAC antiviral strategies, such as hemagglutinin and neuraminidase degraders for influenza and spike proteins of SARS-CoV-2, are detailed in this review. Additionally, the role of PROTACs in targeting cellular mechanisms within the host, thereby preventing viral pathogenesis and eliciting an antiviral effect, is discussed. The potential of PROTACs as vaccines, utilizing proteasome-based virus attenuation to achieve a robust protective immune response, while ensuring safety and enhancing efficient production, is also presented. With the promises exhibited by PROTACs, this technology faces significant challenges, including the emergence of novel viral strains, tissue-specific expression of E3 ligases, and pharmacokinetic constraints. With advanced computational design in molecular platforms, PROTAC-based antiviral development offers an alternative, transformative path in tackling respiratory viruses. Full article
Show Figures

Figure 1

15 pages, 2600 KiB  
Article
Substituted Triazole-3,5-Diamine Compounds as Novel Human Topoisomerase III Beta Inhibitors
by Yasir Mamun, Somaia Haque Chadni, Ramanjaneyulu Rayala, Hasham Shafi, Shomita Ferdous, Rudramani Pokhrel, Adel Nefzi, Prem Chapagain and Yuk-Ching Tse-Dinh
Int. J. Mol. Sci. 2025, 26(13), 6193; https://doi.org/10.3390/ijms26136193 - 27 Jun 2025
Viewed by 469
Abstract
Human topoisomerase III beta (hTOP3B) is a unique and important enzyme in human cells that plays a role in maintaining genome stability, affecting cellular aging, and potentially impacting viral replication. Its dual activity on both DNA and RNA makes it a valuable target [...] Read more.
Human topoisomerase III beta (hTOP3B) is a unique and important enzyme in human cells that plays a role in maintaining genome stability, affecting cellular aging, and potentially impacting viral replication. Its dual activity on both DNA and RNA makes it a valuable target for therapeutic interventions. hTOP3B has been shown to be required for the efficient replication of certain positive-sense ssRNA viruses including Dengue. We performed in silico screening of a library comprising drugs that are FDA-approved or undergoing clinical trials as potential drugs to identify potential inhibitors of hTOP3B. The topoisomerase activity assay of the identified virtual hits showed that bemcentinib, a compound known to target the AXL receptor tyrosine kinase, can inhibit hTOP3B relaxation activity. This is the first small molecule shown to inhibit the complete catalytic cycle of hTOP3B for the potential interference of the function of hTOP3B in antiviral application. Additional small molecules that share the N5,N3-1H-1,2,4-triazole-3,5-diamine moiety of bemcentinib were synthesized and tested for the inhibition of hTOP3B relaxation activity. Five compounds with comparable IC50 to that of bemcentinib for the inhibition of hTOP3B were identified. These results suggest that the exploration of tyrosine kinase inhibitors and their analogs may allow the identification of novel potential topoisomerase inhibitors. Full article
(This article belongs to the Special Issue Small Molecule Drug Design and Research: 3rd Edition)
Show Figures

Figure 1

25 pages, 3008 KiB  
Review
Deep Generative Models for the Discovery of Antiviral Peptides Targeting Dengue Virus: A Systematic Review
by Huynh Anh Duy and Tarapong Srisongkram
Int. J. Mol. Sci. 2025, 26(13), 6159; https://doi.org/10.3390/ijms26136159 - 26 Jun 2025
Cited by 1 | Viewed by 496
Abstract
Dengue virus (DENV) remains a critical global health challenge, with no approved antiviral treatments currently available. The growing prevalence of DENV infections highlights the urgent need for effective therapeutics. Antiviral peptides (AVPs) have gained significant attention due to their potential to inhibit viral [...] Read more.
Dengue virus (DENV) remains a critical global health challenge, with no approved antiviral treatments currently available. The growing prevalence of DENV infections highlights the urgent need for effective therapeutics. Antiviral peptides (AVPs) have gained significant attention due to their potential to inhibit viral replication. However, traditional drug discovery methods are often time-consuming and resource-intensive. Advances in artificial intelligence, particularly deep generative models (DGMs), offer a promising approach to accelerating AVP discovery. This report provides a comprehensive assessment of the role of DGMs in identifying novel AVPs for DENV. It presents an extensive survey of existing antimicrobial and AVP datasets, peptide sequence feature representations, and the integration of DGMs into computational peptide design. Additionally, in vitro and in silico screening data from previous studies highlight the therapeutic potential of AVPs against DENV. Variational autoencoders and generative adversarial networks have been extensively documented in the literature for their applications in AVP generation. These models have demonstrated a remarkable capacity to generate diverse and structurally viable compounds, significantly expanding the repertoire of potential antiviral candidates. Additionally, this report assesses both the strengths and limitations of DGMs, providing valuable insights for guiding future research directions. As a data-driven and scalable framework, DGMs offer a promising avenue for the rational design of potent AVPs targeting DENV and other emerging viral pathogens, contributing to the advancement of next-generation therapeutic strategies. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

11 pages, 652 KiB  
Article
Optimized Enoxolone-Loaded Microsponges for Drug Delivery: A Design of Experiments Approach
by Juste Baranauskaite, Sedef Sefer, Augusta Zevzikoviene, Andrejus Zevzikovas and Liudas Ivanauskas
Pharmaceuticals 2025, 18(7), 938; https://doi.org/10.3390/ph18070938 - 21 Jun 2025
Viewed by 399
Abstract
Enoxolon is widely recognized for its pharmacological potential, exhibiting antioxidant, anti-inflammatory, anticancer, and antiviral properties. Objectives: This study aimed to develop an enhanced formulation of enoxolone-loaded microsponges as a novel drug delivery system. A design of experiments (DoE) approach was employed for [...] Read more.
Enoxolon is widely recognized for its pharmacological potential, exhibiting antioxidant, anti-inflammatory, anticancer, and antiviral properties. Objectives: This study aimed to develop an enhanced formulation of enoxolone-loaded microsponges as a novel drug delivery system. A design of experiments (DoE) approach was employed for the optimization process. Methods: The microsponges were produced using the quasi-emulsion technique. The selected formulation was evaluated for yield, particle size, and entrapment efficiency. Furthermore, the microsponges were incorporated into a 1% MC solution matrix, and in vitro release studies were performed to assess their drug delivery performance. Results: The optimal formulation was determined through the DoE methodology, which involved varying the concentrations of methylcellulose (MC) (0.55–1.87%, w/w), polyvinyl alcohol (PVA) (0.5–1%, w/w), and Tween 80 (TW80) (1.5–2.5%, w/w). The results showed a particle size of 142.8 ± 10.02 µm and an entrapment efficiency of 80.3 ± 1.99%. When comparing the optimized microsponge formulation to pure enoxolon, a 1.29 times higher release rate was observed (p ≤ 0.05). Conclusions: Following optimizationand physicochemical characterization studies were conducted to further assess the formulation. These findings suggest that microsponge-based delivery systems hold considerable potential as an alternative platform for the topical treatment of chronic periodontitis. Full article
(This article belongs to the Section Pharmaceutical Technology)
Show Figures

Graphical abstract

19 pages, 3638 KiB  
Article
Purification and Inhibitor Screening of the Full-Length SARS-CoV-2 Nucleocapsid Protein
by Chen Chen, Zhengfu Zhang, Qiao Zheng, Yingshun Zhou and Shujun Zhang
Molecules 2025, 30(13), 2679; https://doi.org/10.3390/molecules30132679 - 20 Jun 2025
Viewed by 362
Abstract
Severe acute respiratory syndrome coronavirus 2 has undergone several mutations since 2020, and novel variants continue to emerge to this day. The immune escape ability of the emerging mutants is enhanced and results in robust transmissibility. The neutralizing ability of the antibodies produced [...] Read more.
Severe acute respiratory syndrome coronavirus 2 has undergone several mutations since 2020, and novel variants continue to emerge to this day. The immune escape ability of the emerging mutants is enhanced and results in robust transmissibility. The neutralizing ability of the antibodies produced in the human body during previous infections is decreased against some of these mutants, which poses a severe challenge to the preventive and therapeutic effectiveness of vaccines and antibody drugs. The nucleocapsid protein is one of the main structural proteins of the coronavirus and plays an important role in the life cycle of the novel coronavirus. This protein is one of the key targets for drug development, and the first major step in drug development is to obtain pure nucleocapsid proteins. However, since nucleocapsid proteins have a nucleic acid-binding function and automatically undergo liquid–liquid phase separation and agglomeration, the purification of full-length nucleocapsids is challenging. In this context, a set of easy-to-operate processes was developed in this study for the purification of nucleocapsid proteins. Finally, a pure full-length nucleocapsid protein without nucleic acid contamination was obtained, which exhibited significantly enhanced accessibility for structural and functional virological studies, vaccine development, and related research applications. Further, the nucleic acid-binding domain of the nucleocapsid protein was targeted, and potential severe acute respiratory syndrome coronavirus 2 inhibitors were identified using virtual screening and biolayer interferometry technology. Notably, the eukaryotically expressed nucleocapsid protein demonstrated a significantly greater binding affinity for Light Green SF Yellowish (KD = 119.7 nM) compared to that demonstrated by its prokaryotic counterpart (KD = 19.9 × 103 nM). The findings of this study suggest the importance of considering both protein source and post-translational modifications of the target proteins to be used in drug screening workflows. Therefore, this compound not only represents a novel therapeutic candidate for COVID-19 but also a critical tool for elucidating antiviral mechanisms. Full article
Show Figures

Graphical abstract

14 pages, 2607 KiB  
Article
Anti-Influenza A Virus Activity of Rhododendron brachycarpum Extract and Identification of Hyperoside as the Active Constituent
by Yung Hun Park, Soo Yong Shin, Hayeong Choi, Jae Hyeok Lee, You Jin Kim, Seong Ji Woo, Wonkyun Ronny Im and Sung Ho Jeon
Microbiol. Res. 2025, 16(6), 132; https://doi.org/10.3390/microbiolres16060132 - 18 Jun 2025
Viewed by 361
Abstract
Influenza A virus (IAV) poses significant public health challenges due to its rapid mutation and drug resistance, necessitating novel antiviral strategies. Rhododendron brachycarpum, traditionally employed in folk medicine to treat inflammatory conditions, contains bioactive flavonoids with potential antiviral effects. In this study, [...] Read more.
Influenza A virus (IAV) poses significant public health challenges due to its rapid mutation and drug resistance, necessitating novel antiviral strategies. Rhododendron brachycarpum, traditionally employed in folk medicine to treat inflammatory conditions, contains bioactive flavonoids with potential antiviral effects. In this study, we investigated the anti-influenza activity of R. brachycarpum leaf extract and identified hyperoside (quercetin-3-O-galactoside) as the active constituent. The crude extract and its n-butanol fraction markedly reduced IAV replication in Madin–Darby canine kidney (MDCK) cells, with IC50/CC50 values of 74.51/201.09 μg/mL and 24.5/113.1 μg/mL, respectively. Hyperoside, purified via bioactivity-guided fractionation and HPLC analysis, demonstrated potent antiviral activity, with an IC50 of 66.59 μM (30.92 μg/mL) and a CC50 of 318.9 μM (148.1 μg/mL), indicating a favorable selectivity index. It significantly suppressed viral mRNA and protein expression in infected cells. Time-of-addition and hemagglutination inhibition assays suggested that hyperoside exerts antiviral effects during early infection stages, likely interfering with viral entry. In silico molecular docking analysis further supported this mechanism, revealing that hyperoside binds strongly to the receptor-binding domain of hemagglutinin (−11.5 kcal/mol), potentially blocking viral attachment. These findings reveal that hyperoside is a major antiviral component of R. brachycarpum and underscore its therapeutic potential as a natural antiviral candidate against IAV infections. Full article
Show Figures

Figure 1

Back to TopTop