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

Search Results (678)

Search Parameters:
Keywords = novel antiviral drugs

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 2370 KiB  
Article
Designing Effective Drug Therapies Using a Multiobjective Spider-Wasp Optimizer
by Trong-The Nguyen, Thi-Kien Dao, Van-Thien Nguyen and Duc-Tinh Pham
Biomimetics 2025, 10(4), 219; https://doi.org/10.3390/biomimetics10040219 - 2 Apr 2025
Viewed by 42
Abstract
Designing effective drug therapies requires balancing competing objectives, such as therapeutic efficacy, safety, and cost efficiency—a task that poses significant challenges for conventional optimization methods. To address this, we propose the multi-objective spider–wasp optimizer (MOSWO), a novel approach uniquely emulating the cooperative predation [...] Read more.
Designing effective drug therapies requires balancing competing objectives, such as therapeutic efficacy, safety, and cost efficiency—a task that poses significant challenges for conventional optimization methods. To address this, we propose the multi-objective spider–wasp optimizer (MOSWO), a novel approach uniquely emulating the cooperative predation dynamics between spiders and wasps observed in nature. MOSWO integrates adaptive mechanisms for exploration and exploitation to resolve complex trade-offs in multiobjective drug design. Unlike existing approaches, the algorithm employs a dynamic population-partitioning strategy inspired by predator–prey interactions, enabling efficient Pareto frontier discovery. We validate MOSWO’s performance through extensive experiments on synthetic benchmarks and real-world case studies spanning antiviral and antibiotic therapies. Results demonstrate that MOSWO surpasses state-of-the-art methods (NSGA-II, MOEA/D, MOGWO, and MOPSO), achieving 11% higher hypervolume scores, 8% lower inverted generational distance scores, 9% higher spread scores, a 30% faster convergence, and superior robustness against noisy biological datasets. The framework’s adaptability to diverse therapeutic scenarios underscores its potential as a transformative tool for computational pharmacology. Full article
Show Figures

Figure 1

24 pages, 9293 KiB  
Article
Nanotechnology-Driven Strategy Against SARS-CoV-2: Pluronic F127-Based Nanomicelles with or Without Atazanavir Reduce Viral Replication in Calu-3 Cells
by Eduardo Ricci-Junior, Alice Santos Rosa, Tatielle do Nascimento, Ralph Santos-Oliveira, Marcos Alexandre Nunes da Silva, Debora Ferreira Barreto-Vieira, Luísa Tozatto Batista, Giovanna Barbosa da Conceição, Tayane Alvites Nunes Quintão, Vivian Neuza Santos Ferreira and Milene Dias Miranda
Viruses 2025, 17(4), 518; https://doi.org/10.3390/v17040518 - 1 Apr 2025
Viewed by 62
Abstract
Despite extensive efforts, no highly effective antiviral molecule exists for treating moderate and severe COVID-19. Nanotechnology has emerged as a promising approach for developing novel drug delivery systems to enhance antiviral efficacy. Among these, polymeric nanomicelles improve the solubility, bioavailability, and cellular uptake [...] Read more.
Despite extensive efforts, no highly effective antiviral molecule exists for treating moderate and severe COVID-19. Nanotechnology has emerged as a promising approach for developing novel drug delivery systems to enhance antiviral efficacy. Among these, polymeric nanomicelles improve the solubility, bioavailability, and cellular uptake of therapeutic agents. In this study, Pluronic F127-based nanomicelles were developed and evaluated for their antiviral activity against SARS-CoV-2. The nanomicelles, formulated using the direct dissolution method, exhibited an average size of 37.4 ± 8.01 nm and a polydispersity index (PDI) of 0.427 ± 0.01. Their antiviral efficacy was assessed in SARS-CoV-2-infected Vero E6 and Calu-3 cell models, where treatment with a 1:2 dilution inhibited viral replication by more than 90%. Cytotoxicity assays confirmed the nanomicelles were non-toxic to both cell lines after 72 h. In SARS-CoV-2-infected Calu-3 cells (human type II pneumocyte model), treatment with Pluronic F127-based nanomicelles containing atazanavir (ATV) significantly reduced viral replication, even under high MOI (2) and after 48 h, while also preventing IL-6 upregulation. To investigate their mechanism, viral pretreatment with nanomicelles showed no inhibitory effect. However, pre-exposure of Calu-3 cells led to significant viral replication reduction (>85% and >75% for 1:2 and 1:4 dilutions, respectively), as confirmed by transmission electron microscopy. These findings highlight Pluronic F127-based nanomicelles as a promising nanotechnology-driven strategy against SARS-CoV-2, reinforcing their potential for future antiviral therapies. Full article
(This article belongs to the Special Issue Nanovaccines against Viral Infection)
Show Figures

Figure 1

10 pages, 1418 KiB  
Communication
Cellular Receptor Tyrosine Kinase Signaling Plays Important Roles in SARS-CoV-2 Infection
by Shania Sanchez, Brigitte H. Flannery, Hannah Murphy, Qinfeng Huang, Hinh Ly and Yuying Liang
Pathogens 2025, 14(4), 333; https://doi.org/10.3390/pathogens14040333 - 31 Mar 2025
Viewed by 327
Abstract
Current antiviral treatments often target specific viral components, which can lead to the rapid emergence of drug-resistant mutants. Targeting host signaling pathways, including their associated cellular factors, that are important for virus replication is a novel approach toward the development of next-generation antivirals [...] Read more.
Current antiviral treatments often target specific viral components, which can lead to the rapid emergence of drug-resistant mutants. Targeting host signaling pathways, including their associated cellular factors, that are important for virus replication is a novel approach toward the development of next-generation antivirals to overcome drug resistance. Various cellular receptor tyrosine kinases (RTKs) have previously been shown to play important roles in mediating viral replication including coronaviruses. In this study, we examined the roles of RTKs in SARS-CoV-2 replication in two cell lines, A549-ACE2 (human lung epithelial cells) and Vero-E6 (African Green Monkey kidney cell), via chemical inhibitors. We showed that the HER2 inhibitor Lapatinib significantly reduced viral replication in both cell lines, the TrkA inhibitor GW441756 was effective only in A549-ACE2 cells, while the EGFR inhibitor Gefitinib had little effect in either cell line. Lapatinib and GW441756 exhibited a high therapeutic index (CC50/EC50 > 10) in A549-ACE2 cells. Time-of-addition experiments indicated that Lapatinib may inhibit the early entry step, whereas GW441756 can affect post-entry steps of the viral life cycle. These findings suggest the important roles of HER2 and TrkA signaling in SARS-CoV-2 infection in human lung epithelial cells and support further investigation of RTK inhibitors as potential COVID-19 treatments. Full article
(This article belongs to the Section Viral Pathogens)
Show Figures

Figure 1

17 pages, 9210 KiB  
Article
Repurposing Vancomycin as a Potential Antiviral Agent Against PEDV via nsp13 Helicase Inhibition
by Qiao Chen, Mengqi Yu, Jiajing Guo, Jingqi Qiu, Fei Liu and Yanke Shan
Animals 2025, 15(7), 923; https://doi.org/10.3390/ani15070923 - 23 Mar 2025
Viewed by 185
Abstract
Porcine epidemic diarrhea virus (PEDV) causes a highly contagious intestinal disease with severe economic impacts on the global swine industry. The non-structural protein 13 (nsp13), a viral helicase, is essential for viral replication, making it a promising target for antiviral drug development. In [...] Read more.
Porcine epidemic diarrhea virus (PEDV) causes a highly contagious intestinal disease with severe economic impacts on the global swine industry. The non-structural protein 13 (nsp13), a viral helicase, is essential for viral replication, making it a promising target for antiviral drug development. In this study, through virtual screening and molecular dynamics simulations, Vancomycin, a small-molecule drug also clinically used as an antibacterial agent, was identified to exhibit a stable binding affinity for PEDV nsp13. The NTPase and ATP-dependent RNA helicase activities of PEDV nsp13 were confirmed in vitro, and the optimal biochemical reaction conditions for its dsRNA unwinding activity were established. Further experiments demonstrated that Vancomycin effectively inhibited the dual enzymatic activities of PEDV nsp13 and reduced PEDV infections in vitro. This research highlights Vancomycin as a novel inhibitor of PEDV nsp13, providing valuable mechanistic insights and serving as a model for antiviral drug discovery. While this study suggests its potential for repurposing as a therapeutic agent against PEDV, further investigations are required to evaluate its feasibility in vivo, particularly in terms of safety, efficacy, and practical applicability. Full article
(This article belongs to the Section Veterinary Clinical Studies)
Show Figures

Figure 1

22 pages, 6879 KiB  
Article
Glycyl-tRNA Synthetase as a Target for Antiviral Drug Screening Against Influenza Virus
by Jingjing Zhang, Xiaorong Li, Jingxian Liang, Xinru Meng, Chenchen Zhu, Guangpu Yang, Yali Liang, Qikai Zhou, Qianni Qin, Zan Li, Ting Zhang, Gen Liu and Litao Sun
Int. J. Mol. Sci. 2025, 26(7), 2912; https://doi.org/10.3390/ijms26072912 - 23 Mar 2025
Viewed by 290
Abstract
Influenza viruses are characterized by their high variability and pathogenicity, and effective therapeutic options remain limited. Given these challenges, targeting host cell proteins that facilitate viral replication presents a promising strategy for antiviral drug discovery. In the present study, we observed a significant [...] Read more.
Influenza viruses are characterized by their high variability and pathogenicity, and effective therapeutic options remain limited. Given these challenges, targeting host cell proteins that facilitate viral replication presents a promising strategy for antiviral drug discovery. In the present study, we observed a significant upregulation of Glycyl-tRNA synthetase (GlyRS) within 24 h post-PR8 virus infection. The inhibition of GlyRS expression in A549 cells resulted in a marked reduction in infection rates across multiple influenza virus strains, while the overexpression of GlyRS led to an increase in viral infectivity during the early stages of infection. These findings suggest that GlyRS plays a critical role in the replication of influenza virus. Accordingly, we screened for potential inhibitors targeting GlyRS and identified Lycobetaine and Scutellarein using a multifaceted approach. Through a combination of molecular dynamics simulations, we further elucidated the mechanisms of action and potential binding sites of these compounds. Both inhibitors effectively suppressed the replication of influenza viruses, and their antiviral activity was confirmed to be mediated by GlyRS targeting. Therefore, GlyRS inhibitors, such as Lycobetaine and Scutellarein, represent promising candidates for combating influenza infections and provide novel insights into the treatment of influenza and aaRS-related diseases, opening new avenues for the development of aaRS-targeted therapeutics. Full article
(This article belongs to the Special Issue Antimicrobial Agents: Natural Products or Synthetic Compounds)
Show Figures

Figure 1

25 pages, 2091 KiB  
Review
In Vivo Evidence on the Emerging Potential of Non-Digestible Oligosaccharides as Therapeutic Agents in Bacterial and Viral Infections
by Amirmohammad Afsharnia, Yang Cai, Arjen Nauta, Andre Groeneveld, Gert Folkerts, Marc M. S. M. Wösten and Saskia Braber
Nutrients 2025, 17(6), 1068; https://doi.org/10.3390/nu17061068 - 19 Mar 2025
Viewed by 314
Abstract
The issue of antibiotic-resistant bacterial infections, coupled with the rise in viral pandemics and the slow development of new antibacterial and antiviral treatments, underscores the critical need for novel strategies to mitigate the spread of drug-resistant pathogens, enhance the efficacy of existing therapies, [...] Read more.
The issue of antibiotic-resistant bacterial infections, coupled with the rise in viral pandemics and the slow development of new antibacterial and antiviral treatments, underscores the critical need for novel strategies to mitigate the spread of drug-resistant pathogens, enhance the efficacy of existing therapies, and accelerate the discovery and deployment of innovative antimicrobial and antiviral solutions. One promising approach to address these challenges is the dietary supplementation of non-digestible oligosaccharides (NDOs). NDOs, including human milk oligosaccharides (HMOs), play a vital role in shaping and sustaining a healthy gut microbiota. Beyond stimulating the growth and activity of beneficial gut bacteria, NDOs can also interact directly with pathogenic bacteria and viruses. Their antiviral and antibacterial properties arise from their unique interactions with pathogens and their ability to modulate the host’s immune system. NDOs can function as decoy receptors, inhibit pathogen growth, bind to bacterial toxins, stimulate the host immune response, exhibit anti-biofilm properties, and enhance barrier protection. However, a notable gap exists in the comprehensive assessment of in vivo and clinical data on this topic. This review aims to provide an in-depth overview of the in vivo evidence related to the antiviral and antibacterial effects of various NDOs and HMOs, with a focus on discussing their possible mechanisms of action. Full article
(This article belongs to the Section Carbohydrates)
Show Figures

Figure 1

65 pages, 25172 KiB  
Review
Diterpenoids of Marine Organisms: Isolation, Structures, and Bioactivities
by Qi Shi, Shujie Yu, Manjia Zhou, Peilu Wang, Wenlong Li, Xin Jin, Yiting Pan, Yunjie Sheng, Huaqiang Li, Luping Qin and Xiongyu Meng
Mar. Drugs 2025, 23(3), 131; https://doi.org/10.3390/md23030131 - 18 Mar 2025
Viewed by 382
Abstract
Diterpenoids from marine-derived organisms represent a prolific source of secondary metabolites, characterized by their exceptionally promising chemical structures and pronounced pharmacological properties. In recent years, marine diterpenoids have garnered considerable attention and are regarded as a prominent area of scientific research. As a [...] Read more.
Diterpenoids from marine-derived organisms represent a prolific source of secondary metabolites, characterized by their exceptionally promising chemical structures and pronounced pharmacological properties. In recent years, marine diterpenoids have garnered considerable attention and are regarded as a prominent area of scientific research. As a vital class of metabolites, diterpenoids show diverse biological activities, encompassing antibacterial, antifungal, antiviral, anti-inflammatory, inhibitory, and cytotoxic activities, among others. With the rapid advancement of equipment and identified technology, there has been a tremendous surge in the discovery rate of novel diterpenoid skeletons and bioactivities derived from marine fungi over the past decade. The present review compiles the reported diterpenoids from marine fungal sources mainly generated from January 2000 to December 2024. In this paper, 515 diterpenoids from marine organisms are summarized. Among them, a total of 281 structures from various fungal species are included, comprising 55 from sediment, 39 from marine animals (predominantly invertebrates, including 17 from coral and 22 from sponges), and 53 from marine plants (including 34 from algae and 19 from mangrove). Diverse biological activities are exhibited in 244 compounds, and among these, 112 compounds showed great anti-tumor activity (45.90%) and 110 metabolites showed remarkable cytotoxicity (45.08%). Furthermore, these compounds displayed a range of diverse bioactivities, including potent anti-oxidant activity (2.87%), promising anti-inflammatory activity (1.64%), great anti-bacterial activity (1.64%), notable anti-thrombotic activity (1.23%), etc. Moreover, the diterpenoids’ structural characterization and biological activities are additionally elaborated upon. The present critical summary provides a comprehensive overview of the reported knowledge regarding diterpenoids derived from marine fungi, invertebrates, and aquatic plants. The systematic review presented herein offers medical researchers an extensive range of promising lead compounds for the development of marine drugs, thereby furnishing novel and valuable pharmaceutical agents. Full article
(This article belongs to the Special Issue Bioactive Secondary Metabolites of Marine Fungi, 3rd Edition)
Show Figures

Figure 1

28 pages, 4193 KiB  
Review
4(3H)-Quinazolinone: A Natural Scaffold for Drug and Agrochemical Discovery
by Ke Chen, Shumin Wang, Shuyue Fu, Junehyun Kim, Phumbum Park, Rui Liu and Kang Lei
Int. J. Mol. Sci. 2025, 26(6), 2473; https://doi.org/10.3390/ijms26062473 - 10 Mar 2025
Viewed by 298
Abstract
4(3H)-quinazolinone is a functional scaffold that exists widely both in natural products and synthetic organic compounds. Its drug-like derivatives have been extensively synthesized with interesting biological features including anticancer, anti-inflammatory, antiviral, antimalarial, antibacterial, antifungal, and herbicidal, etc. In this review, we [...] Read more.
4(3H)-quinazolinone is a functional scaffold that exists widely both in natural products and synthetic organic compounds. Its drug-like derivatives have been extensively synthesized with interesting biological features including anticancer, anti-inflammatory, antiviral, antimalarial, antibacterial, antifungal, and herbicidal, etc. In this review, we highlight the medicinal and agrochemical versatility of the 4(3H)-quinazolinone scaffold according to the studies published in the past six years (2019–2024), and comprehensively give a summary of the target recognition, structure–activity relationship, and mechanism of its analogs. The present review is expected to provide valuable guidance for discovering novel lead compounds containing 4(3H)-quinazolinone moiety in both drug and agrochemical research. Full article
Show Figures

Figure 1

13 pages, 4558 KiB  
Article
PABPC4 Inhibits SADS-CoV Replication by Degrading the Nucleocapsid Protein Through Selective Autophagy
by Chenchen Zhao, Yan Qin, Haixin Huang, Wei Chen, Yanqing Hu, Xinyu Zhang, Yuying Li, Tian Lan and Wenchao Sun
Vet. Sci. 2025, 12(3), 257; https://doi.org/10.3390/vetsci12030257 - 10 Mar 2025
Viewed by 361
Abstract
Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a novel enteric coronavirus that causes severe clinical diarrhea and intestinal pathological injury in pigs. Selective autophagy is an important mechanism of host defense against virus invasion. However, the mechanism through which SADS-CoV-mediated selective autophagy mediates [...] Read more.
Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a novel enteric coronavirus that causes severe clinical diarrhea and intestinal pathological injury in pigs. Selective autophagy is an important mechanism of host defense against virus invasion. However, the mechanism through which SADS-CoV-mediated selective autophagy mediates the innate immune response remains unknown. Here, we report that the host protein PABPC4 can inhibit SADS-CoV replication through targeting and degrading its N protein. Furthermore, we demonstrate that PABPC4 recruits MARCHF8 (an E3 ubiquitin ligase), which ubiquitinates the N protein and is degraded via NDP52/CALCOCO2 (a selective autophagy cargo receptor). Taken together, these findings reveal a new mechanism by which PABPC4 inhibits virus replication, and reveal a new target for antiviral drug development. Full article
(This article belongs to the Special Issue Viral Infections in Wild and Domestic Animals)
Show Figures

Figure 1

18 pages, 9382 KiB  
Article
A Novel In Vitro Primary Human Alveolar Model (AlveolAir™) for H1N1 and SARS-CoV-2 Infection and Antiviral Screening
by Cindia Ferreira Lopes, Emilie Laurent, Mireille Caul-Futy, Julia Dubois, Chloé Mialon, Caroline Chojnacki, Edouard Sage, Bernadett Boda, Song Huang, Manuel Rosa-Calatrava and Samuel Constant
Microorganisms 2025, 13(3), 572; https://doi.org/10.3390/microorganisms13030572 - 3 Mar 2025
Viewed by 750
Abstract
Lower respiratory infections, mostly caused by viral or bacterial pathogens, remain a leading global cause of mortality. The differences between animal models and humans contribute to inefficiencies in drug development, highlighting the need for more relevant and predictive, non-animal models. In this context, [...] Read more.
Lower respiratory infections, mostly caused by viral or bacterial pathogens, remain a leading global cause of mortality. The differences between animal models and humans contribute to inefficiencies in drug development, highlighting the need for more relevant and predictive, non-animal models. In this context, AlveolAir™, a fully primary in vitro 3D human alveolar model, was characterized and demonstrated the sustained presence of alveolar type I (ATI) and type II (ATII) cells. This model exhibited a functional barrier over a 30-day period, evidenced by high transepithelial electrical resistance (TEER). These findings were further validated by tight junctions’ confocal microscopy and low permeability to Lucifer yellow, confirming AlveolAir™ as robust platform for drug transport assays. Additionally, successful infections with H1N1 and SARS-CoV-2 viruses were achieved, and antiviral treatments with Baloxavir and Remdesivir, respectively, effectively reduced viral replication. Interestingly, both viruses infected only the epithelial layer without replicating in endothelial cells. These findings indicate AlveolAir™ as a relevant model for assessing the toxicity and permeability of xenobiotics and evaluating the efficacy of novel antiviral therapies. Full article
Show Figures

Figure 1

22 pages, 3780 KiB  
Article
Discovery of Arylfuran and Carbohydrate Derivatives from the BraCoLi Library as Potential Zika Virus NS3pro Inhibitors
by Fernanda Kelly Marcelino e Oliveira, Beatriz Murta Rezende Moraes Ribeiro, Ellen Gonçalves de Oliveira, Marina Mol Sena Andrade Verzola, Thales Kronenberger, Vinícius Gonçalves Maltarollo, Ricardo José Alves, Renata Barbosa de Oliveira, Rafaela Salgado Ferreira, Jônatas Santos Abrahão and Mateus Sá Magalhães Serafim
Future Pharmacol. 2025, 5(1), 9; https://doi.org/10.3390/futurepharmacol5010009 - 15 Feb 2025
Viewed by 369
Abstract
Background/Objectives: Zika fever is a disease caused by the Zika virus (ZIKV). Symptomatic cases may be associated with neurological disorders in adults, as well as congenital Zika syndrome and other birth defects during pregnancy. In 2016, Zika fever was considered a public health [...] Read more.
Background/Objectives: Zika fever is a disease caused by the Zika virus (ZIKV). Symptomatic cases may be associated with neurological disorders in adults, as well as congenital Zika syndrome and other birth defects during pregnancy. In 2016, Zika fever was considered a public health problem by the World Health Organization (WHO), highlighting the need to develop new therapies against the disease. Currently, there is no antiviral or vaccine available to treat or prevent severe cases. Due to the lack of available therapeutics and few promising hit molecules, we computationally screened the well-described ZIKV protease (NS3pro) as a drug target to revisit the small-molecule database Brazilian Compound Library (BraCoLi) and select potential inhibitors. Methods: We employed a consensus docking screening of a library of 1176 compounds using GOLD and DockThor. We selected 28 hits based on predicted binding affinity, and only the remnants of three compounds were available in the library at the time of this study for experimental validation. The hits were evaluated for their cytotoxic (CC50) and effective concentrations (EC50) for their potential antiviral activity in Vero cells. Results: The three hit compounds presented modest CC50 values of 89.15 ± 3.72, >100, and 29.67 ± 1.01 μM, with the latter, a carbohydrate derivative, having an EC50 value of >12.5 μM (~40% inhibition) against ZIKV PE243. Additionally, the essentially non-toxic compound, an arylfuran derivative, also inhibited the ZIKV NS3pro with an IC50 value of 17 μM but presented evidence of acting through a promiscuous mechanism for enzyme inhibition. Conclusion: This study highlights the relevance of revisiting existing small-molecule assets to identify novel therapeutic starting points against ZIKV, aiming for potential lead candidates in the future. Full article
Show Figures

Graphical abstract

18 pages, 3309 KiB  
Review
Inhaled Dry Powder of Antiviral Agents: A Promising Approach to Treating Respiratory Viral Pathogens
by Tushar Saha, Zia Uddin Masum, Anik Biswas, Moushumi Afroza Mou, Sohag Ahmed and Tamal Saha
Viruses 2025, 17(2), 252; https://doi.org/10.3390/v17020252 - 12 Feb 2025
Viewed by 643
Abstract
Inhaled dry powder formulations of antiviral agents represent a novel and potentially transformative approach to managing respiratory viral infections. Traditional antiviral therapies in the form of tablets or capsules often face limitations in terms of therapeutic activity, systemic side effects, and delayed onset [...] Read more.
Inhaled dry powder formulations of antiviral agents represent a novel and potentially transformative approach to managing respiratory viral infections. Traditional antiviral therapies in the form of tablets or capsules often face limitations in terms of therapeutic activity, systemic side effects, and delayed onset of action. Dry powder inhalers (DPIs) provide a targeted delivery system, ensuring the direct administration of antivirals to the infection site, the respiratory tract, which potentially enhance therapeutic efficacy and minimize systemic exposure. This review explores the current state of inhaled dry powder antiviral agents, their advantages over traditional routes, and specific formulations under development. We discuss the benefits of targeted delivery, such as improved drug deposition in the lungs and reduced side effects, alongside considerations related to the formulation preparation. In addition, we summarize the developed (published and marketed) inhaled dry powders of antiviral agents. Full article
(This article belongs to the Special Issue Pharmacology of Antiviral Drugs)
Show Figures

Figure 1

23 pages, 3549 KiB  
Article
Efavirenz Repurposing Challenges: A Novel Nanomicelle-Based Antiviral Therapy Against Mosquito-Borne Flaviviruses
by Sofía Maldonado, Pedro Fuentes, Ezequiel Bernabeu, Facundo Bertera, Javier Opezzo, Eduardo Lagomarsino, Hyun J. Lee, Fleming Martínez Rodríguez, Marcelo R. Choi, María Jimena Salgueiro, Elsa B. Damonte, Christian Höcht, Marcela A. Moretton, Claudia S. Sepúlveda and Diego A. Chiappetta
Pharmaceutics 2025, 17(2), 241; https://doi.org/10.3390/pharmaceutics17020241 - 12 Feb 2025
Viewed by 668
Abstract
Background/Objective: World Health Organization latest statistics state that 17% of infectious diseases are transmitted by vectors, causing more than 700,000 deaths each year. Particularly, dengue (DENV), Zika (ZIKV) and yellow fever (YFV) viral infections have generated international awareness due to their epidemic proportion [...] Read more.
Background/Objective: World Health Organization latest statistics state that 17% of infectious diseases are transmitted by vectors, causing more than 700,000 deaths each year. Particularly, dengue (DENV), Zika (ZIKV) and yellow fever (YFV) viral infections have generated international awareness due to their epidemic proportion and risks of international spread. In this framework, the repositioning strategy of Efavirenz (EFV) represents a key clinical feature to improve different antiviral therapies. Therefore, the development of Soluplus®-based nanomicelles (NMs) loaded with EFV (10 mg/mL) for optimized oral pharmacotherapy against ZIKV, DENV and YFV infections was investigated. Methods: EFV-NMs were obtained by an acetone diffusion technique. Micellar size and in vitro micellar interaction with mucin were assessed by dynamic light scattering. In vitro cytocompatibility was investigated in A549 and Vero cells and micellar in vitro antiviral activity against ZIKV, DENV and YFV was evaluated. In vivo oral bioavailability and histological studies were assessed in Wistar rats. Results: EFV encapsulation within Soluplus® NMs increased the drug’s apparent aqueous solubility up to 4803-fold with a unimodal micellar size distribution and a micellar size of ~90 nm at 25 and 37 °C. Micellar in vitro interaction with mucin was also assessed in a pH range of 1.2–7.5 and its storage micellar physicochemical stability at 4 °C was confirmed over 2 years. In vitro cytocompatibility assays in A549 and Vero cells confirmed that EFV micellar dispersions resulted in safe nanoformulations. Interestingly, EFV-loaded NMs exhibited significantly higher in vitro antiviral activity compared with EFV solution for all the tested flaviviruses. In addition, the selectivity index (SI) values reveal that EFV-loaded NMs exhibited considerably more biological efficacy compared to EFV solution in A549 and Vero cell lines and for each viral infection (SI > 10). Further, the drug pharmacokinetics parameters were enhanced after the oral administration of EFV-loaded NMs, being biocompatible by not causing damage in the gastrointestinal segments. Conclusions: Overall, our EFV nanoformulation highlighted its potential as a novel drug delivery platform for optimized ZIKV, DENV and YFV antiviral therapy. Full article
Show Figures

Graphical abstract

10 pages, 3228 KiB  
Article
Early Intervention in Herpes Simplex Virus-1 Replication in Vitro with Allenic Macrolide Archangiumide
by You Li, Jia-Qi Hu, Wen-Hai Feng, Changsheng Wu and Li Gao
Int. J. Mol. Sci. 2025, 26(4), 1537; https://doi.org/10.3390/ijms26041537 - 12 Feb 2025
Viewed by 497
Abstract
Archangiumide is a unique macrolide natural product that features an endocyclic allene functionality, rendering it a prototype of a new class of secondary metabolites of microbial origin. However, its biological and/or pharmaceutical roles remain obscure. In this study, we have unveiled an antiviral [...] Read more.
Archangiumide is a unique macrolide natural product that features an endocyclic allene functionality, rendering it a prototype of a new class of secondary metabolites of microbial origin. However, its biological and/or pharmaceutical roles remain obscure. In this study, we have unveiled an antiviral potency of archangiumide that was effective against herpes simplex virus (HSV-1) replication. We found that archangiumide did not affect host cell viability, nor pathogen infectivity, but suppressed HSV-1 early replication, in terms of early replication genes, such as ICP0, ICP4, etc. Further scrutinizing the underlined master regulator, we found that HSV-1 VP16 protein expression was inhibited by archangiumide, as well as VP16 nuclear translocation. As VP16 is a coactivator of transcription, archangiumide harnessed the master regulator of HSV-1 early replication. Together, here we provide evidence that allene macrolide archangiumide possesses robust antiviral functions that may be valuable for a novel viral infection intervention, as macrolides are generally safe drugs for prolonged treatments. Full article
Show Figures

Figure 1

22 pages, 13927 KiB  
Article
Discovery of TRPV4-Targeting Small Molecules with Anti-Influenza Effects Through Machine Learning and Experimental Validation
by Yan Sun, Jiajing Wu, Beilei Shen, Hengzheng Yang, Huizi Cui, Weiwei Han, Rongbo Luo, Shijun Zhang, He Li, Bingshuo Qian, Lingjun Fan, Junkui Zhang, Tiecheng Wang, Xianzhu Xia, Fang Yan and Yuwei Gao
Int. J. Mol. Sci. 2025, 26(3), 1381; https://doi.org/10.3390/ijms26031381 - 6 Feb 2025
Viewed by 825
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a calcium-permeable cation channel critical for maintaining intracellular Ca2+ homeostasis and is essential in regulating immune responses, metabolic processes, and signal transduction. Recent studies have shown that TRPV4 activation enhances influenza A virus infection, promoting [...] Read more.
Transient receptor potential vanilloid 4 (TRPV4) is a calcium-permeable cation channel critical for maintaining intracellular Ca2+ homeostasis and is essential in regulating immune responses, metabolic processes, and signal transduction. Recent studies have shown that TRPV4 activation enhances influenza A virus infection, promoting viral replication and transmission. However, there has been limited exploration of antiviral drugs targeting the TRPV4 channel. In this study, we developed the first machine learning model specifically designed to predict TRPV4 inhibitory small molecules, providing a novel approach for rapidly identifying repurposed drugs with potential antiviral effects. Our approach integrated machine learning, virtual screening, data analysis, and experimental validation to efficiently screen and evaluate candidate molecules. For high-throughput virtual screening, we employed computational methods to screen open-source molecular databases targeting the TRPV4 receptor protein. The virtual screening results were ranked based on predicted scores from our optimized model and binding energy, allowing us to prioritize potential inhibitors. Fifteen small-molecule drugs were selected for further in vitro and in vivo antiviral testing against influenza. Notably, glecaprevir and everolimus demonstrated significant inhibitory effects on the influenza virus, markedly improving survival rates in influenza-infected mice (protection rates of 80% and 100%, respectively). We also validated the mechanisms by which these drugs interact with the TRPV4 channel. In summary, our study presents the first predictive model for identifying TRPV4 inhibitors, underscoring TRPV4 inhibition as a promising strategy for antiviral drug development against influenza. This pioneering approach lays the groundwork for future clinical research targeting the TRPV4 channel in antiviral therapies. Full article
Show Figures

Graphical abstract

Back to TopTop