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
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
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
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (3,990)

Search Parameters:
Keywords = structured drug review

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 2752 KB  
Review
Capric Acid-Based Therapeutic Deep Eutectic Systems: A Focused Review Within the Framework of Deep Eutectic Solvents
by Faisal Al-Akayleh, Ahmed S. A. Ali Agha, Ali R. Olaimat and Giuseppe Biagini
Pharmaceuticals 2026, 19(1), 159; https://doi.org/10.3390/ph19010159 - 15 Jan 2026
Abstract
Background/Objectives: Capric acid (CA)–therapeutic deep eutectic systems (THEDES) are emerging as a distinct class of biofunctional matrices capable of reshaping drug solubilization, permeability, and bioactivity. Methods: Relevant studies on CA–THEDES were identified through targeted database searches and screened for evidence on [...] Read more.
Background/Objectives: Capric acid (CA)–therapeutic deep eutectic systems (THEDES) are emerging as a distinct class of biofunctional matrices capable of reshaping drug solubilization, permeability, and bioactivity. Methods: Relevant studies on CA–THEDES were identified through targeted database searches and screened for evidence on their design, mechanisms, and pharmaceutical performance. Results: This review synthesizes current evidence on their structural design, mechanistic behavior, and pharmaceutical performance, revealing several unifying principles. Across multiple drug classes, CA consistently drives strong, directional hydrogen bonding and drug amorphization, resulting in marked solubility enhancement and stabilization of non-crystalline or supersaturated states relative to crystalline drugs or conventional solvent systems. Its amphiphilic C10 chain further contributes to membrane fluidization, which explains the improved transdermal and transmucosal permeation repeatedly observed in CA-THEDES. Additionally, synergistic antimicrobial and anticancer effects reported in several systems confirm that CA acts not only as a solvent component but as a bioactive co-therapeutic. Collectively, the reviewed data show that CA serves as a structurally determinant element whose dual hydrogen-bonding and membrane-interacting roles underpin the high pharmaceutical performance of these systems. However, gaps remain in long-term stability, toxicological profiling, and regulatory classification. Emerging Artificial Intelligence (AI) and Machine Learning (ML)-guided predictive approaches offer promising solutions by enabling rational selection of eutectic partners, optimal ratios, and property optimization through computational screening. Conclusions: Overall, CA-THEDES represent a rationally designable platform for next-generation drug delivery, where solvent functionality and therapeutic activity converge within a single, green formulation system. Full article
(This article belongs to the Section Pharmaceutical Technology)
Show Figures

Figure 1

14 pages, 891 KB  
Review
Why Cemiplimab? Defining a Unique Therapeutic Niche in First-Line Non-Small-Cell Lung Cancer with Ultra-High PD-L1 Expression and Squamous Histology
by Satoshi Ikeda, Keigo Araki, Mai Kitagawa, Natsuno Makihara, Yutaro Nagata, Kazuki Fujii, Kiyori Yoshida, Tatsuki Ikoma, Kahori Nakahama, Yuki Takeyasu, Utae Katsushima, Yuta Yamanaka and Takayasu Kurata
Cancers 2026, 18(2), 272; https://doi.org/10.3390/cancers18020272 - 15 Jan 2026
Abstract
The landscape of first-line treatment for metastatic non-small cell lung cancer (NSCLC) without actionable driver mutations is rapidly evolving, currently dominated by pembrolizumab-based regimens. This review discusses the unique molecular characteristics of cemiplimab, a newer anti-PD-1 antibody, and defines its optimal positioning against [...] Read more.
The landscape of first-line treatment for metastatic non-small cell lung cancer (NSCLC) without actionable driver mutations is rapidly evolving, currently dominated by pembrolizumab-based regimens. This review discusses the unique molecular characteristics of cemiplimab, a newer anti-PD-1 antibody, and defines its optimal positioning against established standards. Cemiplimab is a fully human IgG4 monoclonal antibody distinguished by two key features: an engineered hinge-region mutation that prevents Fab-arm exchange, ensuring exceptional molecular stability which minimizes anti-drug antibody (ADA) risks associated with unstable molecules; and a unique interaction with PD-1 glycosylation sites, potentially enhancing binding efficacy. These structural advantages may be particularly relevant in histologies like squamous NSCLC, where accumulating somatic mutations drive high neoantigen loads and heightened immune responses, creating an environment historically prone to ADA formation. Based on data from the pivotal EMPOWER-Lung program, we highlight cemiplimab’s exceptional promise in specific populations. Firstly, in the EMPOWER-Lung 1 trial, cemiplimab monotherapy demonstrated extraordinary survival benefits in a pre-specified analysis of the distinct “ultra-high” PD-L1 expression subgroup (TPS ≥90%), potentially surpassing historical benchmarks. Secondly, cemiplimab displays consistent, robust efficacy in challenging-to-treat squamous histology, both as monotherapy for patients with high PD-L1 expression and in combination with chemotherapy for patients with PD-L1 < 50%. In conclusion, cemiplimab establishes a unique therapeutic niche for patients with squamous histology and ultra-high PD-L1 expression, likely driven by its distinct structural stability and reduced immunogenicity. Full article
(This article belongs to the Special Issue Oncology: State-of-the-Art Research and Initiatives in Japan)
Show Figures

Figure 1

35 pages, 1471 KB  
Review
β-Alanine Is an Unexploited Neurotransmitter in the Pathogenesis and Treatment of Alzheimer’s Disease
by Cindy M. Wozniczka and Donald F. Weaver
NeuroSci 2026, 7(1), 13; https://doi.org/10.3390/neurosci7010013 - 15 Jan 2026
Abstract
Alzheimer’s disease (AD) remains an unmet medical challenge, as there are no effective therapies that alter the disease’s progression. While approaches have targeted molecules like acetylcholine (ACh) and glutamate, these strategies have provided only limited benefits and do not address the complex molecular [...] Read more.
Alzheimer’s disease (AD) remains an unmet medical challenge, as there are no effective therapies that alter the disease’s progression. While approaches have targeted molecules like acetylcholine (ACh) and glutamate, these strategies have provided only limited benefits and do not address the complex molecular mechanisms underlying AD development. This review suggests that β-alanine (3-aminopropanoic acid) is an underexplored neurotransmitter that could serve as a potential AD drug target. Existing evidence indicates that β-alanine modulates GABAergic and glutamatergic neurotransmission, thereby affecting neuronal hyperexcitability. Additionally, studies suggest that β-alanine has antioxidant effects, reducing oxidative stress caused by reactive oxygen species (ROS). We propose that β-alanine might bind to Aβ/tau proteins, possibly targeting the six-amino acid sequences EVHHQK/DDKKAK, which are involved in protein aggregation. β-Alanine may also influence the release of pro-inflammatory cytokines from microglia, potentially reducing neuroinflammation. We also hypothesize that β-alanine may help regulate metal dyshomeostasis, which leads to ROS production. Taurine, structurally like β-alanine, appears to influence comparable mechanisms. Although structural similarity doesn’t ensure therapeutic effectiveness, this evidence supports considering β-alanine as a treatment for AD. Furthermore, β-alanine and its analogues face challenges, including crossing the blood–brain barrier (BBB) and optimizing structure–activity relationships (SAR). This review includes articles through September 2025, sourced from four databases. Full article
Show Figures

Figure 1

24 pages, 3309 KB  
Review
Biomaterials for Improving Skin Penetration in Treatment of Skin Cancer
by Davide Secci, Andrew Urquhart, Vasileios Bekiaris and Katrine Qvortrup
J. Funct. Biomater. 2026, 17(1), 39; https://doi.org/10.3390/jfb17010039 - 15 Jan 2026
Abstract
Skin cancer is among the most common malignancies worldwide, posing a significant societal burden due to its increasing incidence and its limited responsiveness to conventional topical therapies. Treatment is challenged by the presence of the skin barrier which restricts drug penetration. This review [...] Read more.
Skin cancer is among the most common malignancies worldwide, posing a significant societal burden due to its increasing incidence and its limited responsiveness to conventional topical therapies. Treatment is challenged by the presence of the skin barrier which restricts drug penetration. This review discusses the structural and physiological challenges of topical delivery and summarizes efforts to develop functional biomaterials to enhance skin drug penetration. Important aspects to consider when developing formulation strategies, such as drug properties and release mechanisms, are discussed alongside current limitations and future perspectives. Full article
(This article belongs to the Special Issue Review Papers in Biomaterials for Cancer Therapies)
Show Figures

Figure 1

16 pages, 862 KB  
Review
Drug-Induced Acute Generalized Exanthematous Pustulosis: Mechanisms, Diagnosis, and Clinical Differentiation from Other Pustular Eruptions
by Esteban Zavaleta-Monestel, Audry Escudero-Correa, Jeaustin Mora-Jiménez, Andy Jesús Hernández-Vásquez, Luis Carlos Monge-Bogantes, Josephine Hernández-López and Sebastián Arguedas-Chacón
Dermato 2026, 6(1), 3; https://doi.org/10.3390/dermato6010003 - 15 Jan 2026
Abstract
Background/Objectives: Acute generalized exanthematous pustulosis (AGEP) is a severe drug-induced cutaneous reaction characterized by the abrupt onset of sterile pustules, fever, neutrophilia, and a T cell-mediated type IVd hypersensitivity response. This narrative review synthesizes current evidence on pharmacological triggers, immunopathogenic mechanisms, diagnostic criteria, [...] Read more.
Background/Objectives: Acute generalized exanthematous pustulosis (AGEP) is a severe drug-induced cutaneous reaction characterized by the abrupt onset of sterile pustules, fever, neutrophilia, and a T cell-mediated type IVd hypersensitivity response. This narrative review synthesizes current evidence on pharmacological triggers, immunopathogenic mechanisms, diagnostic criteria, and differential diagnosis to provide a clinically oriented framework. Methods: A comprehensive literature search was conducted in PubMed/MEDLINE, Scopus, ScienceDirect, and SpringerLink for studies published between 2000 and 2025, complemented by selected clinical reference sources. Studies addressing clinical features, immunological pathways, pharmacovigilance signals, and diagnostic tools for AGEP were included. Synthesis of Evidence: β-lactam antibiotics remain the most frequent triggers, while increasing associations have been reported with hydroxychloroquine, targeted therapies, immune checkpoint inhibitors, psychotropic agents, and vaccines. Immunopathogenesis is driven by IL-36 activation, CXCL8/IL-8–mediated neutrophil recruitment, and IL36RN mutations, explaining overlap with pustular psoriasis. Diagnostic accuracy improves through integration of drug latency, clinical morphology, histopathology, biomarkers, and standardized tools such as the EuroSCAR score. Conclusions: AGEP is a complex pustular reaction induced by diverse drugs and amplified by IL-36-mediated inflammation. Accurate diagnosis requires a multidimensional approach supported by structured algorithms and robust pharmacovigilance to identify evolving drug-associated patterns. Full article
(This article belongs to the Special Issue Reviews in Dermatology: Current Advances and Future Directions)
Show Figures

Figure 1

34 pages, 4734 KB  
Review
Targeting Bacterial Cell Wall Synthesis: Structural Insights and Emerging Therapeutic Strategies
by Bharat Kumar Reddy Sanapalli, Christopher R. Jones and Vidyasrilekha Sanapalli
Pharmaceutics 2026, 18(1), 106; https://doi.org/10.3390/pharmaceutics18010106 - 13 Jan 2026
Abstract
The emergence of multidrug-resistant (MDR) bacterial pathogens has heightened the urgency for novel antibacterial agents. The bacterial cell wall usually comprises peptidoglycan, which presents a prime target for antibacterial drug development due to its indispensable role in maintaining cellular integrity. Conventional antibiotics such [...] Read more.
The emergence of multidrug-resistant (MDR) bacterial pathogens has heightened the urgency for novel antibacterial agents. The bacterial cell wall usually comprises peptidoglycan, which presents a prime target for antibacterial drug development due to its indispensable role in maintaining cellular integrity. Conventional antibiotics such as β-lactams and glycopeptides hinder peptidoglycan synthesis through competitive binding of penicillin-binding proteins (PBPs) and sequestration of lipid-linked precursor molecules. Nevertheless, prevalent resistance mechanisms including target modification, β-lactamase hydrolysis, and multi-drug efflux pumps have limited their clinical utility. This comprehensive analysis explicates the molecular machinery underlying bacterial cell wall assembly, evaluates both explored and unexplored enzymatic nodes within this pathway, and highlights the transformative impact of high-resolution structural elucidation in accelerating structure-guided drug discovery. Novel targets such as GlmS, GlmM, GlmU, Mur ligases, D,L-transpeptidases are assessed for their inclusiveness for the discovery of next-generation antibiotics. Additionally, cell wall inhibitors are also examined for their mechanisms of action and evolutionary constraints on MDR development. High-resolution crystallographic data provide valuable insights into molecular blueprints for structure-guided optimization of pharmacophores, enhancing binding affinity and circumventing resistance determinants. This review proposes a roadmap for future innovation, advocating for the convergence of computational biology platforms, machine learning-driven compound screening, and nanoscale delivery systems to improve therapeutic efficacy and pharmacokinetics. The synergy of structural insights and cutting-edge technologies offers a multidisciplinary framework for revitalizing the antibacterial arsenal and combating MDR infections efficiently. Full article
(This article belongs to the Special Issue New Era in Antimicrobial Strategies)
Show Figures

Figure 1

18 pages, 1758 KB  
Review
Computational Workflow for Chemical Compound Analysis: From Structure Generation to Molecular Docking
by Jesus Magdiel García-Díaz, Asbiel Felipe Garibaldi-Ríos, Martha Patricia Gallegos-Arreola, Filiberto Gutiérrez-Gutiérrez, Jorge Iván Delgado-Saucedo, Moisés Martínez-Velázquez and Ana María Puebla-Pérez
Sci. Pharm. 2026, 94(1), 9; https://doi.org/10.3390/scipharm94010009 - 13 Jan 2026
Viewed by 63
Abstract
Drug discovery is a complex and expensive process in which only a small proportion of candidate molecules reach clinical approval. Computational methods, particularly computer-aided drug design (CADD), have become fundamental to accelerate and optimize early stages of discovery by integrating chemical, biological, and [...] Read more.
Drug discovery is a complex and expensive process in which only a small proportion of candidate molecules reach clinical approval. Computational methods, particularly computer-aided drug design (CADD), have become fundamental to accelerate and optimize early stages of discovery by integrating chemical, biological, and pharmacokinetic information into predictive models. This review outlines a complete computational workflow for chemical compound analysis, covering molecular structure generation, database selection, evaluation of absorption, distribution, metabolism, excretion and toxicity (ADMET), target prediction, and molecular docking. It focuses on freely accessible and web-based tools that enable reproducible, cost-effective, and scalable in silico studies. Key platforms such as PubChem, ChEMBL, RDKit, SwissADME, TargetNet, and SwissDock are highlighted as examples of how different resources can be integrated to support rational compound design and prioritization. The article also discusses essential methodological principles, data curation strategies, and common limitations in virtual screening and docking analyses. Finally, it explores future directions in computational drug discovery, including the incorporation of artificial intelligence, multi-omics integration, and quantum simulations, to enhance predictive accuracy and translational relevance. Full article
(This article belongs to the Topic Bioinformatics in Drug Design and Discovery—2nd Edition)
Show Figures

Figure 1

13 pages, 366 KB  
Review
Mathematical Modeling of Local Drug Delivery in the Oral Cavity: From Release Kinetics to Mini-PBPK and Local PK/PD with Applications to Periodontal Therapies
by Rafał Rakoczy, Monika Machoy-Rakoczy and Izabela Gutowska
Pharmaceutics 2026, 18(1), 101; https://doi.org/10.3390/pharmaceutics18010101 - 12 Jan 2026
Viewed by 72
Abstract
Background/Objectives: Mathematical modelling provides a quantitative way to describe the fate and action of drugs in the oral cavity, where transport processes are shaped by salivary flow, pellicle formation, biofilm structure and the wash-out effect of gingival crevicular fluid (GCF). Local pharmacokinetics in [...] Read more.
Background/Objectives: Mathematical modelling provides a quantitative way to describe the fate and action of drugs in the oral cavity, where transport processes are shaped by salivary flow, pellicle formation, biofilm structure and the wash-out effect of gingival crevicular fluid (GCF). Local pharmacokinetics in the mouth differ substantially from systemic models, and therefore a dedicated framework is required. The aim of this work was to present a structured, physiologically based concept that links in vitro release testing with local pharmacokinetics and pharmacodynamics. Methods: A narrative review with elements of systematic search was conducted in PubMed, Scopus and Web of Science (1980–2025) for publications describing drug release, local PBPK, and PK/PD modelling in the oral cavity. Mathematical formulations were grouped into release kinetics, mini-PBPK transport and local PK/PD relations. Classical models (Higuchi, Korsmeyer–Peppas, Peppas–Sahlin) were integrated with a mini-PBPK structure describing saliva–mucosa–biofilm–pocket interactions. Results: The combined model captures adsorption to pellicle, diffusion within biofilm and wash-out by GCF. It allows simulation of variable clinical conditions, such as inflammation-related changes in QGCF, and links local exposure to pharmacodynamic outcomes. Case studies with PerioChip®, Arestin®, and Atridox® demonstrate how mechanistic models explain observed therapeutic duration and low-systemic exposure. Conclusions: The proposed mini-PBPK framework bridges empirical release data and physiological transport in the oral cavity. It supports rational formulation design, optimisation of local dosage, and personalised prediction of drug retention in gingival pockets. This modelling approach can become a practical tool for the development of dental biomaterials and subgingival therapies. Full article
38 pages, 2430 KB  
Review
Advances in Natural Products from Mangrove-Associated Fungi Along the Indian Ocean Coast
by Parakkrama Wijerathna, Xinqi Chen, Rongxiang Qiu, P.V.J.S. Wijethilake, Yi Chen, Nuwan Madushanka, I.J.J.U.N. Perera, Jian Cai, Lalith Jayasinghe, Yonghong Liu, Vajira P. Bulugahapitiya and Xuefeng Zhou
Molecules 2026, 31(2), 261; https://doi.org/10.3390/molecules31020261 - 12 Jan 2026
Viewed by 85
Abstract
Mangrove ecosystems along the Indian Ocean coast show great biodiversity, adapting to harsh environmental conditions of high salinity and higher organic matter, and they are a host for a range of microbial communities with special features that produce unique secondary metabolites. Of this, [...] Read more.
Mangrove ecosystems along the Indian Ocean coast show great biodiversity, adapting to harsh environmental conditions of high salinity and higher organic matter, and they are a host for a range of microbial communities with special features that produce unique secondary metabolites. Of this, mangrove-associated endophytic fungi, the second largest ecological group of marine fungi, show the greater potential, being a diverse pool for discovering novel bio-actives with pharmacological and biotechnological interest. This review summarizes the research findings on structural diversity and the associated pharmacological activities of secondary metabolites produced by mangrove-associated fungi along the Indian Ocean coast reported over the period of 2002–2025, based on the literature retrieved from Google Scholar. The total of 302 secondary metabolites is presented mainly from classes of polyketides (208), alkaloids (34), and terpenoids (60). Interestingly, 164 compounds were identified, as first reported in those publications. These compounds have been reported to show diverse biological activities, and the most prominent activities are cytotoxic, antibacterial, antifungal, antioxidant, enzyme inhibitory, and anti-inflammatory effects. The structural novelty and pharmacological activities of these metabolites highlight the importance of mangrove fungi as promising sources for new drug discovery and advancing industrial biotechnology. Therefore, this review highlights the insight into the possible application of these chemical compounds in the future drug industry, as well as in biotechnology for advancing human well-being. Furthermore, though significant progress has been made in exploring the fungi community from mangroves of the African and Middle Eastern coasts, the Indian coast mangrove fungi are yet to be explored more for novel discoveries. Full article
Show Figures

Figure 1

21 pages, 5022 KB  
Review
Phosphatidylinositol-3-Kinase (PI3K) and Histone Deacetylase (HDAC) Multitarget Inhibitors: An Update on Clinical and Preclinical Candidates
by Alef D. S. Lima and Lídia M. Lima
Pharmaceuticals 2026, 19(1), 130; https://doi.org/10.3390/ph19010130 - 12 Jan 2026
Viewed by 80
Abstract
Phosphatidylinositol-3-kinases (PI3Ks) constitute an important validated therapeutic class involved in crucial cellular processes, and their dysregulation is associated with cancer initiation and progression. Nonetheless, intrinsic and acquired resistance mechanisms associated with PI3K pathway modulation have underscored the need for alternative therapeutic strategies. In [...] Read more.
Phosphatidylinositol-3-kinases (PI3Ks) constitute an important validated therapeutic class involved in crucial cellular processes, and their dysregulation is associated with cancer initiation and progression. Nonetheless, intrinsic and acquired resistance mechanisms associated with PI3K pathway modulation have underscored the need for alternative therapeutic strategies. In this context, recent studies have shown that simultaneous inhibition of PI3K and histone deacetylases (HDAC) promotes synergistic antitumor effects in different cancer cell lines. HDACs are validated epigenetic targets that are extensively explored in clinical practice and have a pharmacophore with versatility for structural modifications, which facilitates the design of multitarget inhibitors. This review examines the rational design and synthetic evolution of dual PI3K/HDAC inhibitors, an area catalyzed by the development of fimepinostat, the first clinically evaluated agent exhibiting potent and balanced inhibition of both targets. We provide a critical overview of PI3K/HDAC multitarget inhibitors reported in recent years that have progressed to preclinical or clinical investigation, discussing the structural frameworks employed, medicinal chemistry strategies adopted, and structure–activity relationships established. Particular attention is given to advantageous molecular features as well as challenges related to toxicity, pharmacokinetic behavior, and pharmacodynamic modulation. From this comprehensive analysis, we outline key considerations and emerging design principles that may inform the next generation of PI3K/HDAC multitarget drug candidates. Insights derived from the diversity of chemical scaffolds, activity profiles, and selectivity patterns described herein may support the development of innovative therapeutic agents capable of overcoming current limitations in anticancer treatment. Full article
Show Figures

Graphical abstract

26 pages, 2593 KB  
Review
Experimental and In Silico Approaches to Study Carboxylesterase Substrate Specificity
by Sergio R. Ribone and Mario Alfredo Quevedo
J. Xenobiot. 2026, 16(1), 11; https://doi.org/10.3390/jox16010011 - 12 Jan 2026
Viewed by 163
Abstract
Human carboxylesterases (CES) are enzymes that play a central role in the metabolism and biotransformation of diverse endogenous substances and xenobiotics. The two most relevant isoforms, CES1 and CES2, are crucial in clinical pharmacotherapy as they catalyze the hydrolysis of numerous approved drugs [...] Read more.
Human carboxylesterases (CES) are enzymes that play a central role in the metabolism and biotransformation of diverse endogenous substances and xenobiotics. The two most relevant isoforms, CES1 and CES2, are crucial in clinical pharmacotherapy as they catalyze the hydrolysis of numerous approved drugs and prodrugs. Elucidating the structural basis of CES isoform substrate specificity is essential not only for understanding and anticipating the biological fate of administered drugs, but also for designing prodrugs with optimized site-specific bioactivation. Additionally, this knowledge is also important for the design of biomedically useful molecules such as subtype-targeted CES inhibitors and fluorescent probes. In this context, both experimental and computational methodologies have been used to explore the mechanistic and thermodynamic properties of CES-mediated catalysis. Experimental designs commonly employ recombinant CES or human tissue microsomes as enzyme sources, utilizing quantification methods such as spectrophotometry (UV and fluorescence) and mass spectrometry. Computational approaches fall into two categories: (1) modeling substrate: CES recognition and affinity (molecular docking, molecular dynamics simulation, and free-energy binding calculations), and (2) modeling substrate: CES reaction coordinates (hybrid QM/MM simulations). While experimental and theoretical approaches are highly synergistic in studying the catalytic properties of CES subtypes, they represent distinct technical and scientific fields. This review aims to provide an integrated discussion of the key concepts and the interplay between the most commonly used wet-lab and dry-lab strategies for investigating CES catalytic activity. We hope this report will serve as a concise resource for researchers exploring CES isoform specificity, enabling them to effectively utilize both experimental and computational methods. Full article
Show Figures

Figure 1

24 pages, 893 KB  
Review
Chikungunya in 2025: Comprehensive Insights into Virology, Diagnostics, Vaccines, and Antiviral Therapies
by Zeyong Zheng, Hua Ji, Zeping Shan, Jiangcheng Xu, Jiarui Li, Xueting Zhang, Jiajia Zheng, Shibo Jiang and Naru Zhang
Viruses 2026, 18(1), 100; https://doi.org/10.3390/v18010100 - 12 Jan 2026
Viewed by 213
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus prevalent in more than 110 countries and regions, including Africa, Asia, the Americas, and Europe. It can cause acute fever, rash, and severe joint pain, and some patients may develop chronic arthritis, which significantly impairs quality [...] Read more.
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus prevalent in more than 110 countries and regions, including Africa, Asia, the Americas, and Europe. It can cause acute fever, rash, and severe joint pain, and some patients may develop chronic arthritis, which significantly impairs quality of life. CHIKV infection can occasionally be fatal, with neurologic disease a particularly severe manifestation. Following its resurgence in 2005, CHIKV has emerged as a major threat to global public health. This review summarizes diagnostic techniques, advances in vaccine development, and the latest drug interventions for CHIKV. We also present an overview of the epidemiology, structure, and invasion mechanisms of epidemic hotspots in 2024–2025 and propose evidence-based strategies for effective prevention and control of CHIKV infection. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Figure 1

20 pages, 2139 KB  
Review
Application of Orthoflavivirus Pseudovirus Technology in Antiviral Research
by Yalan Zhang, Yaqi Zhao, Chaojun Wang, Yuanyuan Zhou, Hao Yuan, Xiaodan Li, Yong Wang and Xiaoling Pan
Int. J. Mol. Sci. 2026, 27(2), 722; https://doi.org/10.3390/ijms27020722 - 10 Jan 2026
Viewed by 120
Abstract
Arthropod-borne orthoflaviviruses, including dengue, Zika, Japanese encephalitis, yellow fever and West Nile viruses, pose a significant global public health threat, causing hundreds of millions of infections annually with severe clinical symptoms. However, the lack of effective vaccines and antiviral drugs, coupled with the [...] Read more.
Arthropod-borne orthoflaviviruses, including dengue, Zika, Japanese encephalitis, yellow fever and West Nile viruses, pose a significant global public health threat, causing hundreds of millions of infections annually with severe clinical symptoms. However, the lack of effective vaccines and antiviral drugs, coupled with the biosafety risks associated with handling live highly pathogenic strains, hinders progress in antiviral research. Pseudovirus technology, which uses single-round infectious viral particles lacking replication competence, has thus gained prominence as a safe and versatile tool for antiviral research. This review systematically summarizes the construction, optimization, and applications of orthoflavivirus pseudoviruses in antiviral research. The primary construction strategies of orthoflavivirus pseudoviruses rely on multi-plasmid co-transfection of viral replicons and structural protein expression vectors, leveraging the host cell secretory pathway to mimic natural viral assembly and maturation. The core applications of pseudovirus technology are highlighted, including high-throughput screening and detection of neutralizing antibodies, identification of antiviral drugs targeting viral entry or replication, and evaluation of vaccine immunogenicity. Despite these strengths, the approach still faces limitations, such as incomplete simulation of native viral structures and batch-to-batch titer variability, which may affect the physiological relevance of findings. In summary, orthoflavivirus pseudovirus technology has become an essential platform in both basic virology research and translational medicine, providing critical insights and tools in the ongoing fight against arthropod-borne orthoflaviviruses diseases. Full article
(This article belongs to the Section Molecular Microbiology)
Show Figures

Figure 1

35 pages, 4216 KB  
Review
Anticancer Activity of Schiff Base Metal Complexes Against MCF-7 Breast Cancer Cell Line
by Justyna Samaszko-Fiertek, Barbara Dmochowska and Janusz Madaj
Int. J. Mol. Sci. 2026, 27(2), 678; https://doi.org/10.3390/ijms27020678 - 9 Jan 2026
Viewed by 156
Abstract
According to the World Health Organization, breast cancer is the cancer that affects the largest number of people each year, especially women. Millions of women are diagnosed with it each year, and hundreds of thousands die from it. Research into new types of [...] Read more.
According to the World Health Organization, breast cancer is the cancer that affects the largest number of people each year, especially women. Millions of women are diagnosed with it each year, and hundreds of thousands die from it. Research into new types of drugs, including metal complexes, including those containing tetradentate Schiff bases as ligands, offers a chance to reduce this number. Various cell lines are being used to test their effectiveness in cancer therapy, with the MCF-7 cancer cell line being the most commonly used. A literature search was conducted in four major databases: PubMed, SciELO. The Boolean operator “and” was used to refine the search strategy, combining the terms Schiff base, breast cancer, MCF-7 and metal complexes. Studies published between 2020 and 2025 investigating the cytotoxic activity of metal complexes with Schiff base ligands on the MCF-7 breast cancer cell line were included in the analysis. Articles were considered eligible if they were written in English. As a result of the database search, 37 scientific articles were selected and divided into three groups based on the ligand structure. The largest group of articles described the synthesis, structure, and anticancer activity of metal complexes with ligands based on the salicylaldehyde structure. These were included in the first group of complexes described. The second, extremely interesting and promising group of compounds consisted of metal complexes with ligands containing a sulfur atom. The last group included metal complexes with Schiff base ligands that were not included in the two previously mentioned groups. As indicated by the research results contained in the reviewed articles, Schiff base metal complexes constitute an interesting group of compounds characterized by a range of activities, including anticancer activity, which may in the future be used in anticancer therapy. They may also represent a cheaper and more effective alternative to platinum-based drugs. Full article
(This article belongs to the Special Issue Synthesis and Structural Studies of Potential Anticancer Drugs)
Show Figures

Figure 1

29 pages, 4039 KB  
Review
Targeting Mesenchymal-Epidermal Transition (MET) Aberrations in Non-Small Cell Lung Cancer: Current Challenges and Therapeutic Advances
by Fahua Deng, Weijie Ma and Sixi Wei
Cancers 2026, 18(2), 207; https://doi.org/10.3390/cancers18020207 - 8 Jan 2026
Viewed by 327
Abstract
The mesenchymal–epithelial transition (MET) receptor is a tyrosine kinase activated by its sole known ligand, hepatocyte growth factor (HGF). MET signaling regulates key cellular processes, including proliferation, survival, migration, motility, and angiogenesis. Dysregulation and hyperactivation of this pathway are implicated in multiple malignancies, [...] Read more.
The mesenchymal–epithelial transition (MET) receptor is a tyrosine kinase activated by its sole known ligand, hepatocyte growth factor (HGF). MET signaling regulates key cellular processes, including proliferation, survival, migration, motility, and angiogenesis. Dysregulation and hyperactivation of this pathway are implicated in multiple malignancies, including lung, breast, colorectal, and gastrointestinal cancers. In non–small cell lung cancer (NSCLC), aberrant activation of the MET proto-oncogene contributes to 1% of known oncogenic drivers and is associated with poor clinical outcomes. Several mechanisms can induce MET hyperactivation, including MET gene amplification, transcriptional upregulation of MET or HGF, MET fusion genes, and MET exon 14 skipping mutations. Furthermore, MET pathway activation represents a frequent mechanism of acquired resistance to EGFR- and ALK-targeted tyrosine kinase inhibitors (TKIs) in EGFR- and ALK-driven NSCLCs. Although MET has long been recognized as a promising therapeutic target in NSCLC, the clinical efficacy of MET-targeted therapies has historically lagged behind that of EGFR and ALK inhibitors. Encouragingly, several MET TKIs such as capmatinib, tepotinib, and savolitinib have been approved for the treatment of MET exon 14 skipping mutations. They have also demonstrated potential in overcoming MET-driven resistance to EGFR TKIs or ALK TKIs. On 14 May 2025, the U.S. Food and Drug Administration granted accelerated approval to telisotuzumab vedotin-tllv for adult patients with locally advanced or metastatic non-squamous NSCLC whose tumors exhibit high c-Met protein overexpression and who have already received prior systemic therapy. In this review, we summarize the structure and physiological role of the MET receptor, the molecular mechanisms underlying aberrant MET activation, its contribution to acquired resistance against targeted therapies, and emerging strategies for effectively targeting MET alterations in NSCLC. Full article
Show Figures

Figure 1

Back to TopTop