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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,780)

Search Parameters:
Keywords = novel candidate drug

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 1479 KiB  
Article
Synthesis and Biological Evaluation of β-Phenylalanine Derivatives Containing Sulphonamide and Azole Moieties as Antiproliferative Candidates in Lung Cancer Models
by Vytautas Mickevičius, Kazimieras Anusevičius, Birutė Sapijanskaitė-Banevič, Ilona Jonuškienė, Linas Kapočius, Birutė Grybaitė, Ramunė Grigalevičiūtė and Povilas Kavaliauskas
Molecules 2025, 30(15), 3303; https://doi.org/10.3390/molecules30153303 - 7 Aug 2025
Abstract
In this study, a series of novel β-phenylalanine derivatives were synthesised and evaluated for their anticancer activity. The 3-(4-methylbenzene-1-sulfonamido)-3-phenylpropanoic acid (2) was prepared using β-phenylalanine as a core scaffold. The β-amino acid derivative 2 was converted to the [...] Read more.
In this study, a series of novel β-phenylalanine derivatives were synthesised and evaluated for their anticancer activity. The 3-(4-methylbenzene-1-sulfonamido)-3-phenylpropanoic acid (2) was prepared using β-phenylalanine as a core scaffold. The β-amino acid derivative 2 was converted to the corresponding hydrazide 4, which enabled the development of structurally diverse heterocyclic derivatives including pyrrole 5, pyrazole 6, thiadiazole 8, oxadiazole 11, triazoles 9 and 12 with Schiff base analogues 13 and series1,2,4-triazolo [3,4-b][1,3,4]thiadiazines 14. These modifications were designed to enhance chemical stability, solubility, and biological activity. All compounds were initially screened for cytotoxicity against the A549 human lung adenocarcinoma cell line, identifying N-[3-(3,5-dimethyl-1H-pyrazol-1-yl)-3-oxo-1-phenylpropyl]-4-methylbenzenesulfonamide (5) and (E)-N-{2-[4-[(4-chlorobenzylidene)amino]-5-thioxo-4,5-dihydro-1H-1,2,4-triazol-3-yl]-1-phenylethyl}-4-methylbenzenesulfonamide (13b) as the most active. The two lead candidates were further evaluated in H69 and H69AR small cell lung cancer lines to assess activity in drug-sensitive and multidrug-resistant models. Schiff base 13b containing a 4-chlorophenyl moiety, retained potent antiproliferative activity in both H69 and H69AR cells, comparable to cisplatin, while compound 5 lost efficacy in the resistant phenotype. These findings suggest Schiff base derivative 13b may overcome drug resistance mechanisms, a limitation commonly encountered with standard chemotherapeutics such as doxorubicin. These results demonstrate the potential role of β-phenylalanine derivatives, azole-containing sulphonamides, as promising scaffolds for the development of novel anticancer agents, particularly in the context of lung cancer and drug-resistant tumours. Full article
Show Figures

Graphical abstract

16 pages, 4197 KiB  
Review
Conformational Dynamics and Structural Transitions of Arginine Kinase: Implications for Catalysis and Allergen Control
by Sung-Min Kang
Life 2025, 15(8), 1248; https://doi.org/10.3390/life15081248 - 6 Aug 2025
Abstract
Arginine kinase is a key phosphagen kinase in invertebrates that facilitates rapid ATP regeneration by reversibly transferring phosphate groups between phosphoarginine and ADP. Structural studies have shown that the enzyme adopts distinct conformations in its ligand-free and ligand-bound states, known as the “open” [...] Read more.
Arginine kinase is a key phosphagen kinase in invertebrates that facilitates rapid ATP regeneration by reversibly transferring phosphate groups between phosphoarginine and ADP. Structural studies have shown that the enzyme adopts distinct conformations in its ligand-free and ligand-bound states, known as the “open” and “closed” forms, respectively. These conformational changes are crucial for catalytic activity, enabling precise positioning of active-site residues and loop closure during phosphoryl transfer. Transition-state analog complexes have provided additional insights by mimicking intermediate states of catalysis, supporting the functional relevance of the open/closed structural model. Furthermore, studies across multiple species reveal how monomeric and dimeric forms of arginine kinase contribute to its allosteric regulation and substrate specificity. Beyond its metabolic role, arginine kinase is also recognized as a major allergen in crustaceans. Its structural uniqueness and absence in vertebrates make it a promising candidate for selective drug targeting. By integrating crystallographic data with functional context, this review highlights conserved features and species-specific variations of arginine kinase that may inform the design of inhibitors. Such molecules have the potential to serve both as antiparasitic agents and as novel therapeutics to manage crustacean-related allergic responses in humans. Full article
(This article belongs to the Section Proteins and Proteomics)
Show Figures

Figure 1

13 pages, 286 KiB  
Review
Drug Repurposing and Artificial Intelligence in Multiple Sclerosis: Emerging Strategies for Precision Therapy
by Pedro Henrique Villar-Delfino, Paulo Pereira Christo and Caroline Maria Oliveira Volpe
Sclerosis 2025, 3(3), 28; https://doi.org/10.3390/sclerosis3030028 - 6 Aug 2025
Abstract
Multiple sclerosis (MS) is a chronic, immune-mediated disorder of the central nervous system (CNS) characterized by inflammation, demyelination, axonal degeneration, and gliosis. Its pathophysiology involves a complex interplay of genetic susceptibility, environmental triggers, and immune dysregulation, ultimately leading to progressive neurodegeneration and functional [...] Read more.
Multiple sclerosis (MS) is a chronic, immune-mediated disorder of the central nervous system (CNS) characterized by inflammation, demyelination, axonal degeneration, and gliosis. Its pathophysiology involves a complex interplay of genetic susceptibility, environmental triggers, and immune dysregulation, ultimately leading to progressive neurodegeneration and functional decline. Although significant advances have been made in disease-modifying therapies (DMTs), many patients continue to experience disease progression and unmet therapeutic needs. Drug repurposing—the identification of new indications for existing drugs—has emerged as a promising strategy in MS research, offering a cost-effective and time-efficient alternative to traditional drug development. Several compounds originally developed for other diseases, including immunomodulatory, anti-inflammatory, and neuroprotective agents, are currently under investigation for their efficacy in MS. Repurposed agents, such as selective sphingosine-1-phosphate (S1P) receptor modulators, kinase inhibitors, and metabolic regulators, have demonstrated potential in promoting neuroprotection, modulating immune responses, and supporting remyelination in both preclinical and clinical settings. Simultaneously, artificial intelligence (AI) is transforming drug discovery and precision medicine in MS. Machine learning and deep learning models are being employed to analyze high-dimensional biomedical data, predict drug–target interactions, streamline drug repurposing workflows, and enhance therapeutic candidate selection. By integrating multiomics and neuroimaging data, AI tools facilitate the identification of novel targets and support patient stratification for individualized treatment. This review highlights recent advances in drug repurposing and discovery for MS, with a particular emphasis on the emerging role of AI in accelerating therapeutic innovation and optimizing treatment strategies. Full article
Show Figures

Graphical abstract

33 pages, 640 KiB  
Review
Future Pharmacotherapy for Bipolar Disorders: Emerging Trends and Personalized Approaches
by Giuseppe Marano, Francesco Maria Lisci, Gianluca Boggio, Ester Maria Marzo, Francesca Abate, Greta Sfratta, Gianandrea Traversi, Osvaldo Mazza, Roberto Pola, Gabriele Sani, Eleonora Gaetani and Marianna Mazza
Future Pharmacol. 2025, 5(3), 42; https://doi.org/10.3390/futurepharmacol5030042 - 4 Aug 2025
Viewed by 151
Abstract
Background: Bipolar disorder (BD) is a chronic and disabling psychiatric condition characterized by recurring episodes of mania, hypomania, and depression. Despite the availability of mood stabilizers, antipsychotics, and antidepressants, long-term management remains challenging due to incomplete symptom control, adverse effects, and high relapse [...] Read more.
Background: Bipolar disorder (BD) is a chronic and disabling psychiatric condition characterized by recurring episodes of mania, hypomania, and depression. Despite the availability of mood stabilizers, antipsychotics, and antidepressants, long-term management remains challenging due to incomplete symptom control, adverse effects, and high relapse rates. Methods: This paper is a narrative review aimed at synthesizing emerging trends and future directions in the pharmacological treatment of BD. Results: Future pharmacotherapy for BD is likely to shift toward precision medicine, leveraging advances in genetics, biomarkers, and neuroimaging to guide personalized treatment strategies. Novel drug development will also target previously underexplored mechanisms, such as inflammation, mitochondrial dysfunction, circadian rhythm disturbances, and glutamatergic dysregulation. Physiological endophenotypes, such as immune-metabolic profiles, circadian rhythms, and stress reactivity, are emerging as promising translational tools for tailoring treatment and reducing associated somatic comorbidity and mortality. Recognition of the heterogeneous longitudinal trajectories of BD, including chronic mixed states, long depressive episodes, or intermittent manic phases, has underscored the value of clinical staging models to inform both pharmacological strategies and biomarker research. Disrupted circadian rhythms and associated chronotypes further support the development of individualized chronotherapeutic interventions. Emerging chronotherapeutic approaches based on individual biological rhythms, along with innovative monitoring strategies such as saliva-based lithium sensors, are reshaping the future landscape. Anti-inflammatory agents, neurosteroids, and compounds modulating oxidative stress are emerging as promising candidates. Additionally, medications targeting specific biological pathways implicated in bipolar pathophysiology, such as N-methyl-D-aspartate (NMDA) receptor modulators, phosphodiesterase inhibitors, and neuropeptides, are under investigation. Conclusions: Advances in pharmacogenomics will enable clinicians to predict individual responses and tolerability, minimizing trial-and-error prescribing. The future landscape may also incorporate digital therapeutics, combining pharmacotherapy with remote monitoring and data-driven adjustments. Ultimately, integrating innovative drug therapies with personalized approaches has the potential to enhance efficacy, reduce adverse effects, and improve long-term outcomes for individuals with bipolar disorder, ushering in a new era of precision psychiatry. Full article
Show Figures

Figure 1

21 pages, 6211 KiB  
Article
In Silico and In Vitro Potential Antifungal Insights of Insect-Derived Peptides in the Management of Candida sp. Infections
by Catarina Sousa, Alaka Sahoo, Shasank Sekhar Swain, Payal Gupta, Francisco Silva, Andreia S. Azevedo and Célia Fortuna Rodrigues
Int. J. Mol. Sci. 2025, 26(15), 7449; https://doi.org/10.3390/ijms26157449 - 1 Aug 2025
Viewed by 242
Abstract
The worldwide increase in antifungal resistance, particularly in Candida sp., requires the exploration of novel therapeutic agents. Natural compounds have been a rich source of antimicrobial molecules, where peptides constitute the class of the most bioactive components. Therefore, this study looks into the [...] Read more.
The worldwide increase in antifungal resistance, particularly in Candida sp., requires the exploration of novel therapeutic agents. Natural compounds have been a rich source of antimicrobial molecules, where peptides constitute the class of the most bioactive components. Therefore, this study looks into the target-specific binding efficacy of insect-derived antifungal peptides (n = 37) as possible alternatives to traditional antifungal treatments. Using computational methods, namely the HPEPDOCK and HDOCK platforms, molecular docking was performed to evaluate the interactions between selected key fungal targets, lanosterol 14-demethylase, or LDM (PDB ID: 5V5Z), secreted aspartic proteinase-5, or Sap-5 (PDB ID: 2QZX), N-myristoyl transferase, or NMT (PDB ID: 1NMT), and dihydrofolate reductase, or DHFR, of C. albicans. The three-dimensional peptide structure was modelled through the PEP-FOLD 3.5 tool. Further, we predicted the physicochemical properties of these peptides through the ProtParam and PEPTIDE 2.0 tools to assess their drug-likeness and potential for therapeutic applications. In silico results show that Blap-6 from Blaps rhynchopeter and Gomesin from Acanthoscurria gomesiana have the most antifungal potential against all four targeted proteins in Candida sp. Additionally, a molecular dynamics simulation study of LDM-Blap-6 was carried out at 100 nanoseconds. The overall predictions showed that both have strong binding abilities and are good candidates for drug development. In in vitro studies, Gomesin achieved complete biofilm eradication in three out of four Candida species, while Blap-6 showed moderate but consistent reduction across all species. C. tropicalis demonstrated relative resistance to complete eradication by both peptides. The present study provides evidence to support the antifungal activity of certain insect peptides, with potential to be used as alternative drugs or as a template for a new synthetic or modified peptide in pursuit of effective therapies against Candida spp. Full article
Show Figures

Figure 1

17 pages, 7013 KiB  
Article
A Novel HDAC6 Inhibitor Ameliorates Imiquimod-Induced Psoriasis-Like Inflammation in Mice
by Anqi Cao, Yurong Li, Yanqiao Feng, Xiaoquan Wang, Wenyu Wei, Hongyan Sun and Junmin Quan
Molecules 2025, 30(15), 3224; https://doi.org/10.3390/molecules30153224 - 31 Jul 2025
Viewed by 274
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by abnormal proliferation of keratinocytes and infiltration of inflammatory cells. Significant challenges remain in developing effective and safe targeted therapies for psoriasis. Here, we reported the discovery of novel cystamine derivatives for the treatment of [...] Read more.
Psoriasis is a chronic inflammatory skin disease characterized by abnormal proliferation of keratinocytes and infiltration of inflammatory cells. Significant challenges remain in developing effective and safe targeted therapies for psoriasis. Here, we reported the discovery of novel cystamine derivatives for the treatment of psoriasis. These compounds effectively attenuated LPS-induced inflammation in vitro, and the optimal candidate CS1 ameliorated imiquimod-induced psoriasis-like inflammation in mice. Mechanistically, CS1 bound and inhibited the deacetylase HDAC6, subsequently inhibited the AKT, MAPK, and STAT3 pathways, attenuated the hyperproliferation and altered differentiation of keratinocytes and reduced the infiltration of immune cells. These findings suggest that HDAC6 may serve as a potential target for drug development in the treatment of psoriasis. Full article
Show Figures

Graphical abstract

18 pages, 1988 KiB  
Article
Computational Design of Potentially Multifunctional Antimicrobial Peptide Candidates via a Hybrid Generative Model
by Fangli Ying, Wilten Go, Zilong Li, Chaoqian Ouyang, Aniwat Phaphuangwittayakul and Riyad Dhuny
Int. J. Mol. Sci. 2025, 26(15), 7387; https://doi.org/10.3390/ijms26157387 - 30 Jul 2025
Viewed by 272
Abstract
Antimicrobial peptides (AMPs) provide a robust alternative to conventional antibiotics, combating escalating microbial resistance through their diverse functions and broad pathogen-targeting abilities. While current deep learning technologies enhance AMP generation, they face challenges in developing multifunctional AMPs due to intricate amino acid interdependencies [...] Read more.
Antimicrobial peptides (AMPs) provide a robust alternative to conventional antibiotics, combating escalating microbial resistance through their diverse functions and broad pathogen-targeting abilities. While current deep learning technologies enhance AMP generation, they face challenges in developing multifunctional AMPs due to intricate amino acid interdependencies and limited consideration of diverse functional activities. To overcome this challenge, we introduce a novel de novo multifunctional AMP design framework that enhances a Feedback Generative Adversarial Network (FBGAN) by integrating a global quantitative AMP activity regression module and a multifunctional-attribute integrated prediction module. This integrated approach not only facilitates the automated generation of potential AMP candidates, but also optimizes the network’s ability to assess their multifunctionality. Initially, by integrating an effective pre-trained regression and classification model with feedback-loop mechanisms, our model can not only identify potential valid AMP candidates, but also optimizes computational predictions of Minimum Inhibitory Concentration (MIC) values. Subsequently, we employ a combinatorial predictor to simultaneously identify and predict five multifunctional AMP bioactivities, enabling the generation of multifunctional AMPs. The experimental results demonstrate the efficiency of generating AMPs with multiple enhanced antimicrobial properties, indicating that our work can provide a valuable reference for combating multi-drug-resistant infections. Full article
(This article belongs to the Special Issue Application of Artificial Intelligence in Molecular Sciences)
Show Figures

Figure 1

31 pages, 19845 KiB  
Article
In Silico Approaches for the Discovery of Novel Pyrazoline Benzenesulfonamide Derivatives as Anti-Breast Cancer Agents Against Estrogen Receptor Alpha (ERα)
by Dadang Muhammad Hasyim, Ida Musfiroh, Rudi Hendra, Taufik Muhammad Fakih, Nur Kusaira Khairul Ikram and Muchtaridi Muchtaridi
Appl. Sci. 2025, 15(15), 8444; https://doi.org/10.3390/app15158444 - 30 Jul 2025
Viewed by 398
Abstract
Estrogen receptor alpha (ERα) plays a vital role in the development and progression of breast cancer by regulating the expression of genes associated with cell proliferation in breast tissue. ERα inhibition is a key strategy in the prevention and treatment of breast cancer. [...] Read more.
Estrogen receptor alpha (ERα) plays a vital role in the development and progression of breast cancer by regulating the expression of genes associated with cell proliferation in breast tissue. ERα inhibition is a key strategy in the prevention and treatment of breast cancer. Previous research modified chalcone compounds into pyrazoline benzenesulfonamide derivatives (Modifina) which show activity as an ERα inhibitor. This study aimed to design novel pyrazoline benzenesulfonamide derivatives (PBDs) as ERα antagonists using in silico approaches. Structure-based and ligand-based drug design approaches were used to create drug target molecules. A total of forty-five target molecules were initially designed and screened for drug likeness (Lipinski’s rule of five), cytotoxicity, pharmacokinetics and toxicity using a web-based prediction tools. Promising candidates were subjected to molecular docking using AutoDock 4.2.6 to evaluate their binding interaction with ERα, followed by molecular dynamics simulations using AMBER20 to assess complex stability. A pharmacophore model was also generated using LigandScout 4.4.3 Advanced. The molecular docking results identified PBD-17 and PBD-20 as the most promising compounds, with binding free energies (ΔG) of −11.21 kcal/mol and −11.15 kcal/mol, respectively. Both formed hydrogen bonds with key ERα residues ARG394, GLU353, and LEU387. MM-PBSA further supported these findings, with binding energies of −58.23 kJ/mol for PDB-17 and −139.46 kJ/mol for PDB-20, compared to −145.31 kJ/mol, for the reference compound, 4-OHT. Although slightly less favorable than 4-OHT, PBD-20 demonstrated a more stable interaction with ERα than PBD-17. Furthermore, pharmacophore screening showed that both PBD-17 and PBD-20 aligned well with the generated model, each achieving a match score of 45.20. These findings suggest that PBD-17 and PBD-20 are promising lead compounds for the development of a potent ERα inhibitor in breast cancer therapy. Full article
(This article belongs to the Special Issue Drug Discovery and Delivery in Medicinal Chemistry)
Show Figures

Figure 1

24 pages, 2944 KiB  
Article
Oral Pharmacokinetic Evaluation of a Microemulsion-Based Delivery System for Novel A190 Prodrugs
by Sagun Poudel, Chaolong Qin, Rudra Pangeni, Ziwei Hu, Grant Berkbigler, Madeline Gunawardena, Adam S. Duerfeldt and Qingguo Xu
Biomolecules 2025, 15(8), 1101; https://doi.org/10.3390/biom15081101 - 30 Jul 2025
Viewed by 515
Abstract
Peroxisome proliferator-activated receptor alpha (PPARα) is a key regulator of lipid metabolism, making its agonists valuable therapeutic targets for various diseases, including chronic peripheral neuropathy. Existing PPARα agonists face limitations such as poor selectivity, sub-optimal bioavailability, and safety concerns. We previously demonstrated that [...] Read more.
Peroxisome proliferator-activated receptor alpha (PPARα) is a key regulator of lipid metabolism, making its agonists valuable therapeutic targets for various diseases, including chronic peripheral neuropathy. Existing PPARα agonists face limitations such as poor selectivity, sub-optimal bioavailability, and safety concerns. We previously demonstrated that A190, a novel, potent, and selective PPARα agonist, effectively alleviates chemotherapy-induced peripheral neuropathy and CFA-induced inflammatory pain as a non-opioid therapeutic agent. However, A190 alone has solubility and permeability issues that limits its oral delivery. To overcome this challenge, in this study, four new-generation ester prodrugs of A190; A190-PD-9 (methyl ester), A190-PD-14 (ethyl ester), A190-PD-154 (isopropyl ester), and A190-PD-60 (cyclic carbonate) were synthesized and evaluated for their enzymatic bioconversion and chemical stability. The lead candidate, A190-PD-60, was further formulated as a microemulsion (A190-PD-60-ME) and optimized via Box–Behnken design. A190-PD-60-ME featured nano-sized droplets (~120 nm), low polydispersity (PDI < 0.3), and high drug loading (>90%) with significant improvement in artificial membrane permeability. Crucially, pharmacokinetic evaluation in rats demonstrated that A190-PD-60-ME reached a 16.6-fold higher Cmax (439 ng/mL) and a 5.9-fold increase in relative oral bioavailability compared with an A190-PD-60 dispersion. These findings support the combined prodrug-microemulsion approach as a promising strategy to overcome oral bioavailability challenges and advance PPARα-targeted therapies. Full article
Show Figures

Figure 1

11 pages, 2255 KiB  
Article
Potential Inhibitors of Human–Naegleria fowleri Interactions: An In Vitro Extracellular Matrix-Based Model
by Javier Chao-Pellicer, Iñigo Arberas-Jiménez, Ines Sifaoui, Ana R. Díaz-Marrero, José J. Fernández, Melissa Jamerson, José E. Piñero and Jacob Lorenzo-Morales
Mar. Drugs 2025, 23(8), 306; https://doi.org/10.3390/md23080306 - 30 Jul 2025
Viewed by 250
Abstract
Primary amoebic meningoencephalitis (PAM) is a rapidly progressive and fulminant disease that affects the central nervous system caused by the free-living amoeba Naegleria fowleri. The adhesion to extracellular matrix (ECM) proteins is considered as one of the key steps in the success [...] Read more.
Primary amoebic meningoencephalitis (PAM) is a rapidly progressive and fulminant disease that affects the central nervous system caused by the free-living amoeba Naegleria fowleri. The adhesion to extracellular matrix (ECM) proteins is considered as one of the key steps in the success of the infection and could represent an interesting target to be explored in the prevention and treatment of the disease. In this work, the effect of two sesquiterpenes with proven anti-Naegleria activity on the adhesion of the parasite was evaluated using an in vitro ECM-based model, compared with the reference drugs amphotericin B and staurosporine. Both laurinterol and (+)-elatol inhibited the adhesion of the N. fowleri trophozoites to the main proteins of the ECM when treating them at different concentrations and exposure times. This work not only reinforces the therapeutic potential of laurinterol and (+)-elatol against N. fowleri infection but also introduces the application of ECM-based adhesion assays as a novel and valuable tool for screening candidate compounds that disrupt host–pathogen interactions critical to PAM pathogenesis. Full article
(This article belongs to the Special Issue Marine Antiparasitic Agents, 2nd Edition)
Show Figures

Graphical abstract

25 pages, 14674 KiB  
Article
Eco-Friendly Silver Nanoparticles Synthesis Method Using Medicinal Plant Fungal Endophytes—Biological Activities and Molecular Docking Analyses
by Harish Chandra, Sagar Vishwakarma, Nilesh Makwana, Arun S. Kharat, Vijeta Chaudhry, Sumit Chand, Rajendra Prasad, Soban Prakash, Annapurna Katara, Archana Yadav, Manisha Nigam and Abhay Prakash Mishra
Biology 2025, 14(8), 950; https://doi.org/10.3390/biology14080950 - 28 Jul 2025
Viewed by 495
Abstract
The integration of nanotechnology and green synthesis strategies provides innovative solutions in biomedicine. This study focuses on the biofabrication of silver nanoparticles (AgNPs) using Corynespora smithii, an endophytic fungus isolated from Bergenia ciliata. The eco-friendly synthesis process employed fungal extracts as [...] Read more.
The integration of nanotechnology and green synthesis strategies provides innovative solutions in biomedicine. This study focuses on the biofabrication of silver nanoparticles (AgNPs) using Corynespora smithii, an endophytic fungus isolated from Bergenia ciliata. The eco-friendly synthesis process employed fungal extracts as reducing and stabilizing agents thereby minimizing the need for hazardous chemicals. The AgNPs demonstrated strong potent biological activities, showcasing significant antioxidant, antibacterial, and anticancer properties. The antibacterial efficacy was demonstrated against various Gram-positive and Gram-negative bacteria, while cytotoxicity on the A549 lung cancer cell line revealed an IC50 value of 10.46 µg/mL. A molecular docking analysis revealed interactions between the major bioactive compound, dimethylsulfoxonium formylmethylide, and the pathogenic proteins, Staphylococcus aureus and Salmonella typhi, displaying moderate binding affinities. Furthermore, the ADME analysis of dimethylsulfoxonium formylmethylide indicated favourable pharmacokinetic properties, including high gastrointestinal absorption, minimal lipophilicity, and low potential for drug–drug interactions, making it a promising candidate for oral drug formulations. These findings further support the compound’s suitability for biomedical applications. This research emphasizes the potential of C. smithii as a sustainable source for synthesizing bioactive nanoparticles, paving the way for their application in developing novel therapeutic agents. This study highlights the significance of harnessing endophytic fungi from medicinal plants for sustainable nanotechnology advancements. Full article
Show Figures

Graphical abstract

38 pages, 2987 KiB  
Review
Benzothiazole-Based Therapeutics: FDA Insights and Clinical Advances
by Subba Rao Cheekatla
Chemistry 2025, 7(4), 118; https://doi.org/10.3390/chemistry7040118 - 25 Jul 2025
Viewed by 840
Abstract
Benzothiazole derivatives have emerged as being highly significant in drug discovery due to their versatile biological activities and structural adaptability. Incorporating nitrogen and sulfur, this fused heterocyclic scaffold exhibits wide-ranging pharmacological properties, including anticancer, antimicrobial, anti-inflammatory, antidiabetic, neuroprotective, and diagnostic applications. A diverse [...] Read more.
Benzothiazole derivatives have emerged as being highly significant in drug discovery due to their versatile biological activities and structural adaptability. Incorporating nitrogen and sulfur, this fused heterocyclic scaffold exhibits wide-ranging pharmacological properties, including anticancer, antimicrobial, anti-inflammatory, antidiabetic, neuroprotective, and diagnostic applications. A diverse set of clinically approved and investigational compounds, such as flutemetamol for Alzheimer’s diagnosis, riluzole for ALS, and quizartinib for AML, illustrates the scaffold’s therapeutic potential in varied applications. These agents act via mechanisms such as enzyme inhibition, receptor modulation, and amyloid imaging, demonstrating the scaffold’s high binding affinity and target specificity. Advances in synthetic strategies and our understanding of structure–activity relationships (SARs) continue to drive the development of novel benzothiazole-based therapeutics with improved potency, selectivity, and safety profiles. We also emphasize recent in vitro and in vivo studies, including drug candidates in clinical trials, to provide a comprehensive perspective on the therapeutic potential of benzothiazole-based compounds in modern drug discovery. This review brings together recent progress to help guide the development of new benzothiazole-based compounds for future therapeutic applications. Full article
Show Figures

Graphical abstract

23 pages, 4774 KiB  
Article
Chlorogenic Acid and Cinnamaldehyde in Breast Cancer Cells: Predictive Examination of Pharmacokinetics and Binding Thermodynamics with the Key Mediators of PI3K/Akt Signaling
by Yusuff Olayiwola and Lauren Gollahon
Biomedicines 2025, 13(8), 1810; https://doi.org/10.3390/biomedicines13081810 - 24 Jul 2025
Viewed by 346
Abstract
Background/Objective: In the pursuit of identifying novel therapeutic agents against breast cancer, a major priority is finding agents that effectively and safely inhibit the signaling pathways sustaining cancer cells. To better focus research efforts in validating such candidates, this in silico study assessed [...] Read more.
Background/Objective: In the pursuit of identifying novel therapeutic agents against breast cancer, a major priority is finding agents that effectively and safely inhibit the signaling pathways sustaining cancer cells. To better focus research efforts in validating such candidates, this in silico study assessed the pharmacokinetic profiles, thermodynamics, and binding affinity of chlorogenic acid and cinnamaldehyde with the upstream mediators of the Akt pathway implicated in breast cancer cells. Methods: Various software and online tools were used to conduct molecular docking of the small molecules with the proteins PI3K, Akt, and PDK1, and to examine their absorption, distribution, metabolism, elimination, and toxicity (ADMET) profile. Results: The results show strong binding energy (all within the range of those of FDA-approved drugs) and thermostability between the compounds and the proteins. The phytochemicals were predicted to have moderate oral bioavailability and tissue distribution, and were identified as substrates of drug metabolizing enzymes, but not deactivated. Conclusion: Although these predictive data warrant confirmation in a biological system, they suggest that the compounds have good pharmacokinetics and are strong inhibitors of the Akt pathway, with great potential to shut down breast cancer cell invasion and migration. These data also inform more efficient experimental designs for our planned in vivo studies. Full article
(This article belongs to the Special Issue Signaling of Protein Kinases in Development and Disease)
Show Figures

Figure 1

23 pages, 2594 KiB  
Article
Formation and Characterization of Xylitol-Modified Glycidyl Methacrylate-co-Ethyl Methacrylate Matrices for Controlled Release of Antimicrobial Compounds
by Adam Chyzy, Przemysław Gnatowski, Edyta Piłat, Maciej Sienkiewicz, Katarzyna Wozniak, Marta Wojnicka, Krzysztof Brzezinski and Marta E. Plonska-Brzezinska
Molecules 2025, 30(15), 3083; https://doi.org/10.3390/molecules30153083 - 23 Jul 2025
Viewed by 198
Abstract
Wounds are undeniably important gateways for pathogens to enter the body. In addition to their detrimental local effects, they can also cause adverse systemic effects. For this reason, developing methods for eradicating pathogens from wounds is a challenging medical issue. Polymers, particularly hydrogels, [...] Read more.
Wounds are undeniably important gateways for pathogens to enter the body. In addition to their detrimental local effects, they can also cause adverse systemic effects. For this reason, developing methods for eradicating pathogens from wounds is a challenging medical issue. Polymers, particularly hydrogels, are one of the more essential materials for designing novel drug-delivery systems, thanks to the ease of tuning their structures. This work exploits this property by utilizing copolymerization, microwave modification, and drug-loading processes to obtain antibacterial gels. Synthesized xylitol-modified glycidyl methacrylate-co-ethyl methacrylate ([P(EMA)-co-(GMA)]-Xyl]) matrices were loaded with bacitracin, gentian violet, furazidine, and brilliant green, used as active pharmaceutical ingredients (APIs). The hydrophilic properties, API release mechanism, and antibacterial properties of the obtained hydrogels against Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus epidermidis containing [P(EMA)-co-(GMA)]-Xyl] were studied. The hydrogels with the APIs efficiently inhibit bacteria growth with low doses of drugs, and our findings are statistically significant, confirmed with ANOVA analysis at p = 0.05. The results confirmed that the proposed system is hydrophilic and has extended the drug-release capabilities of APIs with a controlled burst effect based on [P(EMA)-co-(GMA)]-Xyl] content in the hydrogel. Hydrogels are characterized by the prolonged release of APIs in a very short time (a few minutes). Although the amount of released APIs is about 10%, it still exceeds the minimum inhibitory concentrations of drugs. Several kinetic models (first-order, second-order, Baker–Lonsdale, and Korsmeyer–Peppas) were applied to fit the API release data from the [P(EMA)-co-(GMA)]-Xyl-based hydrogel. The best fit of the Korsmeyer–Peppas kinetic model to the experimental data was determined, and it was confirmed that a diffusion-controlled release mechanism of the APIs from the studied hydrogels is dominant, which is desirable for applications requiring a consistent, controlled release of therapeutic agents. A statistical analysis of API release using Linear Mixed Model was performed, examining the relationship between % mass of API, sample (hydrogels and control), time, sample–time interaction, and variability between individuals. The model fits the data well, as evidenced by the determination coefficients close to 1. The analyzed interactions in the data are reliable and statistically significant (p < 0.001). The outcome of this study suggests that the presented acrylate-based gel is a promising candidate for developing wound dressings. Full article
(This article belongs to the Special Issue Advances in Functional Polymers and Their Applications)
Show Figures

Figure 1

21 pages, 14290 KiB  
Article
Identifying Therapeutic Targets for Amyotrophic Lateral Sclerosis Through Modeling of Multi-Omics Data
by François Xavier Blaudin de Thé, Cornelius J. H. M. Klemann, Ward De Witte, Joanna Widomska, Philippe Delagrange, Clotilde Mannoury La Cour, Mélanie Fouesnard, Sahar Elouej, Keith Mayl, Nicolas Lévy, Johannes Krupp, Ross Jeggo, Philippe Moingeon and Geert Poelmans
Int. J. Mol. Sci. 2025, 26(15), 7087; https://doi.org/10.3390/ijms26157087 - 23 Jul 2025
Viewed by 361
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that primarily affects motor neurons, leading to loss of muscle control, and, ultimately, respiratory failure and death. Despite some advances in recent years, the underlying genetic and molecular mechanisms of ALS remain largely elusive. [...] Read more.
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that primarily affects motor neurons, leading to loss of muscle control, and, ultimately, respiratory failure and death. Despite some advances in recent years, the underlying genetic and molecular mechanisms of ALS remain largely elusive. In this respect, a better understanding of these mechanisms is needed to identify new and biologically relevant therapeutic targets that could be developed into treatments that are truly disease-modifying, in that they address the underlying causes rather than the symptoms of ALS. In this study, we used two approaches to model multi-omics data in order to map and elucidate the genetic and molecular mechanisms involved in ALS, i.e., the molecular landscape building approach and the Patrimony platform. These two methods are complementary because they rely upon different omics data sets, analytic methods, and scoring systems to identify and rank therapeutic target candidates. The orthogonal combination of the two modeling approaches led to significant convergences, as well as some complementarity, both for validating existing therapeutic targets and identifying novel targets. As for validating existing targets, we found that, out of 217 different targets that have been or are being investigated for drug development, 10 have high scores in both the landscape and Patrimony models, suggesting that they are highly relevant for ALS. Moreover, through both models, we identified or corroborated novel putative drug targets for ALS. A notable example of such a target is MATR3, a protein that has strong genetic, molecular, and functional links with ALS pathology. In conclusion, by using two distinct and highly complementary disease modeling approaches, this study enhances our understanding of ALS pathogenesis and provides a framework for prioritizing new therapeutic targets. Moreover, our findings underscore the potential of leveraging multi-omics analyses to improve target discovery and accelerate the development of effective treatments for ALS, and potentially other related complex human diseases. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

Back to TopTop