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 (559)

Search Parameters:
Keywords = antibiotic-drug interactions

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 806 KiB  
Proceeding Paper
Enterococcus faecalis Biofilm: A Clinical and Environmental Hazard
by Bindu Sadanandan and Kavyasree Marabanahalli Yogendraiah
Med. Sci. Forum 2025, 35(1), 5; https://doi.org/10.3390/msf2025035005 - 5 Aug 2025
Abstract
This review explores the biofilm architecture and drug resistance of Enterococcus faecalis in clinical and environmental settings. The biofilm in E. faecalis is a heterogeneous, three-dimensional, mushroom-like or multilayered structure, characteristically forming diplococci or short chains interspersed with water channels for nutrient exchange [...] Read more.
This review explores the biofilm architecture and drug resistance of Enterococcus faecalis in clinical and environmental settings. The biofilm in E. faecalis is a heterogeneous, three-dimensional, mushroom-like or multilayered structure, characteristically forming diplococci or short chains interspersed with water channels for nutrient exchange and waste removal. Exopolysaccharides, proteins, lipids, and extracellular DNA create a protective matrix. Persister cells within the biofilm contribute to antibiotic resistance and survival. The heterogeneous architecture of the E. faecalis biofilm contains both dense clusters and loosely packed regions that vary in thickness, ranging from 10 to 100 µm, depending on the environmental conditions. The pathogenicity of the E. faecalis biofilm is mediated through complex interactions between genes and virulence factors such as DNA release, cytolysin, pili, secreted antigen A, and microbial surface components that recognize adhesive matrix molecules, often involving a key protein called enterococcal surface protein (Esp). Clinically, it is implicated in a range of nosocomial infections, including urinary tract infections, endocarditis, and surgical wound infections. The biofilm serves as a nidus for bacterial dissemination and as a reservoir for antimicrobial resistance. The effectiveness of first-line antibiotics (ampicillin, vancomycin, and aminoglycosides) is diminished due to reduced penetration, altered metabolism, increased tolerance, and intrinsic and acquired resistance. Alternative strategies for biofilm disruption, such as combination therapy (ampicillin with aminoglycosides), as well as newer approaches, including antimicrobial peptides, quorum-sensing inhibitors, and biofilm-disrupting agents (DNase or dispersin B), are also being explored to improve treatment outcomes. Environmentally, E. faecalis biofilms contribute to contamination in water systems, food production facilities, and healthcare environments. They persist in harsh conditions, facilitating the spread of multidrug-resistant strains and increasing the risk of transmission to humans and animals. Therefore, understanding the biofilm architecture and drug resistance is essential for developing effective strategies to mitigate their clinical and environmental impact. Full article
(This article belongs to the Proceedings of The 4th International Electronic Conference on Antibiotics)
Show Figures

Figure 1

26 pages, 1426 KiB  
Review
Mycobacteriophages in the Treatment of Mycobacterial Infections: From Compassionate Use to Targeted Therapy
by Magdalena Druszczynska, Beata Sadowska, Agnieszka Zablotni, Lesia Zhuravska, Jakub Kulesza and Marek Fol
Appl. Sci. 2025, 15(15), 8543; https://doi.org/10.3390/app15158543 - 31 Jul 2025
Viewed by 331
Abstract
This review addresses the urgent need for alternative strategies to combat drug-resistant mycobacterial infections, including multidrug-resistant (MDR) and extensively drug-resistant (XDR) tuberculosis, as well as non-tuberculous mycobacterial (NTM) diseases. Traditional antibiotics are increasingly limited by resistance, toxicity, and poor efficacy, particularly in immunocompromised [...] Read more.
This review addresses the urgent need for alternative strategies to combat drug-resistant mycobacterial infections, including multidrug-resistant (MDR) and extensively drug-resistant (XDR) tuberculosis, as well as non-tuberculous mycobacterial (NTM) diseases. Traditional antibiotics are increasingly limited by resistance, toxicity, and poor efficacy, particularly in immunocompromised patients. A comprehensive literature search was conducted using PubMed, Scopus, and Google Scholar, covering publications primarily from 2000 to 2025. Only articles published in English were included to ensure consistency in data interpretation. Search terms included “mycobacteriophages,” “phage therapy,” “drug-resistant mycobacteria, “diagnostic phages,” and “phage engineering.” The review examines the therapeutic and diagnostic potential of mycobacteriophages—viruses that specifically infect mycobacteria—focusing on their molecular biology, engineering advances, delivery systems, and clinical applications. Evidence suggests that mycobacteriophages offer high specificity, potent bactericidal activity, and adaptability, positioning them as promising candidates for targeted therapy. Although significant obstacles remain—including immune interactions, limited host range, and regulatory challenges—rapid progress in synthetic biology and delivery platforms continues to expand their clinical potential. As research advances and clinical frameworks evolve, mycobacteriophages are poised to become a valuable asset in the fight against drug-resistant mycobacterial diseases, offering new precision-based solutions where conventional therapies fail. Full article
(This article belongs to the Special Issue Tuberculosis—a Millennial Disease in the Age of New Technologies)
Show Figures

Figure 1

15 pages, 2101 KiB  
Article
Identification of Two Critical Contact Residues in a Pathogenic Epitope from Tetranectin for Monoclonal Antibody Binding and Preparation of Single-Chain Variable Fragments
by Juncheng Wang, Meng Liu, Rukhshan Zahid, Wenjie Zhang, Zecheng Cai, Yan Liang, Die Li, Jiasheng Hao and Yuekang Xu
Biomolecules 2025, 15(8), 1100; https://doi.org/10.3390/biom15081100 - 30 Jul 2025
Viewed by 276
Abstract
Sepsis is a fetal disease that requires a clear diagnostic biomarker for timely antibiotic treatment. Recent research has identified a pyroptosis-inducing epitope known as P5-5 in tetranectin (TN), a plasma protein produced by monocytes. Previously, we produced a 12F1 monoclonal antibody against the [...] Read more.
Sepsis is a fetal disease that requires a clear diagnostic biomarker for timely antibiotic treatment. Recent research has identified a pyroptosis-inducing epitope known as P5-5 in tetranectin (TN), a plasma protein produced by monocytes. Previously, we produced a 12F1 monoclonal antibody against the P5-5 and discovered that it could not only diagnose the presence but also monitor the progress of sepsis in the clinic. In the current study, we further investigated the structure site of the P5-5 and the recognition mechanism between the 12F1 mAb and the P5-5 epitope. To this end, 10 amino acids (NDALYEYLRQ) in the P5-5 were individually mutated to alanine, and their binding to the mAb was tested to confirm the most significant antigenic recognition sites. In the meanwhile, the spatial conformation of 12F1 mAb variable regions was modeled, and the molecular recognition mechanisms in detail of the mAb to the P5-5 epitope were further studied by molecular docking. Following epitope prediction and experimental verification, we demonstrated that the motif “DALYEYL” in the epitope sequence position 2−8 of TN-P5-5 is the major binding region for mAb recognition, in which two residues (4L and 8L) were essential for the interaction between the P5-5 epitope and the 12F1 mAb. Therefore, our study greatly narrowed down the previously reported motif from ten to seven amino acids and identified two Leu as critical contact residues. Finally, a single-chain variable fragment (scFv) from the 12F1 hybridoma was constructed, and it was confirmed that the identified motif and residues are prerequisites for the strong binding between P5-5 and 12F1. Altogether, the data of the present work could serve as a theoretic guide for the clinical design of biosynthetic drugs by artificial intelligence to treat sepsis. Full article
Show Figures

Figure 1

16 pages, 317 KiB  
Review
Combination Antibiotic Therapy for Orthopedic Infections
by Eric Bonnet and Julie Lourtet-Hascoët
Antibiotics 2025, 14(8), 761; https://doi.org/10.3390/antibiotics14080761 - 29 Jul 2025
Viewed by 302
Abstract
Background/Objectives: Limited robust data support the use of antibiotic combinations in the treatment of orthopedic infections. However, in certain situations, the combination of antibiotics seems to be beneficial. This review aims to outline the circumstances under which a combination of antibiotics may [...] Read more.
Background/Objectives: Limited robust data support the use of antibiotic combinations in the treatment of orthopedic infections. However, in certain situations, the combination of antibiotics seems to be beneficial. This review aims to outline the circumstances under which a combination of antibiotics may be utilized in the treatment of orthopedic infections. Methods: We reviewed the existing guidelines on orthopedic infections and focused on situations where antibiotic combinations are recommended or proposed optionally. We chose vitro and animal studies that provide evidence for the effectiveness of several widely recommended combinations. Results: The combinations serve multiple purposes: they provide empirical coverage while awaiting microbiological results, offer targeted treatment for difficult-to-treat infections, and facilitate oral treatment primarily for staphylococcal infections. The objectives include enhancing bacterial coverage against Gram-positive and Gram-negative bacteria, achieving synergistic effects with bactericidal agents, and reducing the risk of antibiotic resistance. The review outlines specific combinations for fracture-related infections, periprosthetic joint infections, spinal infections, and anterior cruciate ligament reconstruction infections, emphasizing the importance of tailoring antibiotic choices based on local epidemiology and patient history. The review also addresses potential drawbacks of combination therapy, such as toxicity, higher costs, and drug interactions, underscoring the complexity of managing orthopedic infections effectively. Conclusions: According to the guidelines, several different proposals are made, depending in part on the countries’ epidemiology. In a well-defined situation, various authors propose either monotherapy or a combination of antibiotics. When a combination is suggested, the choice of antibiotics is based on the expected effect: broadening the spectrum, enhancing bactericidal activity, achieving a synergistic effect, or reinforcing biofilm activity to optimize the treatment. Full article
(This article belongs to the Section Antibiotic Therapy in Infectious Diseases)
17 pages, 985 KiB  
Review
Advances in Forensic Entomotoxicology for Decomposed Corpses: A Review
by Sen Hou, Zengjia Liu, Jiali Su, Zeyu Yang, Zhongjiang Wang, Xinyi Yao, Zhou Lyu, Yang Xia, Shuguang Zhang, Wen Cui, Yequan Wang and Lipin Ren
Insects 2025, 16(7), 744; https://doi.org/10.3390/insects16070744 - 21 Jul 2025
Viewed by 462
Abstract
Forensic entomotoxicology is a subdiscipline that utilizes necrophagous insects as bioindicators for detecting drugs and toxicants in decomposed remains, particularly in cases where conventional biological matrices are no longer available. Toxic substances can profoundly alter insect development, physiology, and community succession, potentially impacting [...] Read more.
Forensic entomotoxicology is a subdiscipline that utilizes necrophagous insects as bioindicators for detecting drugs and toxicants in decomposed remains, particularly in cases where conventional biological matrices are no longer available. Toxic substances can profoundly alter insect development, physiology, and community succession, potentially impacting the accuracy of postmortem interval (PMI) estimation. This review systematically summarizes the effects of various xenobiotics, including pesticides, illicit drugs, sedatives, heavy metals, and antibiotics on larval growth, physiological traits, and gut microbial composition in forensically relevant flies. However, most studies to date have relied primarily on phenotypic observations, with limited insight into underlying molecular mechanisms. Significant interspecies and dose-dependent variability also exists in the absorption, metabolism, and physiological responses to xenobiotics. We highlight recent advances in multi-omics technologies that facilitate the identification of molecular biomarkers associated with xenobiotic exposure, particularly within the insect detoxification system. Key components such as cytochrome P450 monooxygenases (P450s), glutathione S-transferases (GSTs), and ATP-binding cassette (ABC) transporters play essential roles in xenobiotic metabolism and insecticide resistance. Additionally, the insect fat body serves as a central hub for detoxification, hormonal regulation, and energy metabolism. It integrates signals related to xenobiotic exposure and modulates larval development, making it a promising model for future mechanistic studies in insect toxicology. Altogether, this review offers a comprehensive and reliable framework for understanding the complex interactions between toxic substance exposure, insect ecology, and decomposition in forensic investigations. Full article
(This article belongs to the Section Medical and Livestock Entomology)
Show Figures

Graphical abstract

13 pages, 1243 KiB  
Review
Evidence-Based Medicine: Past, Present, Future
by Filippos Triposkiadis and Dirk L. Brutsaert
J. Clin. Med. 2025, 14(14), 5094; https://doi.org/10.3390/jcm14145094 - 17 Jul 2025
Viewed by 648
Abstract
Early medical traditions include those of ancient Babylonia, China, Egypt, and India. The roots of modern Western medicine, however, go back to ancient Greece. During the Renaissance, physicians increasingly relied on observation and experimentation to understand the human body and develop new techniques [...] Read more.
Early medical traditions include those of ancient Babylonia, China, Egypt, and India. The roots of modern Western medicine, however, go back to ancient Greece. During the Renaissance, physicians increasingly relied on observation and experimentation to understand the human body and develop new techniques for diagnosis and treatment. The discovery of antibiotics, antiseptics, and other drugs in the 19th century accelerated the development of modern medicine, the latter being fueled further by advances in technology, research, a better understanding of the human body, and, most recently, the introduction of evidence-based medicine (EBM). The EBM model de-emphasized intuition, unsystematic clinical experience, and pathophysiologic rationale as sufficient grounds for clinical decision-making and stressed the examination of evidence from clinical research. A later EBM model additionally incorporated clinical expertise and the latest model of EBM patients’ preferences and actions. In this review article, we argue that in the era of precision medicine, major EBM principles must be based on (a) the systematic identification, analysis, and utility of big data using artificial intelligence; (b) the magnifying effect of medical interventions by means of the physician–patient interaction, the latter being guided by the physician’s expertise, intuition, and philosophical beliefs; and (c) the patient preferences, since, in healthcare under precision medicine, the patient will be a central stakeholder contributing data and actively participating in shared decision-making. Full article
(This article belongs to the Section Clinical Research Methods)
Show Figures

Figure 1

7 pages, 806 KiB  
Communication
Two Cocrystals of Phenazine with Different Phenylboronic Acids
by Stijn Germonpré, Subhrajyoti Bhandary and Kristof Van Hecke
Molbank 2025, 2025(3), M2036; https://doi.org/10.3390/M2036 - 14 Jul 2025
Viewed by 376
Abstract
Boronic acids are an important class of molecules diversely used in organic synthesis, catalysis, medicinal chemistry, and for the design of functional materials. Particularly, aryl boronic acids in the solid state are known to exhibit pharmaceutical and photoluminescent properties for antimicrobial, sensing, and [...] Read more.
Boronic acids are an important class of molecules diversely used in organic synthesis, catalysis, medicinal chemistry, and for the design of functional materials. Particularly, aryl boronic acids in the solid state are known to exhibit pharmaceutical and photoluminescent properties for antimicrobial, sensing, and drug delivery applications. Furthermore, the phenazine molecule is known for its diverse pharmacological properties, including antibiotic activity. In the case of molecular crystalline solids, it is well established that understanding noncovalent interactions remains key to designing or engineering their functional properties. While both aryl boronic acids and phenazine molecules individually represent an important class of compounds, their co-assembly in the crystalline state is of interest within the context of supramolecular chemistry and crystal engineering. Herein, we report the supramolecular features of two newly synthesized cocrystals, which are composed of para-F/CF3-substituted phenylboronic acids, respectively, and phenazine, as demonstrated by structure analysis by single-crystal X-ray diffraction. Full article
Show Figures

Figure 1

24 pages, 7709 KiB  
Article
Quaternized Polysulfones as Matrix for the Development of Broad-Spectrum Antimicrobial Coatings for Medical Devices
by Oana Dumbrava, Irina Rosca, Daniela Ailincai and Luminita Marin
Polymers 2025, 17(13), 1869; https://doi.org/10.3390/polym17131869 - 3 Jul 2025
Viewed by 502
Abstract
The development and application of antimicrobial coatings has become increasingly important in both medical and industrial settings due to the rising threat of microbial contamination and antibiotic resistance. This paper focuses on the formulation, characterization, and investigation of coatings based on quaternized polysulfone, [...] Read more.
The development and application of antimicrobial coatings has become increasingly important in both medical and industrial settings due to the rising threat of microbial contamination and antibiotic resistance. This paper focuses on the formulation, characterization, and investigation of coatings based on quaternized polysulfone, which are designed to encapsulate two broad-spectrum antimicrobial drugs with complementary activity, amphotericin B (AmB) and norfloxacin (NFX), with the primary aim of inhibiting pathogen colonization on surgical instruments. Structural characterization using FTIR, 1H-NMR, and UV-Vis spectroscopy, along with supramolecular analysis via X-ray diffraction and polarized optical microscopy (POM), revealed strong physical interactions between the drugs and the quaternized polysulfone matrix. Scanning electron microscopy (SEM) confirmed a uniform distribution of the antimicrobial agents within the polymeric matrix. Surface wettability, assessed through water contact angle measurements, indicated moderate hydrophilicity (70–90°). The coatings also exhibited notable antioxidant activity, showing a 12-fold increase in DPPH radical inhibition compared to the control. Furthermore, all formulations demonstrated strong antimicrobial efficacy against three reference strains frequently associated with hospital-acquired infections, S. aureus, E. coli, and C. albicans, with inhibition zones ranging from 32 to 39.67 mm for bacterial strains and 13.86 to 20.86 mm for C. albicans. These data points indicate that these materials may be useful as antimicrobial coatings. Full article
(This article belongs to the Special Issue Polymer Coatings for High-Performance Applications)
Show Figures

Graphical abstract

26 pages, 905 KiB  
Review
Advancements in Antimicrobial Surface Coatings Using Metal/Metaloxide Nanoparticles, Antibiotics, and Phytochemicals
by Preetha Ebenezer, S. P. S. N. Buddhika Sampath Kumara, S. W. M. A. Ishantha Senevirathne, Laura J. Bray, Phurpa Wangchuk, Asha Mathew and Prasad K. D. V. Yarlagadda
Nanomaterials 2025, 15(13), 1023; https://doi.org/10.3390/nano15131023 - 1 Jul 2025
Viewed by 446
Abstract
The growing prevalence of bacterial infections and the alarming rise of antimicrobial resistance (AMR) have driven the need for innovative antimicrobial coatings for medical implants and biomaterials. However, implant surface properties, such as roughness, chemistry, and reactivity, critically influence biological interactions and must [...] Read more.
The growing prevalence of bacterial infections and the alarming rise of antimicrobial resistance (AMR) have driven the need for innovative antimicrobial coatings for medical implants and biomaterials. However, implant surface properties, such as roughness, chemistry, and reactivity, critically influence biological interactions and must be engineered to ensure biocompatibility, corrosion resistance, and sustained antibacterial activity. This review evaluates three principal categories of antimicrobial agents utilized in surface functionalization: metal/metaloxide nanoparticles, antibiotics, and phytochemical compounds. Metal/metaloxide-based coatings, especially those incorporating silver (Ag), zinc oxide (ZnO), and copper oxide (CuO), offer broad-spectrum antimicrobial efficacy through mechanisms such as reactive oxygen species (ROS) generation and bacterial membrane disruption, with a reduced risk of resistance development. Antibiotic-based coatings enable localized drug delivery but often face limitations related to burst release, cytotoxicity, and diminishing effectiveness against multidrug-resistant (MDR) strains. In contrast, phytochemical-derived coatings—using bioactive plant compounds such as curcumin, eugenol, and quercetin—present a promising, biocompatible, and sustainable alternative. These agents not only exhibit antimicrobial properties but also provide anti-inflammatory, antioxidant, and osteogenic benefits, making them multifunctional tools for implant surface modification. The integration of these antimicrobial strategies aims to reduce bacterial adhesion, inhibit biofilm formation, and enhance tissue regeneration. By leveraging the synergistic effects of metal/metaloxide nanoparticles, antibiotics, and phytochemicals, next-generation implant coatings hold the potential to significantly improve infection control and clinical outcomes in implant-based therapies. Full article
(This article belongs to the Special Issue Nanocoating for Antibacterial Applications)
Show Figures

Graphical abstract

22 pages, 6421 KiB  
Article
Therapeutic Optimization of Pseudomonas aeruginosa Phages: From Isolation to Directed Evolution
by Sara Bolognini, Caterina Ferretti, Claudia Campobasso, Elisabetta Trovato, Magda Marchetti, Laura Rindi, Arianna Tavanti and Mariagrazia Di Luca
Viruses 2025, 17(7), 938; https://doi.org/10.3390/v17070938 - 30 Jun 2025
Viewed by 504
Abstract
Pseudomonas aeruginosa is a major opportunistic pathogen with high levels of antibiotic resistance. Phage therapy represents a promising alternative for the treatment of difficult infections both alone and in combination with antibiotics. Here, we isolated and characterized three novel lytic myoviruses, Cisa, Nello, [...] Read more.
Pseudomonas aeruginosa is a major opportunistic pathogen with high levels of antibiotic resistance. Phage therapy represents a promising alternative for the treatment of difficult infections both alone and in combination with antibiotics. Here, we isolated and characterized three novel lytic myoviruses, Cisa, Nello, and Moonstruck. Genomic analysis revealed that Cisa and Nello belong to the Pbunavirus genus, while Moonstruck is a novel Pakpunavirus species. All lacked lysogeny, virulence, or resistance-associated genes, supporting their therapeutic suitability. Phage Nello and Moonstruck were active against P. aeruginosa Pa3GrPv, isolated from a patient with lung infection candidate for phage therapy. Moonstruck exhibited superior lytic activity with ciprofloxacin sub-MIC value (0.125 µg/mL), achieving bacterial suppression for 48 h. However, to improve the lytic efficacy of the phages on the clinical isolate, phage adaptation via serial passage was investigated. The killing efficacy of Nello was enhanced, whereas Moonstruck showed a less consistent improvement, suggesting phage-specific differences in evolutionary dynamics. Sequencing of the evolved phages revealed point mutations in tail-associated genes, potentially linked to a better phage–host interaction. These results support the use of phage–antibiotic combinations and directed evolution as strategies to enhance phage efficacy against drug-resistant infections. Overall, these findings support the therapeutic potential of the newly isolated phages in treating P. aeruginosa lung infections. Full article
(This article belongs to the Section Bacterial Viruses)
Show Figures

Figure 1

20 pages, 4236 KiB  
Article
Study of PVP and PLA Systems and Fibers Obtained by Solution Blow Spinning for Chlorhexidine Release
by Oliver Rosas, Manuel Acevedo and Itziar Vélaz
Polymers 2025, 17(13), 1839; https://doi.org/10.3390/polym17131839 - 30 Jun 2025
Viewed by 368
Abstract
Antimicrobial resistance arises from treatment non-adherence and ineffective delivery systems. Optimal wound dressings combine localized drug release, exudate management, and bacterial encapsulation through hydrogel-forming nanofibers for enhanced therapy. In this work, polylactic acid (PLA) and polyvinylpyrrolidone (PVP) fibers loaded with chlorhexidine (CHX) were [...] Read more.
Antimicrobial resistance arises from treatment non-adherence and ineffective delivery systems. Optimal wound dressings combine localized drug release, exudate management, and bacterial encapsulation through hydrogel-forming nanofibers for enhanced therapy. In this work, polylactic acid (PLA) and polyvinylpyrrolidone (PVP) fibers loaded with chlorhexidine (CHX) were developed using Solution Blow Spinning (SBS), a scalable electrospinning alternative that enables in situ deposition. Molecular interactions between CHX and polymers in solution (by UV-Vis and fluorescence spectroscopy) and in solid state (by FTIR, XRD and thermal analysis) were studied. The morphology of the polymeric fibers was determined by optical microscopy, showing that PVP fibers are thinner (1625 nm) and more uniform than those of PLA (2237 nm). Finally, drug release from single-polymer fibers discs, overlapping fibers discs (PLA/PVP/PLA and PVP/PLA/PVP), and solid dispersions was determined by UV-Vis spectrometry. PVP-based fibers exhibited faster CHX release due to their hydrophilic nature, while PLA fibers proved sustained release, attributed to their hydrophobic matrix. This study highlights the potential of PLA/PVP-CHX fibers made from SBS as advanced wound dressings, combining biocompatibility and personalized drug delivery, offering a promising platform for localized and controlled antibiotic delivery. Full article
Show Figures

Figure 1

14 pages, 578 KiB  
Article
Food–Drug Interactions: Effect of Propolis on the Pharmacokinetics of Enrofloxacin and Its Active Metabolite Ciprofloxacin in Rabbits
by Ali Sorucu, Cengiz Gokbulut, Busra Aslan Akyol and Osman Bulut
Pharmaceuticals 2025, 18(7), 967; https://doi.org/10.3390/ph18070967 - 27 Jun 2025
Viewed by 500
Abstract
Propolis is a natural resinous substance produced by honeybees that has many biological activities. For thousands of years, it has been widely used as a dietary supplement and traditional medicine to treat a variety of ailments due to its antimicrobial, anti-inflammatory, antioxidant, immunomodulatory, [...] Read more.
Propolis is a natural resinous substance produced by honeybees that has many biological activities. For thousands of years, it has been widely used as a dietary supplement and traditional medicine to treat a variety of ailments due to its antimicrobial, anti-inflammatory, antioxidant, immunomodulatory, and wound-healing properties. Nutritional supplements and foods may interact with drugs both pharmacodynamically and pharmacokinetically, which could raise clinical concerns. Background/Objectives: This study aimed to investigate the effect of propolis on the plasma disposition of enrofloxacin and to assess the potential pharmacokinetic interaction in rabbits. Methods: In this study, enrofloxacin was applied per os (20 mg/kg) and IM (10 mg/kg) and with propolis (100 mg resin/kg) administration in four groups of rabbits (each of six individuals). Heparinized blood samples were collected at 0, 0.1, 0.3, 0.5, 1, 2, 4, 8, 12, and 24 h post-administration. HPLC-FL was used to analyze the plasma concentrations of enrofloxacin and its active metabolite ciprofloxacin following liquid–liquid phase extraction, i.e., protein precipitation with acetonitrile and partitioning with sodium sulfate. Results: The results revealed that propolis coadministration significantly affected the plasma disposition of enrofloxacin and its active metabolite after both per os and intramuscular administration routes. Significantly greater AUC (48.91 ± 11.53 vs. 26.11 ± 12.44 µg.h/mL), as well as longer T1/2λz (11.75 ± 3.20 vs. 5.93 ± 2.51 h) and MRT (17.26 ± 4.55 vs. 8.96 ± 3.82 h) values of enrofloxacin and its metabolite ciprofloxacin, were observed after the coadministration of propolis compared to enrofloxacin alone following both per os and IM routes in rabbits. Conclusions: The concurrent use of propolis and prescription medications may prolong the half-life (T1/2λz) and increase the systemic availability of chronically used drugs with narrow therapeutic indices. The repeated use of drugs such as antibiotics, heart medications, and antidepressants, or drugs with a narrow therapeutic index such as antineoplastic and anticoagulant agents, can cause toxic effects by raising blood plasma levels. Considering the varied metabolism of rabbits and humans, further validation of this study may require thorough clinical trials in humans. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

36 pages, 2055 KiB  
Review
Towards Effective Helicobacter pylori Eradication: Emerging Therapies in the Wake of Antibiotic Resistance
by Barathan Muttiah, Wathiqah Wahid, Asif Sukri and Alfizah Hanafiah
Int. J. Mol. Sci. 2025, 26(13), 6064; https://doi.org/10.3390/ijms26136064 - 24 Jun 2025
Viewed by 1095
Abstract
Helicobacter pylori (H. pylori) infection is a leading cause of gastritis, peptic ulcers, and gastric cancer, affecting more than half of the global population. Its persistence in the acidic gastric environment and its ability to evade host immunity present major treatment challenges. [...] Read more.
Helicobacter pylori (H. pylori) infection is a leading cause of gastritis, peptic ulcers, and gastric cancer, affecting more than half of the global population. Its persistence in the acidic gastric environment and its ability to evade host immunity present major treatment challenges. Although antibiotics remain the standard therapy, rising antimicrobial resistance has reduced treatment efficacy, prompting the search for alternative and adjunct approaches. Emerging therapies include probiotics, antimicrobial peptides (AMPs), and plant-derived compounds, which target H. pylori through membrane disruption, immunomodulation, or direct antimicrobial activity. Novel drug delivery systems and microbiota-sparing interventions are also being investigated. Additionally, vaccine development offers a promising strategy for long-term protection, though challenges related to antigenic variability and host-specific responses remain. Despite these advances, treatment variability and the limited clinical validation of alternatives hinder progress. A multifaceted approach integrating microbiome research, host–pathogen interactions, and new therapeutic agents is essential for future success. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

16 pages, 637 KiB  
Review
Structural Innovations in Vancomycin: Overcoming Resistance and Expanding the Antibacterial Spectrum
by Ricardo Cartes-Velásquez, Felipe Morales-León, Franco Valdebenito-Maturana, Pablo Sáez-Riquelme, Nicolás Rodríguez-Ortíz and Hernán Carrillo-Bestagno
Organics 2025, 6(3), 28; https://doi.org/10.3390/org6030028 - 23 Jun 2025
Viewed by 854
Abstract
Vancomycin, a cornerstone antibiotic against severe Gram-positive infections, is increasingly challenged by resistance in Methicillin-resistant Staphylococcus aureus (MRSA) and Vancomycin Enterococcus spp. (VRE), necessitating the development of novel therapeutic strategies. This review examines how structural modifications to vancomycin can enhance its antibacterial activity [...] Read more.
Vancomycin, a cornerstone antibiotic against severe Gram-positive infections, is increasingly challenged by resistance in Methicillin-resistant Staphylococcus aureus (MRSA) and Vancomycin Enterococcus spp. (VRE), necessitating the development of novel therapeutic strategies. This review examines how structural modifications to vancomycin can enhance its antibacterial activity and explores the critical role of computational approaches in designing the next generation of analogs. By analyzing the existing literature, we highlight how strategic alterations, such as the introduction of lipophilic side chains, substitutions on the sugar moieties, and modifications to the aglycone core, have yielded derivatives with improved antibacterial potency. Notably, certain analogs (e.g., Vanc-83, Dipi-Van-Zn) have demonstrated expanded activity against Gram-negative bacteria and exhibited enhanced pharmacokinetic profiles, including prolonged half-lives and improved tissue penetration, crucial for effective treatment. Semisynthetic glycopeptides like telavancin, dalbavancin, and oritavancin exemplify successful translation of structural modifications, offering sustained plasma concentrations and simplified dosing regimens that improve patient compliance. Complementing these experimental efforts, computational methods, including molecular docking and molecular dynamics simulations, provide valuable insights into drug–target interactions, guiding the rational design of more effective analogs. Furthermore, physiologically based pharmacokinetic modeling aids in predicting the in vivo behavior and optimizing the pharmacokinetic properties of these novel compounds. This review highlights a critical path forward in the fight against multidrug-resistant infections. By meticulously examining the previously carried out structural refinement of vancomycin, guided by computational predictions and validated through rigorous experimental testing, we underscore its immense potential. Full article
Show Figures

Figure 1

20 pages, 1729 KiB  
Article
Development of a Cyclodextrin-Based Drug Delivery System to Improve the Physicochemical Properties of Ceftobiprole as a Model Antibiotic
by Dariusz Boczar, Wojciech Bocian, Jerzy Sitkowski, Karolina Pioruńska and Katarzyna Michalska
Int. J. Mol. Sci. 2025, 26(13), 5953; https://doi.org/10.3390/ijms26135953 - 20 Jun 2025
Viewed by 375
Abstract
This study presents a methodology for developing a cyclodextrin-based delivery system for ceftobiprole, a poorly water-soluble and amphoteric drug, chemically stable in acidic conditions. Ceftobiprole is a broad-spectrum cephalosporin antibiotic administered clinically as its water-soluble prodrug, ceftobiprole medocaril, due to limited aqueous solubility [...] Read more.
This study presents a methodology for developing a cyclodextrin-based delivery system for ceftobiprole, a poorly water-soluble and amphoteric drug, chemically stable in acidic conditions. Ceftobiprole is a broad-spectrum cephalosporin antibiotic administered clinically as its water-soluble prodrug, ceftobiprole medocaril, due to limited aqueous solubility of the parent compound. Solubility enhancement was achieved through complexation with anionic sulfobutylether-β-cyclodextrin (SBE-β-CD). At a pH below 3, ceftobiprole is protonated and cationic, which facilitates electrostatic interactions with the anionic cyclodextrin. An optimised high-performance liquid chromatography (HPLC) method was used to assess solubility, the impurity profile, and long-term chemical stability. X-ray powder diffraction (XRPD) confirmed the amorphous nature of the system and the absence of recrystallization. Nuclear magnetic resonance (NMR) and attenuated total reflection Fourier transform infrared (ATR-FTIR) spectroscopy supported the formation of a host–guest complex. The freeze-dried system prepared from 0.1 M formic acid solution contained negligible residual acid due to nearly complete sublimation. The most promising formulation was a ternary system of ceftobiprole, maleic acid, and SBE-β-CD (1:25:4 molar ratio), showing ~300-fold solubility improvement, low levels of degradation products, and stability after eight months at −20 °C. After pH adjustment to a parenterally acceptable level, the formulation demonstrated solubility and a pH comparable to the marketed drug product. Full article
(This article belongs to the Section Molecular Informatics)
Show Figures

Graphical abstract

Back to TopTop