Background: As a widely used chemotherapeutic agent, cisplatin frequently induces acute kidney injury (AKI), which severely compromises patient survival and limits its clinical use. While the natural flavonoid diosmetin (Dio) shows promise in mitigating cisplatin-induced nephrotoxicity, its clinical translation is challenged by poor solubility, low bioavailability, and incompletely elucidated mechanisms. This study aimed to overcome these limitations by developing a novel drug delivery system using the microalgae Dunaliella salina (D. salina, Ds) to load Dio (Ds-Dio), thereby enhancing its efficacy and exploring its therapeutic potential. Methods: We first characterized the physicochemical properties of Ds and Dio, and then Ds-Dio complex was synthesized via co-incubation. Its nephroprotective efficacy and safety were systematically evaluated in a cisplatin-induced mouse AKI model by assessing renal function (serum creatinine, blood urea nitrogen), injury biomarkers, histopathology, body weight, and organ index. The underlying mechanism was predicted by network pharmacology and subsequently validated experimentally. Results: The novel Ds-Dio delivery system has been successfully established. In the AKI model, Ds-Dio significantly improved renal function and exhibited a superior protective effect over Dio alone; this benefit is attributed to the enhanced bioavailability provided by Ds carrier. In addition, Ds-Dio also demonstrated safety performance, with no evidence of toxicity to major organs. Network pharmacology analysis predicted the involvement of PI3K/AKT pathway, which was experimentally verified. Specifically, we confirmed that Ds-Dio alleviates AKI by modulating the PI3K/AKT pathway, resulting in concurrent suppression of NF-κB-mediated inflammation and activation of NRF2-dependent antioxidant responses. Conclusions: This study successfully developed a microalgae-based drug delivery system, Ds-Dio, which significantly enhances the nephroprotective efficacy of Dio against cisplatin-induced AKI. The nephroprotective mechanism is associated with modulation of the PI3K/AKT pathway, resulting in the simultaneous attenuation of oxidative stress and inflammation.
{"title":"<i>Dunaliella salina</i>-Loaded Diosmetin Carriers Alleviate Oxidative Stress and Inflammation in Cisplatin-Induced Acute Kidney Injury via PI3K/AKT Pathway.","authors":"Yujing Huangfu, Wei Chen, Dandan Guo, Peiyao Wang, Aifang Li, Yi Yang, Shuxuan Li, Qianfang Wang, Baiyan Wang, Shuying Feng","doi":"10.3390/pharmaceutics18010102","DOIUrl":"https://doi.org/10.3390/pharmaceutics18010102","url":null,"abstract":"<p><p><b>Background:</b> As a widely used chemotherapeutic agent, cisplatin frequently induces acute kidney injury (AKI), which severely compromises patient survival and limits its clinical use. While the natural flavonoid diosmetin (Dio) shows promise in mitigating cisplatin-induced nephrotoxicity, its clinical translation is challenged by poor solubility, low bioavailability, and incompletely elucidated mechanisms. This study aimed to overcome these limitations by developing a novel drug delivery system using the microalgae <i>Dunaliella salina</i> (<i>D. salina</i>, <i>Ds</i>) to load Dio (<i>Ds</i>-Dio), thereby enhancing its efficacy and exploring its therapeutic potential. <b>Methods:</b> We first characterized the physicochemical properties of <i>Ds</i> and Dio, and then <i>Ds</i>-Dio complex was synthesized via co-incubation. Its nephroprotective efficacy and safety were systematically evaluated in a cisplatin-induced mouse AKI model by assessing renal function (serum creatinine, blood urea nitrogen), injury biomarkers, histopathology, body weight, and organ index. The underlying mechanism was predicted by network pharmacology and subsequently validated experimentally. <b>Results:</b> The novel <i>Ds</i>-Dio delivery system has been successfully established. In the AKI model, <i>Ds</i>-Dio significantly improved renal function and exhibited a superior protective effect over Dio alone; this benefit is attributed to the enhanced bioavailability provided by <i>Ds</i> carrier. In addition, <i>Ds</i>-Dio also demonstrated safety performance, with no evidence of toxicity to major organs. Network pharmacology analysis predicted the involvement of PI3K/AKT pathway, which was experimentally verified. Specifically, we confirmed that <i>Ds</i>-Dio alleviates AKI by modulating the PI3K/AKT pathway, resulting in concurrent suppression of NF-κB-mediated inflammation and activation of NRF2-dependent antioxidant responses. <b>Conclusions:</b> This study successfully developed a microalgae-based drug delivery system, <i>Ds</i>-Dio, which significantly enhances the nephroprotective efficacy of Dio against cisplatin-induced AKI. The nephroprotective mechanism is associated with modulation of the PI3K/AKT pathway, resulting in the simultaneous attenuation of oxidative stress and inflammation.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"18 1","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"146065496","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-12DOI: 10.3390/pharmaceutics18010099
Maša Roganović, Mirjana Cvetković, Ivana Gojković, Brankica Spasojević, Marija Jovanović, Branislava Miljković, Katarina Vučićević
Background/Objectives: Cyclosporine A (CsA) is a key immunosuppressant in post-transplantation therapy protocol characterized by large interindividual and intraindividual pharmacokinetic (PK) variability and a narrow therapeutic range necessitating therapeutic drug monitoring (TDM) to prevent graft rejection and minimize side effects. TDM data can be used for developing PK models with the objective of identification and quantification of variability factors that contribute to the differences in CsA concentrations. Methods: Retrospectively collected data from medical records of 58 patients (children and young adults) regarding CsA blood concentrations, concomitant medications, and laboratory findings of significance were used for the population PK model development in NONMEM® (version 7.5) with first-order conditional estimation method with interaction (FOCE-I). Simulation of the concentrations and area under the curve (AUC) was performed in the web application e-campsis®. RStudio (version 4.5.0) was used for the purpose of descriptive statistics analysis and graphs plotting. Results: A one-compartment model with first-order absorption and elimination best described the data. Value of clearance (CL/F) was estimated to be 15 L/h, and volume of distribution (V/F) was 71.1 L for a typical patient weighing 40 kg. Interindividual variability (IIV) on CL/F and V/F was 34.91% and 43.05%, respectively. Interoccasional variability (IOV) was 12.25%. Body weight (WT) was introduced allometrically on CL/F and V/F, with the estimated exponent of 0.89 for CL/F and 1 (fixed) for V/F. According to the final model, CL/F decreases with increasing haemoglobin (HGB) value. A difference of almost 22.5% in CL/F was observed among patients' HGB values reported in the study. Conclusions: Our findings indicate that HGB levels significantly influence CsA PK, particularly minimum concentration (Cmin), highlighting the importance of regular HGB levels monitoring together with CsA levels.
{"title":"Population Pharmacokinetics Model of Cyclosporin A in Children and Young Adult Renal Transplant Patients: Focus on Haemoglobin Contribution to Exposure Variability.","authors":"Maša Roganović, Mirjana Cvetković, Ivana Gojković, Brankica Spasojević, Marija Jovanović, Branislava Miljković, Katarina Vučićević","doi":"10.3390/pharmaceutics18010099","DOIUrl":"https://doi.org/10.3390/pharmaceutics18010099","url":null,"abstract":"<p><p><b>Background/Objectives</b>: Cyclosporine A (CsA) is a key immunosuppressant in post-transplantation therapy protocol characterized by large interindividual and intraindividual pharmacokinetic (PK) variability and a narrow therapeutic range necessitating therapeutic drug monitoring (TDM) to prevent graft rejection and minimize side effects. TDM data can be used for developing PK models with the objective of identification and quantification of variability factors that contribute to the differences in CsA concentrations. <b>Methods</b>: Retrospectively collected data from medical records of 58 patients (children and young adults) regarding CsA blood concentrations, concomitant medications, and laboratory findings of significance were used for the population PK model development in NONMEM<sup>®</sup> (version 7.5) with first-order conditional estimation method with interaction (FOCE-I). Simulation of the concentrations and area under the curve (AUC) was performed in the web application e-campsis<sup>®</sup>. RStudio (version 4.5.0) was used for the purpose of descriptive statistics analysis and graphs plotting. <b>Results</b>: A one-compartment model with first-order absorption and elimination best described the data. Value of clearance (CL/F) was estimated to be 15 L/h, and volume of distribution (V/F) was 71.1 L for a typical patient weighing 40 kg. Interindividual variability (IIV) on CL/F and V/F was 34.91% and 43.05%, respectively. Interoccasional variability (IOV) was 12.25%. Body weight (WT) was introduced allometrically on CL/F and V/F, with the estimated exponent of 0.89 for CL/F and 1 (fixed) for V/F. According to the final model, CL/F decreases with increasing haemoglobin (HGB) value. A difference of almost 22.5% in CL/F was observed among patients' HGB values reported in the study. <b>Conclusions</b>: Our findings indicate that HGB levels significantly influence CsA PK, particularly minimum concentration (C<sub>min</sub>), highlighting the importance of regular HGB levels monitoring together with CsA levels.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"18 1","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"146066108","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-12DOI: 10.3390/pharmaceutics18010100
María García-Hervalejo, José Germán Sánchez-Hernández, Irene Conde-González, Alejandro Avendaño Pita, María José Otero
Background: Teicoplanin is widely used for the empirical and targeted treatment of febrile neutropenia in patients with hematological malignancies. However, the pathophysiological alterations typical of this population may substantially affect drug exposure. The aim of this study was to develop and validate a population pharmacokinetic (PopPK) model of teicoplanin in adult hematological patients and to propose individualized dosing strategies. Methods: A retrospective, single-center study including 151 patients and 263 serum concentrations was conducted, with participants assigned to development (n = 100) and validation (n = 51) cohorts. Concentrations were quantified using a turbidimetric immunoassay, and the PopPK model was developed in NONMEM using FOCE-I. Results: Teicoplanin pharmacokinetics were described by a one-compartment model with first-order elimination. Ideal body weight, estimated glomerular filtration rate, and age were identified as significant predictors of clearance. Internal and external validation confirmed the robustness and predictive performance of the model. Monte Carlo simulations showed that conventional regimens (6 mg/kg every 12 h for three loading doses, followed by 6 mg/kg once-daily, or 600 mg every 12 h for three loading doses, followed by 600 mg once-daily) are insufficient to achieve therapeutic trough concentrations (≥15-20 mg/L) within the first 72 h, particularly in patients with preserved renal function or higher body weight. An intensified regimen consisting of five loading doses of 12 mg/kg every 12 h, followed by 12 mg/kg once-daily, enabled rapid attainment and maintenance of trough concentrations ≥ 20 mg/L in patients with lower to intermediate ideal body weight. Conclusions: These findings underscore the importance of intensified dosing strategies and covariate-guided individualization supported by therapeutic drug monitoring to achieve optimal teicoplanin exposure in this vulnerable patient group.
{"title":"Population Pharmacokinetics and Model-Informed Dose Optimization of Teicoplanin in Adults with Hematological Malignancies.","authors":"María García-Hervalejo, José Germán Sánchez-Hernández, Irene Conde-González, Alejandro Avendaño Pita, María José Otero","doi":"10.3390/pharmaceutics18010100","DOIUrl":"https://doi.org/10.3390/pharmaceutics18010100","url":null,"abstract":"<p><p><b>Background:</b> Teicoplanin is widely used for the empirical and targeted treatment of febrile neutropenia in patients with hematological malignancies. However, the pathophysiological alterations typical of this population may substantially affect drug exposure. The aim of this study was to develop and validate a population pharmacokinetic (PopPK) model of teicoplanin in adult hematological patients and to propose individualized dosing strategies. <b>Methods:</b> A retrospective, single-center study including 151 patients and 263 serum concentrations was conducted, with participants assigned to development (<i>n</i> = 100) and validation (<i>n</i> = 51) cohorts. Concentrations were quantified using a turbidimetric immunoassay, and the PopPK model was developed in NONMEM using FOCE-I. <b>Results:</b> Teicoplanin pharmacokinetics were described by a one-compartment model with first-order elimination. Ideal body weight, estimated glomerular filtration rate, and age were identified as significant predictors of clearance. Internal and external validation confirmed the robustness and predictive performance of the model. Monte Carlo simulations showed that conventional regimens (6 mg/kg every 12 h for three loading doses, followed by 6 mg/kg once-daily, or 600 mg every 12 h for three loading doses, followed by 600 mg once-daily) are insufficient to achieve therapeutic trough concentrations (≥15-20 mg/L) within the first 72 h, particularly in patients with preserved renal function or higher body weight. An intensified regimen consisting of five loading doses of 12 mg/kg every 12 h, followed by 12 mg/kg once-daily, enabled rapid attainment and maintenance of trough concentrations ≥ 20 mg/L in patients with lower to intermediate ideal body weight. <b>Conclusions:</b> These findings underscore the importance of intensified dosing strategies and covariate-guided individualization supported by therapeutic drug monitoring to achieve optimal teicoplanin exposure in this vulnerable patient group.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"18 1","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"146066072","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-12DOI: 10.3390/pharmaceutics18010103
Hayley Fong, Anjan Debnath
Objectives: Free-living amoeba Naegleria fowleri causes primary amoebic meningoencephalitis (PAM). While infection is rare, PAM's fatality rate exceeds 97%. The recommended treatment includes combination therapy, which does not result in uniform survival. Thus, there is a critical unmet need for finding better therapy for PAM. Drug repurposing can expedite the discovery of effective treatment for PAM. Isavuconazonium is approved for the treatment of fungal infections. Given that isavuconazole is the major metabolite of isavuconazonium and isavuconazole penetrates into the brain with high efficiency, our objective was to determine the activity of both isavuconazonium and isavuconazole on N. fowleri trophozoites. Methods: To test the effect of both compounds, we determined their dose-responses against N. fowleri and two mammalian cells. To establish how fast the prodrug and the metabolite kill the trophozoites, we measured potency at different time points. Finally, we investigated the effect of combining isavuconazonium or isavuconazole with amphotericin B on both N. fowleri and mammalian cells. Results: Both isavuconazonium and the metabolite isavuconazole were active against multiple strains, with clinically relevant isavuconazole exhibiting potency ranging between 0.1 and 0.6 µM. They were less toxic on mammalian cells. Isavuconazonium and isavuconazole required 24 h to achieve nanomolar potency. Combination with amphotericin B was synergistic without eliciting toxicity on mammalian cells. Conclusions: Our findings, together with the use of intravenous and oral formulations of isavuconazonium to treat pediatric and adult patients, support further in vivo efficacy study of isavuconazonium for its potential use for the treatment of PAM.
{"title":"The Activity of FDA-Approved Prodrug Isavuconazonium Sulfate and Its Major Metabolite Isavuconazole Against <i>Naegleria fowleri</i>.","authors":"Hayley Fong, Anjan Debnath","doi":"10.3390/pharmaceutics18010103","DOIUrl":"https://doi.org/10.3390/pharmaceutics18010103","url":null,"abstract":"<p><p><b>Objectives</b>: Free-living amoeba <i>Naegleria fowleri</i> causes primary amoebic meningoencephalitis (PAM). While infection is rare, PAM's fatality rate exceeds 97%. The recommended treatment includes combination therapy, which does not result in uniform survival. Thus, there is a critical unmet need for finding better therapy for PAM. Drug repurposing can expedite the discovery of effective treatment for PAM. Isavuconazonium is approved for the treatment of fungal infections. Given that isavuconazole is the major metabolite of isavuconazonium and isavuconazole penetrates into the brain with high efficiency, our objective was to determine the activity of both isavuconazonium and isavuconazole on <i>N. fowleri</i> trophozoites. <b>Methods</b>: To test the effect of both compounds, we determined their dose-responses against <i>N. fowleri</i> and two mammalian cells. To establish how fast the prodrug and the metabolite kill the trophozoites, we measured potency at different time points. Finally, we investigated the effect of combining isavuconazonium or isavuconazole with amphotericin B on both <i>N. fowleri</i> and mammalian cells. <b>Results</b>: Both isavuconazonium and the metabolite isavuconazole were active against multiple strains, with clinically relevant isavuconazole exhibiting potency ranging between 0.1 and 0.6 µM. They were less toxic on mammalian cells. Isavuconazonium and isavuconazole required 24 h to achieve nanomolar potency. Combination with amphotericin B was synergistic without eliciting toxicity on mammalian cells. <b>Conclusions</b>: Our findings, together with the use of intravenous and oral formulations of isavuconazonium to treat pediatric and adult patients, support further in vivo efficacy study of isavuconazonium for its potential use for the treatment of PAM.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"18 1","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"146065836","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Oral insulin improves compliance and convenience in patients with diabetes who require regular needle injections. However, the clinical application of oral insulin preparations has been limited due to instability and inefficient permeation through the gastrointestinal tract. In this study, a novel cationic polysaccharide nanodrug delivery platform was designed for efficient oral insulin delivery. Methods: The innovative thiolated trimethyl chitosan-grafted β-cyclodextrin (NCT) was synthesized by utilizing N-trimethyl chitosan (TMC) as the polymer backbone. This involved modifying TMC with thiol group-containing N-acetylcysteine and carboxymethyl-β-cyclodextrin that possesses hydrophobic cavities via an amide condensation reaction. Subsequently, this polymer was employed to construct the NCT nanoparticle system using an ionic cross-linking method. The physicochemical properties of the NCT nanoparticles were systematically analyzed, and their therapeutic efficacy was comprehensively evaluated in streptozotocin (STZ)-induced animal models. Results: The NCT nanoparticles demonstrated mucus adhesion, permeability, and pH sensitivity, which facilitated a slow and controlled release within the gastrointestinal microenvironment due to both ionic electrostatic interactions and disulfide bonding interactions. The experiments revealed in vivo that insulin/NCT nanoparticles extended the retention time of insulin in the small intestine. Blood glucose levels decreased to approximately 39% of the initial level at 5 h post-administration while exhibiting smooth hypoglycemic efficacy. Simultaneously, insulin bioavailability increased to 12.58%. Conclusions: The NCT nanoparticles effectively protect insulin from degradation in the gastrointestinal microenvironment while overcoming intestinal barriers, thereby providing a promising approach to oral biomolecule delivery.
{"title":"Efficient Oral Insulin Delivery Through Thiolated Trimethyl Chitosan-Grafted β-Cyclodextrin Nanoparticles.","authors":"Lizhen Yu, Fengge Wang, Shuyun Bao, Yue Zhang, Xuebin Shen, Desheng Wang, Zhisheng Liu, Xinyi Liu, Lihua Li, Renmin Gong","doi":"10.3390/pharmaceutics18010097","DOIUrl":"https://doi.org/10.3390/pharmaceutics18010097","url":null,"abstract":"<p><p><b>Background</b>: Oral insulin improves compliance and convenience in patients with diabetes who require regular needle injections. However, the clinical application of oral insulin preparations has been limited due to instability and inefficient permeation through the gastrointestinal tract. In this study, a novel cationic polysaccharide nanodrug delivery platform was designed for efficient oral insulin delivery. <b>Methods</b>: The innovative thiolated trimethyl chitosan-grafted β-cyclodextrin (NCT) was synthesized by utilizing N-trimethyl chitosan (TMC) as the polymer backbone. This involved modifying TMC with thiol group-containing N-acetylcysteine and carboxymethyl-β-cyclodextrin that possesses hydrophobic cavities via an amide condensation reaction. Subsequently, this polymer was employed to construct the NCT nanoparticle system using an ionic cross-linking method. The physicochemical properties of the NCT nanoparticles were systematically analyzed, and their therapeutic efficacy was comprehensively evaluated in streptozotocin (STZ)-induced animal models. <b>Results</b>: The NCT nanoparticles demonstrated mucus adhesion, permeability, and pH sensitivity, which facilitated a slow and controlled release within the gastrointestinal microenvironment due to both ionic electrostatic interactions and disulfide bonding interactions. The experiments revealed in vivo that insulin/NCT nanoparticles extended the retention time of insulin in the small intestine. Blood glucose levels decreased to approximately 39% of the initial level at 5 h post-administration while exhibiting smooth hypoglycemic efficacy. Simultaneously, insulin bioavailability increased to 12.58%. <b>Conclusions</b>: The NCT nanoparticles effectively protect insulin from degradation in the gastrointestinal microenvironment while overcoming intestinal barriers, thereby providing a promising approach to oral biomolecule delivery.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"18 1","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"146065882","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-12DOI: 10.3390/pharmaceutics18010096
Sari Risheq, Athira Venugopal, Andres Sancho, Michael Friedman, Irit Gati, Ron Eliashar, Doron Steinberg, Menachem Gross
Background: Non-absorbable polyvinyl alcohol sponges (Merocel) are widely used in otolaryngology for nasal and ear packing but are prone to bacterial colonization and biofilm formation, which may increase infection risk and drive frequent use of systemic antibiotics. Sustained-release drug delivery systems enable prolonged local antiseptic activity at the site of packing while minimizing systemic exposure. Methods: We developed a sustained-release varnish containing chlorhexidine (SRV-CHX) and coated sterile Merocel sponges. Antibacterial, in vitro, activity against Staphylococcus aureus and Pseudomonas aeruginosa was evaluated using kinetic diffusion assays on agar, optical density (OD600) measurements of planktonic cultures, drop plate, ATP-based viability assays, biofilm analysis by MTT metabolic assay, crystal violet bio-mass staining, high-resolution scanning electron microscopy (HR-SEM), and spinning disk confocal microscopy. Results: SRV-CHX-coated sponges produced sustained zones of inhibition on agar plates for up to 37 days against S. aureus and 39 days against P. aeruginosa, far exceeding the usual 3-5 days of clinical sponge use. Planktonic growth was significantly reduced compared with SRV-placebo, and a bactericidal effect persisted for up to 16 days for S. aureus and 5 days for P. aeruginosa before becoming predominantly bacteriostatic. Biofilm formation was markedly inhibited, with suppression of metabolic activity and biomass for at least 33 days for S. aureus and up to 16 days for P. aeruginosa. HR-SEM and confocal imaging confirmed sparse, discontinuous biofilms and predominance of non-viable bacteria on SRV-CHX-coated sponges compared with dense, viable biofilms on the placebo controls. Conclusions: Coating Merocel sponges with SRV-CHX provides prolonged antibacterial and anti-biofilm activity against clinically relevant pathogens. This strategy may reduce dependence on systemic antibiotics and improve infection control in nasal and ear packing applications in otolaryngology.
{"title":"Sustained Release Varnish of Chlorhexidine for Prevention of Biofilm Formation on Non-Absorbable Nasal and Ear Sponges.","authors":"Sari Risheq, Athira Venugopal, Andres Sancho, Michael Friedman, Irit Gati, Ron Eliashar, Doron Steinberg, Menachem Gross","doi":"10.3390/pharmaceutics18010096","DOIUrl":"https://doi.org/10.3390/pharmaceutics18010096","url":null,"abstract":"<p><p><b>Background</b>: Non-absorbable polyvinyl alcohol sponges (Merocel) are widely used in otolaryngology for nasal and ear packing but are prone to bacterial colonization and biofilm formation, which may increase infection risk and drive frequent use of systemic antibiotics. Sustained-release drug delivery systems enable prolonged local antiseptic activity at the site of packing while minimizing systemic exposure. <b>Methods</b>: We developed a sustained-release varnish containing chlorhexidine (SRV-CHX) and coated sterile Merocel sponges. Antibacterial, in vitro, activity against Staphylococcus aureus and Pseudomonas aeruginosa was evaluated using kinetic diffusion assays on agar, optical density (OD<sub>600</sub>) measurements of planktonic cultures, drop plate, ATP-based viability assays, biofilm analysis by MTT metabolic assay, crystal violet bio-mass staining, high-resolution scanning electron microscopy (HR-SEM), and spinning disk confocal microscopy. <b>Results</b>: SRV-CHX-coated sponges produced sustained zones of inhibition on agar plates for up to 37 days against <i>S. aureus</i> and 39 days against <i>P. aeruginosa</i>, far exceeding the usual 3-5 days of clinical sponge use. Planktonic growth was significantly reduced compared with SRV-placebo, and a bactericidal effect persisted for up to 16 days for S. aureus and 5 days for <i>P. aeruginosa</i> before becoming predominantly bacteriostatic. Biofilm formation was markedly inhibited, with suppression of metabolic activity and biomass for at least 33 days for <i>S. aureus</i> and up to 16 days for <i>P. aeruginosa</i>. HR-SEM and confocal imaging confirmed sparse, discontinuous biofilms and predominance of non-viable bacteria on SRV-CHX-coated sponges compared with dense, viable biofilms on the placebo controls. <b>Conclusions</b>: Coating Merocel sponges with SRV-CHX provides prolonged antibacterial and anti-biofilm activity against clinically relevant pathogens. This strategy may reduce dependence on systemic antibiotics and improve infection control in nasal and ear packing applications in otolaryngology.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"18 1","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"146065766","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-12DOI: 10.3390/pharmaceutics18010098
Jingjing Wang, Rui Ni, Ziwei Li, Jianhong Chen, Yao Liu
Background/Objectives: Wound healing represents a pervasive and urgent clinical challenge. Hard-to-heal chronic wounds are frequently complicated by infections, inflammatory responses, and oxidative stress. Currently, wound dressings are broadly categorized into dry and moist types, with moist wound dressings for chronic wounds accounting for approximately 70% of market revenue. Recently, adipose-derived stem cells (ADSCs), which possess self-renewal and multi-lineage differentiation capabilities, have emerged as a promising strategy for promoting tissue regeneration and wound repair. Methods: In this study, we developed a novel luteolin nanoparticle-ADSCs composite hydrogel (GelCA@LUT@ADSCs). This system was constructed by first encapsulating ADSCs within a chitosan/alginate hydrogel (GelCA), followed by coating the hydrogel with luteolin-loaded nanoparticles (LUT@NPs). Results: The sustained release of LUT@NPs from the hydrogel modulates the wound microenvironment, enhancing the pro-healing functions of ADSCs at the wound site. The GelCA hydrogel exhibited excellent biocompatibility. Both in vitro and in vivo results demonstrated that GelCA@LUT@ADSCs treatment effectively reduced inflammation, promoted angiogenesis and collagen deposition, stimulated cell proliferation and migration, and polarized macrophages toward an anti-inflammatory, pro-healing M2 phenotype, thereby accelerating wound healing. Conclusions: Overall, this innovative therapeutic approach provides a novel strategy for wound management through a synergistic division of labor between pharmaceutical agents and stem cells.
{"title":"A Multifunctional Hydrogel Incorporating Luteolin-Encapsulated ROS-Responsive Nanoparticles and Stem Cells Promotes Bacterial-Infected Wound Healing.","authors":"Jingjing Wang, Rui Ni, Ziwei Li, Jianhong Chen, Yao Liu","doi":"10.3390/pharmaceutics18010098","DOIUrl":"https://doi.org/10.3390/pharmaceutics18010098","url":null,"abstract":"<p><p><b>Background/Objectives:</b> Wound healing represents a pervasive and urgent clinical challenge. Hard-to-heal chronic wounds are frequently complicated by infections, inflammatory responses, and oxidative stress. Currently, wound dressings are broadly categorized into dry and moist types, with moist wound dressings for chronic wounds accounting for approximately 70% of market revenue. Recently, adipose-derived stem cells (ADSCs), which possess self-renewal and multi-lineage differentiation capabilities, have emerged as a promising strategy for promoting tissue regeneration and wound repair. <b>Methods:</b> In this study, we developed a novel luteolin nanoparticle-ADSCs composite hydrogel (GelCA@LUT@ADSCs). This system was constructed by first encapsulating ADSCs within a chitosan/alginate hydrogel (GelCA), followed by coating the hydrogel with luteolin-loaded nanoparticles (LUT@NPs). <b>Results:</b> The sustained release of LUT@NPs from the hydrogel modulates the wound microenvironment, enhancing the pro-healing functions of ADSCs at the wound site. The GelCA hydrogel exhibited excellent biocompatibility. Both in vitro and in vivo results demonstrated that GelCA@LUT@ADSCs treatment effectively reduced inflammation, promoted angiogenesis and collagen deposition, stimulated cell proliferation and migration, and polarized macrophages toward an anti-inflammatory, pro-healing M<sub>2</sub> phenotype, thereby accelerating wound healing. <b>Conclusions:</b> Overall, this innovative therapeutic approach provides a novel strategy for wound management through a synergistic division of labor between pharmaceutical agents and stem cells.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"18 1","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"146065549","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-11DOI: 10.3390/pharmaceutics18010095
Byeol Yoon, Jang Ah Kim, Yoo Kyung Kang
Antimicrobial resistance (AMR) is escalating worldwide, posing a serious threat to global public health by driving infections that are no longer treatable with conventional antibiotics. CRISPR-Cas technology offers a programmable and highly specific therapeutic alternative by directly targeting the genetic determinants responsible for resistance. Various CRISPR systems can restore antibiotic susceptibility and induce selective bactericidal effects by eliminating resistance genes, disrupting biofilm formation, and inhibiting virulence pathways. Moreover, CRISPR can suppress horizontal gene transfer (HGT) by removing mobile genetic elements such as plasmids, thereby limiting the ecological spread of AMR across humans, animals, and the environment. Advances in delivery platforms-including conjugative plasmids, phagemids, and nanoparticle-based carriers-are expanding the translational potential of CRISPR-based antimicrobial strategies. Concurrent progress in Cas protein engineering, spatiotemporal activity regulation, and AI-driven optimization is expected to overcome current technical barriers. Collectively, these developments position CRISPR-based antimicrobials as next-generation precision therapeutics capable of treating refractory bacterial infections while simultaneously suppressing the dissemination of antibiotic resistance.
{"title":"CRISPR-Cas-Mediated Reprogramming Strategies to Overcome Antimicrobial Resistance.","authors":"Byeol Yoon, Jang Ah Kim, Yoo Kyung Kang","doi":"10.3390/pharmaceutics18010095","DOIUrl":"https://doi.org/10.3390/pharmaceutics18010095","url":null,"abstract":"<p><p>Antimicrobial resistance (AMR) is escalating worldwide, posing a serious threat to global public health by driving infections that are no longer treatable with conventional antibiotics. CRISPR-Cas technology offers a programmable and highly specific therapeutic alternative by directly targeting the genetic determinants responsible for resistance. Various CRISPR systems can restore antibiotic susceptibility and induce selective bactericidal effects by eliminating resistance genes, disrupting biofilm formation, and inhibiting virulence pathways. Moreover, CRISPR can suppress horizontal gene transfer (HGT) by removing mobile genetic elements such as plasmids, thereby limiting the ecological spread of AMR across humans, animals, and the environment. Advances in delivery platforms-including conjugative plasmids, phagemids, and nanoparticle-based carriers-are expanding the translational potential of CRISPR-based antimicrobial strategies. Concurrent progress in Cas protein engineering, spatiotemporal activity regulation, and AI-driven optimization is expected to overcome current technical barriers. Collectively, these developments position CRISPR-based antimicrobials as next-generation precision therapeutics capable of treating refractory bacterial infections while simultaneously suppressing the dissemination of antibiotic resistance.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"18 1","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"146065932","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Diabetic nephropathy (DN) is largely driven by transforming growth factor-β1 (TGF-β1)-mediated fibrosis. Clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) ribonucleoprotein (RNP) complexes offer precise gene disruption, yet effective non-viral delivery remains a challenge. This study developed cationic lipid-based hybrid nanostructured lipid carriers (NLCs) for intracellular delivery of TGFB1-targeting RNP as an early-stage platform for DN gene modulation. Methods: A single-guide RNA (sgRNA) targeting human TGFB1 was assembled with Cas9 protein (1:1 and 1:2 molar ratios). Hybrid NLCs comprising squalene, glyceryl trimyristate, and the cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) were formulated via optimized emulsification-sonication to achieve sub-100 nm particles. Physicochemical properties, including polydispersity index (PDI), were assessed via dynamic light scattering (DLS), while silencing efficacy in HEK293T cells was quantified using quantitative reverse transcription PCR (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). Results: Optimized NLCs achieved hydrodynamic diameters of 65-99 nm (PDI < 0.5) with successful RNP complexation. The 1:2 Cas9:sgRNA formulation produced the strongest gene-editing response, reducing TGFB1 mRNA by 67% (p < 0.01) compared with 39% for the 1:1 ratio. This translated to a significant reduction in TGF-β1 protein (p < 0.05) within 24 h. Conclusions: DOTAP-based hybrid NLCs enable efficient delivery of CRISPR-Cas9 RNP and achieve significant suppression of TGFB1 expression at both transcriptional and protein levels. These findings establish a promising non-viral platform for upstream modulation of profibrotic signaling in DN and support further evaluation in kidney-derived cells and in vivo renal models.
{"title":"DOTAP-Based Hybrid Nanostructured Lipid Carriers for CRISPR-Cas9 RNP Delivery Targeting <i>TGFB1</i> in Diabetic Nephropathy.","authors":"Nurul Jummah, Hanifa Syifa Kamila, Satrialdi, Aluicia Anita Artarini, Ebrahim Sadaqa, Anindyajati, Diky Mudhakir","doi":"10.3390/pharmaceutics18010094","DOIUrl":"https://doi.org/10.3390/pharmaceutics18010094","url":null,"abstract":"<p><p><b>Background</b>: Diabetic nephropathy (DN) is largely driven by transforming growth factor-β1 (TGF-β1)-mediated fibrosis. Clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) ribonucleoprotein (RNP) complexes offer precise gene disruption, yet effective non-viral delivery remains a challenge. This study developed cationic lipid-based hybrid nanostructured lipid carriers (NLCs) for intracellular delivery of <i>TGFB1</i>-targeting RNP as an early-stage platform for DN gene modulation. <b>Methods</b>: A single-guide RNA (sgRNA) targeting human <i>TGFB1</i> was assembled with Cas9 protein (1:1 and 1:2 molar ratios). Hybrid NLCs comprising squalene, glyceryl trimyristate, and the cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) were formulated via optimized emulsification-sonication to achieve sub-100 nm particles. Physicochemical properties, including polydispersity index (PDI), were assessed via dynamic light scattering (DLS), while silencing efficacy in HEK293T cells was quantified using quantitative reverse transcription PCR (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). <b>Results</b>: Optimized NLCs achieved hydrodynamic diameters of 65-99 nm (PDI < 0.5) with successful RNP complexation. The 1:2 Cas9:sgRNA formulation produced the strongest gene-editing response, reducing <i>TGFB1</i> mRNA by 67% (<i>p</i> < 0.01) compared with 39% for the 1:1 ratio. This translated to a significant reduction in TGF-β1 protein (<i>p</i> < 0.05) within 24 h. <b>Conclusions</b>: DOTAP-based hybrid NLCs enable efficient delivery of CRISPR-Cas9 RNP and achieve significant suppression of <i>TGFB1</i> expression at both transcriptional and protein levels. These findings establish a promising non-viral platform for upstream modulation of profibrotic signaling in DN and support further evaluation in kidney-derived cells and in vivo renal models.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"18 1","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"146065914","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-10DOI: 10.3390/pharmaceutics18010091
Ilaria Trozzi, Beatrice Giorgi, Riccardo De Paola, Milo Gatti, Federico Pea
Background/Objectives: Therapeutic drug monitoring (TDM) of β-lactams (BL), BL/β-lactamase inhibitor (BLI) combinations (BL/BLIc), and of fosfomycin may play a key role in optimizing antimicrobial therapy and in preventing resistance development, especially when used by continuous infusion in critically ill or immunocompromised patients. Unfortunately, analytical methods for simultaneously quantifying multiple BL/BLIc in plasma are still lacking. Methods: The aim of this study was to develop and validate two rapid, sensitive, and accurate UPLC-qTOF-MS/MS methods for the simultaneous quantification of five novel β-lactam or β-lactam/β-lactamase inhibitor combinations (ceftolozane/tazobactam, ceftazidime/avibactam, meropenem/vaborbactam, cefiderocol, and ceftobiprole) along with fosfomycin. Methods: Human plasma samples were prepared by protein precipitation using methanol containing isotopically labeled internal standards. Chromatographic separation was achieved within 10-12 min using two Agilent Poroshell columns (EC-C18 and PFP) under positive and negative electrospray ionization modes. The method was validated according to the EMA guidelines by assessing selectivity, linearity, precision, accuracy, matrix effects, extraction recovery, and stability. Results: The methods exhibited excellent linearity (R2 ≥ 0.998) across the calibration ranges for all of the analytes (1.56-500 µg/mL), with limits of quantification ranging from 1.56 to 15.62 µg/mL. Intra- and inter-day precision and accuracy were always within ±15%. Extraction recovery always exceeded 92%, and the matrix effects were effectively corrected through isotopic internal standards. No carry-over or isobaric interferences were observed. All the analytes were stable for up to five days at 4 °C, but the BL and BL/BLIc stability was affected by multiple freeze-thaw cycles. Conclusions: These UPLC-qTOF-MS/MS multi-analyte methods enabled a simultaneous, reliable quantification in plasma of five novel beta-lactams and of fosfomycin. Robustness, high throughput, and sensitivity make these multi-methods feasible for real-time TDM, supporting personalized antimicrobial dosing and improved therapeutic outcomes in patients with severe or multidrug-resistant infections.
{"title":"Two Comprehensive Liquid Chromatography High-Resolution Mass Spectrometry (UPLC-MS/MS) Multi-Methods for Real-Time Therapeutic Drug Monitoring (TDM) of Five Novel Beta-Lactams and of Fosfomycin Administered by Continuous Infusion.","authors":"Ilaria Trozzi, Beatrice Giorgi, Riccardo De Paola, Milo Gatti, Federico Pea","doi":"10.3390/pharmaceutics18010091","DOIUrl":"https://doi.org/10.3390/pharmaceutics18010091","url":null,"abstract":"<p><p><b>Background/Objectives</b>: Therapeutic drug monitoring (TDM) of β-lactams (BL), BL/β-lactamase inhibitor (BLI) combinations (BL/BLIc), and of fosfomycin may play a key role in optimizing antimicrobial therapy and in preventing resistance development, especially when used by continuous infusion in critically ill or immunocompromised patients. Unfortunately, analytical methods for simultaneously quantifying multiple BL/BLIc in plasma are still lacking. <b>Methods</b>: The aim of this study was to develop and validate two rapid, sensitive, and accurate UPLC-qTOF-MS/MS methods for the simultaneous quantification of five novel β-lactam or β-lactam/β-lactamase inhibitor combinations (ceftolozane/tazobactam, ceftazidime/avibactam, meropenem/vaborbactam, cefiderocol, and ceftobiprole) along with fosfomycin. Methods: Human plasma samples were prepared by protein precipitation using methanol containing isotopically labeled internal standards. Chromatographic separation was achieved within 10-12 min using two Agilent Poroshell columns (EC-C18 and PFP) under positive and negative electrospray ionization modes. The method was validated according to the EMA guidelines by assessing selectivity, linearity, precision, accuracy, matrix effects, extraction recovery, and stability. <b>Results</b>: The methods exhibited excellent linearity (R<sup>2</sup> ≥ 0.998) across the calibration ranges for all of the analytes (1.56-500 µg/mL), with limits of quantification ranging from 1.56 to 15.62 µg/mL. Intra- and inter-day precision and accuracy were always within ±15%. Extraction recovery always exceeded 92%, and the matrix effects were effectively corrected through isotopic internal standards. No carry-over or isobaric interferences were observed. All the analytes were stable for up to five days at 4 °C, but the BL and BL/BLIc stability was affected by multiple freeze-thaw cycles. <b>Conclusions</b>: These UPLC-qTOF-MS/MS multi-analyte methods enabled a simultaneous, reliable quantification in plasma of five novel beta-lactams and of fosfomycin. Robustness, high throughput, and sensitivity make these multi-methods feasible for real-time TDM, supporting personalized antimicrobial dosing and improved therapeutic outcomes in patients with severe or multidrug-resistant infections.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"18 1","pages":""},"PeriodicalIF":5.5,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"146065849","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}