[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (6,975)

Search Parameters:
Keywords = drug dosing

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 7657 KiB  
Article
Optimization and Evaluation of Cannabis-Based Magistral Formulations: A Path to Personalized Therapy
by Bożena Grimling, Magdalena Fast, Magdalena Okoniewska, Artur Owczarek and Bożena Karolewicz
Pharmaceuticals 2025, 18(1), 73; https://doi.org/10.3390/ph18010073 - 9 Jan 2025
Viewed by 269
Abstract
Introduction: The official implementation of pharmaceutical-grade cannabis raw materials for medicinal use has permitted doctors to prescribe and pharmacists to prepare cannabis-based formulations. The objective of the pharmaceutical development and manufacturing process optimization work was to propose a suppository formulation containing doses of [...] Read more.
Introduction: The official implementation of pharmaceutical-grade cannabis raw materials for medicinal use has permitted doctors to prescribe and pharmacists to prepare cannabis-based formulations. The objective of the pharmaceutical development and manufacturing process optimization work was to propose a suppository formulation containing doses of 25 mg and 50 mg of tetra-hydrocannabinol (∆-9-THC) as an alternative to existing inhalable or orally administered formulations. The formulation could be used for rectal or vaginal administration, thereby providing dosage control in the treatment of endometriosis and other conditions involving pain. In this study, two substrates from suppositories with standardized Cannabis extractum normatum (CEX) were used: cocoa butter and Witepsol® H15. Materials and Methods: The long-term stability of CEX was investigated over a period of up to 24 months. The concentrations of ∆-9-THC, cannabidiol (CBD), and cannabinol (CBN) were determined using an HPLC method. Furthermore, the water content of the extract, the ethanol residue, and the microbiological purity were determined. The pharmaceutical properties of CEX-incorporated suppositories, namely content uniformity, hardness, softening time, total deformation time, disintegration time, and the release profile of ∆-9-THC, CBD, and CBN, were evaluated in order to develop optimal preparation procedures for pharmacists. Results and Discussion: Following a 24-month stability study on CEX, no significant alterations in component content were observed beyond the specified requirements. The disintegration time, total deformation time, and hardness of the suppositories based on Witepsol® H15 with CEX were found to be longer and higher, respectively, than those of suppositories formulated with cocoa butter. In vitro studies demonstrated that suppositories prepared with Witepsol® H15 exhibited superior release of ∆-9-THC compared to those prepared with cocoa butter. Conclusions: We suggest that pharmacists making prescription drugs in a pharmacy setting in the form of medical marijuana suppositories will receive a better release profile of the drug by choosing Witepsol® H15 as a substrate. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Appearances of suppository formulations with <span class="html-italic">Cannabis extractum normatum</span> prepared based on cocoa butter and Witepsol<sup>®</sup> H15: (<b>a</b>) CB; (<b>b</b>) CB_EXT_25; (<b>c</b>) CB_EXT_50; (<b>d</b>) W; (<b>e</b>) W_EXT_25; (<b>f</b>) W_EXT_50.</p>
Full article ">Figure 2
<p>In vitro ∆-9-THC release profiles from suppositories. Each data point represents mean ± SD, n = 3.</p>
Full article ">Figure 3
<p>Comparing THC dissolution efficiency (DE) from suppository formulations using parametric one-way ANOVA.</p>
Full article ">Figure 4
<p>In vitro CBD release profiles from suppositories. Each data point represents mean ± SD, n = 3.</p>
Full article ">Figure 5
<p>In vitro CBN release profiles from suppositories. Each data point represents mean ± SD, n = 3.</p>
Full article ">Figure 6
<p>Scheme of technology preparation of suppositories with <span class="html-italic">Cannabis extractum normatum</span> on fatty bases: (<b>a</b>) cocoa butter; (<b>b</b>) Witepsol<sup>®</sup> H15. Created with Biorender.com.</p>
Full article ">
20 pages, 4162 KiB  
Review
Polyphenolic Compounds in Fabaceous Plants with Antidiabetic Potential
by Lucia Guerrero-Becerra, Sumiko Morimoto, Estefania Arrellano-Ordoñez, Angélica Morales-Miranda, Ramón G. Guevara-Gonzalez, Ana Angélica Feregrino-Pérez and Consuelo Lomas-Soria
Pharmaceuticals 2025, 18(1), 69; https://doi.org/10.3390/ph18010069 - 9 Jan 2025
Viewed by 210
Abstract
Diabetes mellitus (DM) is a chronic non-communicable disease with an increasing prevalence in Latin America and worldwide, impacting various social and economic areas. It causes numerous complications for those affected. Current treatments for diabetes include oral hypoglycemic drugs, which can lead to adverse [...] Read more.
Diabetes mellitus (DM) is a chronic non-communicable disease with an increasing prevalence in Latin America and worldwide, impacting various social and economic areas. It causes numerous complications for those affected. Current treatments for diabetes include oral hypoglycemic drugs, which can lead to adverse effects and health complications. Other natural alternatives for DM treatment have been studied as adjunct therapies that could reduce or eliminate the need for antidiabetic medications. Several natural supplements may offer an alternative way to improve the quality of life for patients with DM, and they may have other nutraceutical applications. Due to their phenolic compound content, some leguminous substances have been proposed as these alternatives. Phenolic compounds, with their high antioxidant activity, have shown promising potential in insulin synthesis, secretion, and the functionality of the endocrine pancreas. This review provides valuable information on various leguminous plants with anti-diabetic properties, including antioxidant, hypoglycemic, anti-fat-induced damage, and anti-apoptotic properties in vitro and in vivo, attributed to the high content of phenolic compounds in their seeds. Natural products with antidiabetic and pharmacological treatment potential improve diabetes management by offering more effective and complementary alternatives. To integrate these herbal remedies into modern medicine, further research on phenolic compound type, doses, efficacy, and safety in the human population is needed. Full article
(This article belongs to the Special Issue Natural Products in Diabetes Mellitus: 2nd Edition)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Secondary metabolite production in plants (N = nitrogen, S = sulfur) (created by BioRender).</p>
Full article ">Figure 2
<p>General diagram for the identification and selection of articles (created by BioRender).</p>
Full article ">Figure 3
<p>The process of obtaining and developing phytotherapeutics from fabaceous plants (created by BioRender).</p>
Full article ">Figure 4
<p>Potential mechanisms of action of phenolic compounds [<a href="#B93-pharmaceuticals-18-00069" class="html-bibr">93</a>,<a href="#B95-pharmaceuticals-18-00069" class="html-bibr">95</a>] (created by BioRender).</p>
Full article ">
18 pages, 3905 KiB  
Article
Methyl Canthin-6-one-2-carboxylate Inhibits the Activation of the NLRP3 Inflammasome in Synovial Macrophages by Upregulating Nrf2 Expression
by Yuanyuan Chen, Zongying Zhang, Yuan Yao, Xiaorong Zhou, Yong Ling, Liming Mao and Zhifeng Gu
Curr. Issues Mol. Biol. 2025, 47(1), 38; https://doi.org/10.3390/cimb47010038 - 9 Jan 2025
Viewed by 194
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder that leads to severe cartilage deterioration and synovial impairment in the joints. Previous studies have indicated that the aberrant activation of the NLRP3 inflammasome in synovial macrophages plays a significant role in the pathogenesis of RA [...] Read more.
Rheumatoid arthritis (RA) is an autoimmune disorder that leads to severe cartilage deterioration and synovial impairment in the joints. Previous studies have indicated that the aberrant activation of the NLRP3 inflammasome in synovial macrophages plays a significant role in the pathogenesis of RA and has been regarded as a therapeutic target for the disease. In this study, we synthesized a novel canthin-6-one alkaloid, namely methyl canthin-6-one-2-carboxylate (Cant), and assessed its effects on NLRP3 inflammasome activation in macrophages. Our data reveal that exposure to Cant significantly suppressed the transcription and secretion of multiple pro-inflammatory mediators, including IL-1β, IL-6, IL-18, TNF-α, NO, and COX2, in a dose-dependent manner. These alterations were associated with changes in the activation of various signaling pathways, including NF-kB, MAPK, and PI3K-AKT pathways. Notably, pretreatment with Cant significantly reduced LPS/ATP-induced activation of the NLRP3 inflammasome, as evidenced by the decline in the cleaved forms of IL-1β and caspase-1 in cell culture supernatants of BMDMs. Regarding the mechanisms, our data show that Cant could enhance the expression of Nrf2 in macrophages, which play an inhibitory role in ROS production. Collectively, our data demonstrate that Cant might suppress the activation of the NLRP3 inflammasome by upregulating the production of Nrf2, suggesting that Cant could serve as a candidate for the further development of anti-RA drugs. Full article
(This article belongs to the Special Issue The Role of Bioactives in Inflammation)
Show Figures

Figure 1

Figure 1
<p>The chemical structure of methyl canthin-6-one-2-carboxylate (Cant).</p>
Full article ">Figure 2
<p>Pharmacological analysis indicates a potential anti-RA role of Cant. (<b>A</b>) Cant target genes were searched using SwissTargetPrediction and SuperPred databases and were intersected with RA-related genes. (<b>B</b>) A Cant–RA–target network was drawn using Cytoscape 3.10.2 software. (<b>C</b>) The PPI network was established to visualize the interactions between the targets. (<b>D</b>) GO pathway enrichment analysis identified the enriched terms of BPs, CCs, and MFs associated with Cant targets. (<b>E</b>) KEGG pathway analysis identified the enriched signaling pathways associated with Cant target genes. (<b>F</b>) Mouse BMDM cells were exposed to various concentrations of Cant; then, the cell viability were examined using CKK8 assay at indicated time points. (<b>G</b>) BMDM cells were exposed to various concentrations of Cant; then, the cells were stimulated with LPS, and the cell viability was detected using a CKK8 assay at indicated time points. Data in (<b>F</b>,<b>G</b>) were presented as means ± SD (<span class="html-italic">n</span> = 3) (* = <span class="html-italic">p</span> &lt; 0.05, ** = <span class="html-italic">p</span> &lt; 0.01, ns = not significant, vs. control group).</p>
Full article ">Figure 3
<p>Cant inhibits the production of pro-inflammatory mediators. (<b>A</b>) Mouse BMDM cells were exposed to indicated concentrations of Cant and then were stimulated with LPS; the culture supernatants were collected for detection of IL-6 and TNF-α by ELISA. (<b>B</b>) BMDM cells were treated as indicated in (<b>A</b>); the cells were lysed and subjected to Western blot detection of iNOS, pro-IL-1β, and COX-2; GAPDH was used as an internal control. (<b>C</b>) BMDM cells were treated as indicated in (<b>A</b>); the cells were subjected to RNA extraction and subsequent detection of transcription of TNF-α, IL-6, IL-18, and COX-2 by RT-PCR. RAW264.7 cells were used to validate the results obtained using BMDMs. (<b>D</b>) IL-6 and TNF-α levels in culture supernatants of the cells were detected using ELISA. (<b>E</b>) iNOS, pro-IL-1β, and COX-2, GAPDH levels in cell lysates were detected using Western blot. (<b>F</b>) The transcription of TNF-α, IL-6, IL-18, and COX-2 was detected using RT-PCR. The data in (<b>A</b>,<b>C</b>,<b>D</b>,<b>F</b>) are presented as means ± SD (<span class="html-italic">n</span> = 3) (ns = not significant, *, <span class="html-italic">p</span> &lt; 0.05, ***, <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, vs. LPS group).</p>
Full article ">Figure 4
<p>Cant inhibits the NF-kB and MAPK signaling pathways. (<b>A</b>) BMDM cells were pretreated with indicated concentrations of Cant; the cells were then stimulated with LPS for 2 h. Then, the cells were lysed, and the cell lysates were subjected to detection of phospho-p65, p65, phospho-p38, p38, phospho-ERK, ERK, phospho-JNK, and JNK using Western blot analysis. GAPDH was used as an internal control. (<b>B</b>) Quantification of relative levels of phosphorylated p65, p38, ERK, and JNK in comparison to the non-phosphorylated proteins in (<b>A</b>). The same stimulating method was applied to RAW264.7 cells, and the same proteins were detected using Western blot (<b>C</b>). The quantified data are shown in (<b>D</b>). (<b>E</b>) The BMDM lysates were subjected to detection of AKT, phospho-AKT, PI3K, and phospho-PI3K using Western blot analysis. GAPDH was used as an internal control. (<b>F</b>) Quantification of relative level of phosphorylated AKT and PI3K in comparison to the non-phosphorylated proteins in (<b>E</b>). The data in (<b>A</b>,<b>C</b>,<b>E</b>) are representative of three independent experiments. Data in (<b>B</b>,<b>D</b>,<b>F</b>) are presented as means ± SD. * = <span class="html-italic">p</span> &lt; 0.05; ** = <span class="html-italic">p</span> &lt; 0.01; *** = <span class="html-italic">p</span> &lt; 0.001; **** = <span class="html-italic">p</span> &lt; 0.0001. One-way analysis of variance was used. ns = not significant.</p>
Full article ">Figure 5
<p>Cant suppresses the activation of the NLRP3 inflammasome by promoting the level of Nrf2 and inhibiting ROS production. BMDMs were stimulated with LPS (1 μg/mL) for 6 h in serum-free culture medium and were then treated with various concentrations of Cant. The cells were then stimulated with ATP (5 mM) for 20 min to activate the NLRP3 inflammasome. The total proteins in the cell culture supernatants were precipitated and applied in Western blot analysis of caspase-1 p20 and IL-1β p17. The cells were harvested and lysed, the cell lysates were subjected to Western blot analysis of pro-caspase-1, pro-IL-1β, and NLRP3. GAPDH was used as an internal control. (<b>B</b>) BMDMs were stimulated as indicated in (<b>A</b>) and then the cells were subjected to ROS detection using DCFH-DA staining using flow cytometry. (<b>C</b>, left) The cell lysates obtained in (<b>A</b>) were used in the detection of NLRP3 and Nrf2 using Western blot. The quantification of the expression levels of NLRP3 (<b>C</b>, middle) and Nrf2 (<b>C</b>, right) are shown. The data are representative of three independent experiments. Data in (<b>B</b>,<b>C</b>) are presented as the means ± SD. * = <span class="html-italic">p</span> &lt; 0.05; ** = <span class="html-italic">p</span> &lt; 0.01; **** = <span class="html-italic">p</span> &lt; 0.0001. One-way analysis of variance was used.</p>
Full article ">
16 pages, 4655 KiB  
Article
Ruthenium(II) Complex with 8-Hydroxyquinoline Exhibits Antitumor Activity in Breast Cancer Cell Lines
by Amr Khalifa, Salah A. Sheweita, Asmaa Namatalla, Mohamed A. Khalifa, Alessio Nencioni and Ahmed S. Sultan
Cancers 2025, 17(2), 195; https://doi.org/10.3390/cancers17020195 - 9 Jan 2025
Viewed by 233
Abstract
Background/Objectives: Breast cancer (BC) remains one of the most prevalent and deadly cancers worldwide, with limited access to advanced treatments in developing regions. There is a critical need for novel therapies with unique mechanisms of action, especially to overcome resistance to conventional platinum-based [...] Read more.
Background/Objectives: Breast cancer (BC) remains one of the most prevalent and deadly cancers worldwide, with limited access to advanced treatments in developing regions. There is a critical need for novel therapies with unique mechanisms of action, especially to overcome resistance to conventional platinum-based drugs. This study investigates the anticancer potential of the ruthenium complex Bis(quinolin-8-olato)bis(triphenylphosphine)ruthenium(II) (Ru(quin)2) in ER-positive (T47D) and triple-negative (MDA-MB-231) BC cell lines. Results: Ru(quin)2 demonstrated dose-dependent cytotoxicity, with IC50 values of 48.3 μM in T47D cells and 45.5 μM in MDA-MB-231 cells. Its cytotoxic effects are primarily driven by apoptosis, as shown by increased BAX expression, enhanced caspase-3 activity, reduced Aurora B kinase levels, and elevated histone release. Ru(quin)2 also induced autophagy, evidenced by LC3-I to LC3-II conversion and reduced SQSTM1, partially mediated through MAPK signaling. Furthermore, Ru(quin)2 induced G0/G1 cell cycle arrest by downregulating cyclin D1, CDK4, and CDK6, alongside upregulation of the CDK inhibitor p21. Conclusions: Ru(quin)2 emerges as a potent candidate for BC treatment, with multiple mechanisms of action involving apoptosis, autophagy, and cell cycle arrest. Further studies are warranted to elucidate its detailed molecular mechanisms and evaluate its therapeutic potential in vivo, moving toward clinical applications for both ER-positive and triple-negative BC management. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Ru(quin)<sub>2</sub> triggers cytotoxicity in ER+ and TNBC breast cancer cells. (<b>A</b>) Chemical structure of Ru(quin)<sub>2</sub>. (<b>B</b>,<b>C</b>) Representative phase-contrast micrographs showing morphological changes in T47D (<b>B</b>) and MDA-MB-231 (<b>C</b>) cells after 72 h. (<b>D</b>) Dose-dependent inhibition of cell growth in T47D and MDA-MB-231 cells as assessed by the SRB assay after 72 h of Ru(quin)<sub>2</sub> treatment. (<b>E</b>,<b>F</b>) Colony formation assay showing the inhibitory effects of Ru(quin)<sub>2</sub> on T47D (<b>E</b>) and MDA-MB-231 (<b>F</b>) cells after 72 h. Colonies were stained with sulforhodamine B and quantified. Data represent the mean ± SD of three independent experiments performed in triplicate. Statistical significance: ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001. Abbreviations: ER+—estrogen receptor-positive, TNBC—triple-negative breast cancer.</p>
Full article ">Figure 2
<p>Ru(quin)<sub>2</sub> induces apoptosis and alters apoptotic markers in ER+ and TNBC breast cancer cells. (<b>A</b>) Quantification of apoptosis levels in T47D and MDA-MB-231 cells using an enzyme-linked immunosorbent assay. (<b>B</b>) Caspase-3 activity assay demonstrating Ru(quin)<sub>2</sub>-induced caspase activation in levels of T47D and MDA-MB-231 cells. (<b>C</b>) AURKB expression levels in T47D and MDA-MB-231 cells assessed by quantitative PCR. (<b>D</b>) Western blot analysis showing the expression of BAX and vinculin in T47D cells treated with Ru(quin)<sub>2</sub>. (<b>E</b>) Western blot analysis showing the expression of BAX and β-actin in MDA-MB-231 cells treated with Ru(quin)<sub>2</sub>. Representative blot from three independent experiments is shown. Data represent the mean ± SD of three independent experiments performed in triplicate. Statistical significance: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001. Abbreviations: ER+—estrogen receptor-positive, TNBC—triple-negative breast cancer, AURKB—Aurora B kinase. The original Western blot figures can be found in <a href="#app1-cancers-17-00195" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 3
<p>Ru(quin)<sub>2</sub> promotes autophagy in ER+ and TNBC breast cancer cells. (<b>A</b>) Immunoblot analysis of autophagy-related markers LC3, Atg13, FIP200, SQSTM1/p62, vinculin, phosphorylated ERK1/2 (Thr 202 and Tyr 204), and ERK1/2 in T47D cells treated with Ru(quin)<sub>2</sub>. The intensity of the LC3-II, Atg13, FIP200, and SQSTM1/p62 bands was quantified and normalized to that of vinculin bands, whereas the phosphorylated ERK1/2 bands were quantified and normalized to the respective total ERK1/2 bands. (<b>B</b>) Immunoblot analysis of autophagy-related markers LC3, Atg13, FIP200, SQSTM1/p62, β-actin, phosphorylated ERK1/2 (Thr 202 and Tyr 204), and ERK1/2 in MDA-MB-231 cells treated with Ru(quin)<sub>2</sub>. The intensity of the LC3-II, Atg13, FIP200, and SQSTM1/p62 bands was quantified and normalized to that of β-actin bands, whereas the phosphorylated ERK1/2 bands were quantified and normalized to the respective total ERK1/2 bands. Representative blot from three independent experiments is shown. Abbreviations: ER+—estrogen receptor-positive, TNBC—triple-negative breast cancer, LC3—microtubule-associated proteins 1A/1B light chain 3, Atg13—autophagy-related protein 13, FIP200—focal adhesion kinase family interacting protein of 200 kDa, SQSTM1/p62—sequestosome 1. The original Western blot figures can be found in <a href="#app1-cancers-17-00195" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 4
<p>Ru(quin)<sub>2</sub> induces G0/G1 phase cell cycle arrest in ER+ and TNBC breast cancer cells. (<b>A</b>) Flow cytometry analysis of the percentage of cells in each cell cycle phase (G1, S, and G2/M) in T47D cells treated with Ru(quin)<sub>2</sub>. (<b>B</b>) Flow cytometry analysis of cell cycle phases in MDA-MB-231 cells treated with Ru(quin)<sub>2</sub>. (<b>C</b>) Immunoblot analysis of CCND1, CDK4, CDK6, p21, and vinculin in T47D cells following Ru(quin)<sub>2</sub> treatment. The intensity of the CCND1, CDK4, CDK6, and p21 bands was quantified and normalized to that of vinculin bands. (<b>D</b>) Immunoblot analysis of the same markers in MDA-MB-231 cells. The intensity of the CCND1, CDK6, and p21 bands was quantified and normalized to that of vinculin bands. Data represent the mean ± SD of three independent experiments performed in triplicate. Representative blot from three independent experiments is shown. Statistical significance: * <span class="html-italic">p</span> &lt; 0.05; **: <span class="html-italic">p</span> &lt; 0.01; n.s: non-significant. Abbreviations: ER+—estrogen receptor-positive, TNBC—triple-negative breast cancer, CDK—cyclin-dependent kinase, CCND1—cyclin D1. The original Western blot figures can be found in <a href="#app1-cancers-17-00195" class="html-app">Supplementary File S1</a>.</p>
Full article ">
12 pages, 1140 KiB  
Article
Plasma Concentrations of Benzylpenicillin and Cloxacillin in Infective Endocarditis—With Special Reference to Delayed Hypersensitivity Reactions
by Malin Hägglund, Ulrika Snygg-Martin, Lars Olaison, Michael Stofkoper, Bert Ove Larsson and Magnus Brink
Antibiotics 2025, 14(1), 56; https://doi.org/10.3390/antibiotics14010056 - 9 Jan 2025
Viewed by 266
Abstract
Background: Current antibiotic regimens for infective endocarditis (IE) are effective but pose a high risk of delayed hypersensitivity reactions (DHR). Dose adjustments guided by therapeutic drug monitoring (TDM) could mitigate these risks while maintaining treatment efficacy. This study aimed to investigate the plasma [...] Read more.
Background: Current antibiotic regimens for infective endocarditis (IE) are effective but pose a high risk of delayed hypersensitivity reactions (DHR). Dose adjustments guided by therapeutic drug monitoring (TDM) could mitigate these risks while maintaining treatment efficacy. This study aimed to investigate the plasma concentration of benzylpenicillin and cloxacillin in patients with IE and explore associations between antibiotic concentrations and DHR. Methods: Plasma concentrations of benzylpenicillin and cloxacillin were measured as centre (midpoint concentrations between consecutive doses) and trough values during the first and third weeks of treatment in patients with IE. Patient characteristics and outcomes, including DHR, were documented. Results: A total of 55 patients were included, with 37 patients (67%) receiving benzylpenicillin and 18 (33%) receiving cloxacillin. The 90-day mortality rate was 3%. Both centre and trough concentration exhibited substantial interpatient variation for the two antibiotics, while intra-patient variability between weeks 1 and 3 remained low for most patients. Kidney function could explain, at best, 54% of the variation, and a multiple regression model including kidney function, body mass index, age, and albumin explained up to 68% of the variation for benzylpenicillin. There was no relation between high plasma concentration and the prevalence of DHR; conversely, we observed a tendency of low plasma concentrations in these patients. Conclusions: This study revealed significant interindividual variation in plasma concentrations for both studied penicillins. TDM might be useful in situations where concentrations are hard to predict, such as severe obesity or kidney failure. Additionally, we found no indication that high plasma concentrations are related to the prevalence of DHR. Full article
Show Figures

Figure 1

Figure 1
<p>Flowchart of the study. * Missing single sample.</p>
Full article ">Figure 2
<p>Centre and trough total plasma concentrations of benzylpenicillin and cloxacillin in treatment weeks 1 and 3. Benzylpenicillin, median (IQR); centre value, median 10 (5.1–21.5) and 20 (6.5–29.0), trough value 1.6 (0.79–5.3) and 3.6 (1.1–6.8). Cloxacillin, median (IQR); centre value 18 (7.3–27.0) and 17.5 (9.1–25.8). Trough value 3.7 (1.7–6.2) and 3.85 (1.7–7.4).</p>
Full article ">Figure 3
<p>Paired plasma concentrations (treatment weeks 1 and 3) of benzylpenicillin and cloxacillin. Wilcoxon signed-rank test with no significant intra-patient difference between weeks. Effective pairing with Spearman (one-tailed) 0.7–0.9, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
29 pages, 4274 KiB  
Review
Role of Ionizing Radiation Techniques in Polymeric Hydrogel Synthesis for Tissue Engineering Applications
by Ion Călina, Maria Demeter, Anca Scărișoreanu, Awn Abbas and Muhammad Asim Raza
Gels 2025, 11(1), 47; https://doi.org/10.3390/gels11010047 - 8 Jan 2025
Viewed by 317
Abstract
Hydrogels are widely utilized in industrial and scientific applications owing to their ability to immobilize active molecules, cells, and nanoparticles. This capability has led to their growing use in various biomedical fields, including cell culture and transplantation, drug delivery, and tissue engineering. Among [...] Read more.
Hydrogels are widely utilized in industrial and scientific applications owing to their ability to immobilize active molecules, cells, and nanoparticles. This capability has led to their growing use in various biomedical fields, including cell culture and transplantation, drug delivery, and tissue engineering. Among the available synthesis techniques, ionizing-radiation-induced fabrication stands out as an environmentally friendly method for hydrogel preparation. In alignment with the current requirements for cleaner technologies, developing hydrogels using gamma and electron beam irradiation technologies represents a promising and innovative approach for their biomedical applications. A key advantage of these methods is their ability to synthesize homogeneous three-dimensional networks in a single step, without the need for chemical initiators or catalysts. Additionally, the fabrication process is controllable by adjusting the radiation dose and dose rate. Full article
(This article belongs to the Special Issue Novel Gels for Topical Applications)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>a</b>) Multi-component polymeric hydrogels fabricated via electron (E)-beam irradiation. (<b>b</b>) Fluorescence images of hydrogels after 24 h and 72 h of cell seeding, obtained from the LIVE/DEAD assay (red fluorescence indicates dead cells; green fluorescence indicates living cells). Reproduced with permission [<a href="#B80-gels-11-00047" class="html-bibr">80</a>]. Copyright 2023, MDPI.</p>
Full article ">Figure 2
<p>Qualitative evaluation of the mineralization and calcium deposition of rabbit-bone-marrow-derived mesenchymal stem cells on tragacanth gum/poly(vinyl alcohol)/halloysite nanotube hydrogels after up to 21 days of incubation. Reproduced with permission [<a href="#B81-gels-11-00047" class="html-bibr">81</a>]. Copyright 2023, Elsevier.</p>
Full article ">Figure 3
<p>Appearance of silk-fibroin-based soft and tough tissue engineering scaffolds. Reproduced with permission from [<a href="#B97-gels-11-00047" class="html-bibr">97</a>]. Copyright 2020 American Chemical Society.</p>
Full article ">Figure 4
<p>Human clinical trial outcomes of γ-irradiation-sterilized wound dressing applied to a nevus resection wound measuring 5.2 × 2.7 cm<sup>2</sup> over 4 weeks. (<b>A</b>) Congenital melanocytic nevus, (<b>B</b>) skin wound post-surgical resection, (<b>C</b>) grafting procedure using small dressing pieces, (<b>D</b>) complete wound coverage by the dressing, (<b>E</b>) early biodegradability observed after seven days, and (<b>F</b>) early regeneration observed after four weeks. Reproduced with permission from [<a href="#B103-gels-11-00047" class="html-bibr">103</a>]. Copyright 2017 Elsevier.</p>
Full article ">Figure 5
<p>The potential applications of the use of ionizing radiation in TE applications.</p>
Full article ">Scheme 1
<p>Advantages and limitations of E-beam and gamma irradiation technology.</p>
Full article ">Scheme 2
<p>Radiation-induced polymerization [<a href="#B54-gels-11-00047" class="html-bibr">54</a>].</p>
Full article ">Scheme 3
<p>Radiation-induced graft polymerization [<a href="#B46-gels-11-00047" class="html-bibr">46</a>].</p>
Full article ">
15 pages, 2181 KiB  
Article
Characterization and Otoprotective Effects of Polysaccharides from Arthrospira platensis
by Matteo Banti, Mercedes Garcia-Gil, Lorenzo Guidotti, Graziano Di Giuseppe, Simona Rapposelli, Daniela Monti, Silvia Tampucci, Marinella De Leo, Francesca Gado, Paola Nieri and Clementina Manera
Molecules 2025, 30(2), 224; https://doi.org/10.3390/molecules30020224 - 8 Jan 2025
Viewed by 262
Abstract
Hearing loss is one of the most common sensory disorders in humans, and a large number of cases are due to ear cell damage caused by ototoxic drugs including anticancer agents, such as cisplatin. The recent literature reported that hearing loss is promoted [...] Read more.
Hearing loss is one of the most common sensory disorders in humans, and a large number of cases are due to ear cell damage caused by ototoxic drugs including anticancer agents, such as cisplatin. The recent literature reported that hearing loss is promoted by an excessive generation of reactive oxygen species (ROS) in cochlea cells, which causes oxidative stress. Recently, polysaccharides from the cyanobacterium Arthrospira platensis showed many biological activities, including antioxidant activity, suggesting their potential use to combat hearing loss. On these bases, this study describes the extraction, purification, and characterization of water-soluble polysaccharides from A. platensis (SPPs) and the investigation of their protective role against cisplatin toxicity on House Ear Institute-Organ of Corti (HEI-OC1) cells. The results showed that SPPs (5–80 µg/mL) induced a dose-dependent increase in viability, statistically significant at 40 µg/mL and 80 µg/mL. Moreover, SPPs, evaluated at 80 µg/mL, inhibited the cisplatin-induced ROS level increase in HEI-OC1. This evidence highlights the potential of SPPs as natural candidates to protect cochlear ear cells against ototoxic oxidative agents. Moreover, in view of the potential use of microalgal polysaccharides to realize hydrogels, SPPs could also represent a healthy carrier for other topically administered otoprotective agents. Full article
Show Figures

Figure 1

Figure 1
<p>FT-IR analysis of SPPs isolated from <span class="html-italic">Spirulina platensis</span>.</p>
Full article ">Figure 2
<p>The <sup>1</sup>H-NMR (<b>a</b>) and <sup>13</sup>C-NMR (<b>b</b>) spectrum of SPPs.</p>
Full article ">Figure 3
<p>LC-MS profile registered in negative ionization mode of 1-phenyl-3-methyl-5-pyrazolone (PMP)-labeled monosaccharides of the hydrolyzed SPPs (<b>a</b>) and standard monosaccharides (<b>b</b>).</p>
Full article ">Figure 4
<p>Radical scavenging activities of SPPs (curve of inhibition percentage versus SPP concentration). (<b>a</b>) ABTS radical scavenging activity (ABTS<sup>•</sup> assay); (<b>b</b>) hydroxyl radical scavenging activity (<sup>•</sup>OH assay). VITC, ascorbic acid. Data are means ± SEM from 3 experiments.</p>
Full article ">Figure 5
<p>(<b>a</b>) Effect of SPPs on viability of HEI-OC1 cells. Data were normalized to the absorbance of the cells grown in the absence of SPPs or their solvent; (<b>b</b>) effect of preincubation with SPPs on viability of HEI-OC1 cells treated with cisplatin. Data were normalized to the absorbance of the cells grown in the absence of SPPs and their solvent. Data are presented as means ± SEM from 3–4 experiments. Significance was measured using a non-parametric ANOVA followed by Tukey’s multiple comparisons test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. cells cultured in the absence of SPPs.</p>
Full article ">Figure 6
<p>HEI-OC1 cells were treated with 30 µM cisplatin in the presence or the absence of SPPs. ROS levels were normalized to the values from cells grown in the absence of SPPs and their solvent. The data are expressed as means ± SEM of 3 experiments. Statistical significance obtained with Student’s <span class="html-italic">t</span>-test: ** <span class="html-italic">p</span> &lt; 0.01 vs. control (correspondent blank bar). CisPt, cisplatin.</p>
Full article ">
15 pages, 4795 KiB  
Article
Epithelial–Mesenchymal Transition Suppression by ML210 Enhances Gemcitabine Anti-Tumor Effects on PDAC Cells
by Keisuke Takemura, Kyohei Ikeda, Hayato Miyake, Yoshio Sogame, Hiroaki Yasuda, Nobuhiro Okada, Kazumi Iwata, Junichi Sakagami, Kanji Yamaguchi, Yoshito Itoh and Atsushi Umemura
Biomolecules 2025, 15(1), 70; https://doi.org/10.3390/biom15010070 - 6 Jan 2025
Viewed by 335
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in the world. Neoadjuvant chemotherapy (NAC) has become a standard treatment for patients scheduled for surgical resection, but the high rate of postoperative recurrence is a critical problem. Optimization of NAC is desirable [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in the world. Neoadjuvant chemotherapy (NAC) has become a standard treatment for patients scheduled for surgical resection, but the high rate of postoperative recurrence is a critical problem. Optimization of NAC is desirable to reduce postoperative recurrence and achieve long-term survival. However, if a patient’s general condition deteriorates due to NAC toxicity, surgical outcomes may be compromised. Therefore, we aimed to identify drug(s) that can be used in combination with gemcitabine (GEM), a drug widely used for the treatment of PDAC, to inhibit distant metastatic recurrence, particularly after surgery. After several screening steps, ML210, a low molecular weight chemical, was found to suppress the epithelial–mesenchymal transition (EMT) in PDAC cells in combination with GEM. Specifically, low dose ML210 in combination with GEM was sufficient for cell migration without apparent toxicity or cell death. Mechanistically, ML210, which was developed as a glutathione peroxidase 4 (GPX4) inhibitor to induce lipid peroxidation, increased the oxidized lipid concentrations in PDAC cells. The oxidization of the cell membrane lipids may suppress EMT, including cell migration. Since EMT is a major malignant phenotype of PDAC, our findings may lead to the advancement of PDAC therapy, especially in the prevention of postoperative recurrence. Full article
(This article belongs to the Section Cellular Biochemistry)
Show Figures

Figure 1

Figure 1
<p>Drug screening. (<b>A</b>) Schematic explanation for the drug screening process. A total of 10,000 cells were seeded in a 96-well dish with 100 µL of culture medium, and we replaced the medium with each drug compound at concentrations of 3 µM after 24 h. The cells were harvested after a 48 h incubation. (<b>B</b>) Vimentin and N-cadherin gene expression analyzed by real-time qPCR. (<b>C</b>) Cell viability of PANC-1 cells. A total of 5000 cells/well (100 μL) were treated with each compound at the indicated concentrations (0.3, 3, 10, 30 μM), and their viabilities were assessed after a 48 h incubation (vs. non-treatment control). Data were obtained from three independent experiments. Mean ± SD data are displayed as fold changes relative to the non-treated group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (vs. the non-treated group).</p>
Full article ">Figure 2
<p>(<b>A</b>) Vimentin, N-cadherin, and GATA6 gene expression analyzed by real-time qPCR. A total of 10,000 cells were seeded in a 96-well dish with 100 µL of culture medium, and we replaced the medium with each chemical compound after 24 h. Each drug was used at the concentration of 3 µM, and the evaluation was performed 48 h later. (<b>B</b>,<b>C</b>) Diff-Quik staining images and the graphs of quantified cells that were treated with 0.05/0.1/0.3 μM ML210 or CA-074 methyl ester the after migration assay was performed. Migrated cells were counted in × 200 images (<span class="html-italic">n</span> = 5). Scale bar: 200 μm. Mean ± SD data are displayed as fold changes relative to the non-treated group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (vs. the non-treated group).</p>
Full article ">Figure 3
<p>(<b>A</b>) Cell viability of PANC-1 cells (vs the non-treatment control). A total of 5000 cells/well (100 μL) were treated with 3mg/L GEM or 3 mg/L GEM + ML210 at the indicated doses. Data were obtained from three independent experiments. (<b>B</b>) LDH leakage of PANC-1 cells (vs the high control (HL)). A total of 5000 cells/well (100 μL) were treated with 3 mg/L GEM or 3mg/L GEM + ML210 at indicated doses. (<b>C</b>,<b>D</b>) Diff-Quik staining images and the graphs of quantified cells that were treated with 3 mg/L GEM or 3 mg/L GEM + 0.025/0.05/0.1/0.3 μM ML210 after migration. Migrated cells were counted in x 200 images (<span class="html-italic">n</span> = 5). Scale bar: 200 μm. (<b>E</b>) The protein expression of vimentin, GPX4, and tubulin were measured by western blotting analysis. PANC-1 cells were pretreated with 3 mg/L GEM and 0.025/0.05/0.1/0.3μM ML210 for 48 h. Numbers indicated below the blotting bands are quantified protein levels relative to the non-treatment control. Western blotting original images can be found in <a href="#app1-biomolecules-15-00070" class="html-app">Supplementary figure</a>. Mean ± SD data are displayed as fold changes relative to the non-treated group. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (vs. the non-treated group).</p>
Full article ">Figure 4
<p>(<b>A</b>) The protein expression of vimentin, GPX4, cleaved caspase-3, and tubulin were measured by a western blotting assay. PANC-1 cells were treated with VP16 or 3mg/L GEM and/or ML210 at the indicated concentrations. VP16, or etoposide, is known as an apoptosis inducer. Numbers indicated below the blotting bands are quantified protein levels relative to the non-treatment control. Western blotting original images can be found in <a href="#app1-biomolecules-15-00070" class="html-app">Supplementary figures</a>. (<b>B</b>,<b>C</b>) Flow cytometer analysis of BODIPY581/591 C11 positive cells to detect lipid peroxidation and the graphs of data obtained from five independent experiments. The cells were treated with 0.25 μM ML210, 3 mg/L GEM + 0.1 μM ML210, or 100 μM Tert-butyl hydroperoxide (tBHP), which is an oxidative stress inducer that was used as a positive control drug. After a 24 h incubation with the indicated drugs, the positive cells were evaluated. Mean ± SD data are displayed. ** <span class="html-italic">p</span> &lt; 0.01 (vs. the non-treated group).</p>
Full article ">Figure 5
<p>(<b>A</b>) The protein expressions of vimentin, GPX4, and tubulin were measured by western blotting analysis. MiaPaCa-2 cells were treated with 3mg/L GEM and ML210 at the indicated concentrations for 48 h. Numbers indicated below the blotting bands are quantified protein levels relative to the non-treatment control. Western blotting original images can be found in <a href="#app1-biomolecules-15-00070" class="html-app">Supplementary figure</a>. (<b>B</b>) Vimentin gene expression analyzed by real-time qPCR. (<b>C</b>,<b>D</b>) Diff-Quik staining images of the migration assay. The graphs of quantified cells that were treated with 3 mg/L GEM + 0.025/0.05/0.1/0.2 μM ML210. The invaded cells were counted in ×200 images (<span class="html-italic">n</span> = 5). Scale bar indicates 200 μm. Mean ± SD data are displayed as fold changes relative to the non-treated group. ** <span class="html-italic">p</span> &lt; 0.01 (vs. the non-treated group).</p>
Full article ">
38 pages, 4554 KiB  
Review
Oncolytic Viruses and Immunotherapy for the Treatment of Uveal Melanoma and Retinoblastoma: The Current Landscape and Novel Advances
by Merve Kulbay, Nicolas Tuli, Massimo Mazza, Armaan Jaffer, Sarinee Juntipwong, Emily Marcotte, Stuti Misty Tanya, Anne Xuan-Lan Nguyen, Miguel N. Burnier and Hakan Demirci
Biomedicines 2025, 13(1), 108; https://doi.org/10.3390/biomedicines13010108 - 6 Jan 2025
Viewed by 513
Abstract
Intraocular malignant tumors are rare; however, they can cause serious life-threatening complications. Uveal melanoma (UM) and retinoblastoma (RB) are the most common intraocular tumors in adults and children, respectively, and come with a great disease burden. For many years, several different treatment modalities [...] Read more.
Intraocular malignant tumors are rare; however, they can cause serious life-threatening complications. Uveal melanoma (UM) and retinoblastoma (RB) are the most common intraocular tumors in adults and children, respectively, and come with a great disease burden. For many years, several different treatment modalities for UM and RB have been proposed, with chemotherapy for RB cases and plaque radiation therapy for localized UM as first-line treatment options. Extraocular extension, recurrence, and metastasis constitute the major challenges of conventional treatments. To overcome these obstacles, immunotherapy, which encompasses different treatment options such as oncolytic viruses, antibody-mediated immune modulations, and targeted immunotherapy, has shown great potential as a novel therapeutic tool for cancer therapy. These anti-cancer treatment options provide numerous advantages such as selective cancer cell death and the promotion of an anti-tumor immune response, and they prove useful in preventing vision impairment due to macular and/or optic disc involvement. Numerous factors such as the vector choice, route of administration, dosing, and patient characteristics must be considered when engineering an oncolytic virus or other forms of immunotherapy vectors. This manuscript provides an in-depth review of the molecular design of oncolytic viruses (e.g., virus capsid proteins and encapsulation technologies, vectors for delivery, cell targeting) and immunotherapy. The most recent advances in preclinical- and clinical-phase studies are further summarized. The recent developments in virus-like drug conjugates (i.e., AU011), oncolytic viruses for metastatic UM, and targeted immunotherapies have shown great results in clinical trials for the future clinical application of these novel technologies in the treatment algorithm of certain intraocular tumors. Full article
Show Figures

Figure 1

Figure 1
<p>Downstream cellular signaling dysregulations involved in the pathogenesis of uveal melanoma following <span class="html-italic">GNAQ</span> and <span class="html-italic">GNA11</span> mutations. <span class="html-italic">GNAQ</span> and <span class="html-italic">GNA11</span> mutations lead to the uncontrolled activation of G protein signaling pathways, ultimately inducing cancer cell survival, proliferation, and the inhibition of apoptosis. Created in BioRender. Kulbay, M. (2024) <a href="https://BioRender.com/d29b243" target="_blank">https://BioRender.com/d29b243</a>.</p>
Full article ">Figure 2
<p>Schematic illustration of pathways altered in retinoblastoma pathogenesis. Four main pathways are involved in retinoblastoma (Rb) pathogenesis: DNA damage pathways and Wnt signaling, RAS signaling, and Notch signaling pathways. Mutations in gene expression lead to disruptions in downstream signalization. Created in BioRender. Kulbay, M. (2024) <a href="https://BioRender.com/x11l167" target="_blank">https://BioRender.com/x11l167</a>.</p>
Full article ">Figure 3
<p>Schematic illustration of the mechanism of action of oncolytic viruses. Oncolytic viruses (OVs), which can be dsDNA-, dsRNA-, or ssRNA-based, interact with cancerous cells through surface proteins or fibers that recognize the extracellular receptors. The OV is internalized through receptor-mediated endocytosis or through cell membrane fusion. Once internalized, dsDNA OVs translocate to the nucleus where their genome is integrated within the host DNA to induce the transcription of viral proteins. RNA-based OVs (not represented in this illustration) undergo replication within the cytoplasm. Upon viral progeny assembly, viral particles and cell antigens are expulsed into the tumor microenvironment, leading to cell oncolysis and the continuation of the infectious cycle. Created in BioRender. Kulbay, M. (2024) <a href="https://BioRender.com/w63e114" target="_blank">https://BioRender.com/w63e114</a>.</p>
Full article ">Figure 4
<p>Modifying oncolytic viruses to enhance tumor recognition. (<b>A</b>) Oncolytic viruses (OVs) are modified to express bispecific ligands, such as single-chain antibodies, that facilitate the interaction between viral capsid fiber/envelope proteins and their target receptor. (<b>B</b>) OVs are modified to have their capsid fibers/envelope fused to a tumor-targeting ligand which enhances tumor-specific protein recognition. (<b>C</b>) OVs are modified to have their viral capsid protein/fibers replaced with another virus’s protein to enhance the recognition of specific tumors. Created in BioRender. Tuli, N. (2025) <a href="https://BioRender.com/b97r111" target="_blank">https://BioRender.com/b97r111</a>.</p>
Full article ">Figure 5
<p>Arming oncolytic viruses to enhance tumor elimination through four mechanisms. (<b>A</b>) Oncolytic viruses (OVs) are armed to express pro-immunogenic cell death (ICD) proteins that enhance apoptosis, necrosis, pyroptosis, or autophagy. (<b>B</b>) Arming OVs to express chemokines or cytokines, which are immune mediators that recruit T cells (chemokines) or enhance their activation and proliferation (cytokines). (<b>C</b>) OVs armed with immune checkpoint inhibitors (ICIs) that interfere with tumor-mediated immunosuppression, leading to enhanced T cell activation and proliferation. (<b>D</b>) Arming OVs with proteins such as bispecific T cell engager (BiTE) facilitates the interaction between T cells and tumor-associated antigens, enhancing T cell function. Created in BioRender. Tuli, N. (2025) <a href="https://BioRender.com/b97r111" target="_blank">https://BioRender.com/b97r111</a>.</p>
Full article ">Figure 6
<p>Schematic illustration of the mechanism of action of Talimogene Laherparepvec. Created in BioRender. Kulbay, M. (2024) <a href="https://BioRender.com/c38m199" target="_blank">https://BioRender.com/c38m199</a>.</p>
Full article ">Figure 7
<p>A schematic illustration of the mechanism of action of immune checkpoint inhibitors. (<b>A</b>) Nivolumab and pembrolizumab block the PD-1 receptor, interfering with PD-L1 mediated immunosuppression. (<b>B</b>) Ipilimumab binds CTLA-4, which inhibits it from competing with CD-28 for CD80/86, leading to increased costimulatory T cell signaling. (<b>C</b>) Anti-GD2 antibodies block tumor-expressed GD2 from interacting with its receptor, Siglec-7, restoring NK cell function. Created in BioRender. Tuli, N. (2025) <a href="https://BioRender.com/b97r111" target="_blank">https://BioRender.com/b97r111</a>.</p>
Full article ">Figure 8
<p>A schematic of adoptive cell transfer therapies. LEFT: A schematic of CAR T cell therapy, where patient-derived T cells are transfected with tumor-specific CAR transgenes, leading to the expression of CARs. These cells are then expanded and re-infused into the patient. RIGHT: A schematic of TIL therapy whereby TILs are isolated from a patient-derived tumor specimen, isolated, and then expanded with cytokines ex vivo. The TILs are then re-infused into the patient. Created in BioRender. Kulbay, M. (2025) <a href="https://BioRender.com/x32x257" target="_blank">https://BioRender.com/x32x257</a>.</p>
Full article ">
15 pages, 449 KiB  
Article
Efficacy and Safety Assessment of Antifungal Prophylaxis with Posaconazole Using Therapeutic Drug Monitoring in Pediatric Patients with Oncohematological Disorders—A Single-Centre Study
by Karolina Liszka, Paweł Marschollek, Dawid Przystupski, Jowita Frączkiewicz, Monika Mielcarek-Siedziuk, Igor Olejnik, Zuzanna Gamrot, Natalia Haze, Agnieszka Kwella, Paulina Zalewska, Matylda Resztak, Marek Ussowicz and Krzysztof Kałwak
J. Fungi 2025, 11(1), 38; https://doi.org/10.3390/jof11010038 - 6 Jan 2025
Viewed by 308
Abstract
Introduction: Posaconazole is recommended for prophylaxis in pediatric immunocompromised patients. Due to its variability in bioavailability and drug-to-drug interactions, EBMT recommends regimens based on therapeutic drug monitoring (TDM). Materials and methods: In total, 171 oncohematological pediatric patients on posaconazole prophylaxis were included. Full [...] Read more.
Introduction: Posaconazole is recommended for prophylaxis in pediatric immunocompromised patients. Due to its variability in bioavailability and drug-to-drug interactions, EBMT recommends regimens based on therapeutic drug monitoring (TDM). Materials and methods: In total, 171 oncohematological pediatric patients on posaconazole prophylaxis were included. Full pharmacokinetic posaconazole profiles were assessed in 51 children. The efficacy and safety of posaconazole was evaluated by measuring the drug concentration, with dose modification attempted in patients with suboptimal results. The influence of modifying factors on the posaconazole plasma concentration (PPC) was investigated. Results: An insufficient PPC was the main issue, but no significant increase in prophylaxis failure was reported. The modification of the dosage resulted in the optimization of PPC in 50% of patients. No significant correlation between age, gender, diagnosis or the posaconazole dosage and the PPC was found. HCT, total parenteral nutrition and diarrhea were associated with a lower PPC. Hypoalbuminemia was related to both higher and lower PPC. The concomitant administration of specified drugs significantly impacted the PPC. Conclusions: TDM allows the identification of patients receiving non-optimal treatment and offers an opportunity to improve the efficacy and safety of the therapy. However, further research involving larger patient groups and longer observation periods are needed to determine the optimal dosing and target PPC in pediatric patients. Full article
(This article belongs to the Special Issue Advances in Antifungal Drugs, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Distribution of posaconazole concentrations across different age groups.</p>
Full article ">
9 pages, 1468 KiB  
Case Report
Severe Lipoatrophy in a Growth Hormone Deficient Toddler Girl Treated with a Non-Pegylated Long-Acting Growth Hormone
by Atilla Büyükgebiz and And Demir
Children 2025, 12(1), 58; https://doi.org/10.3390/children12010058 - 4 Jan 2025
Viewed by 478
Abstract
Background: Recombinant growth hormone (rhGH) has been used since 1985 to treat growth hormone (GH)-induced short stature, typically associated with transient adverse events. However, lipoatrophy, characterized by irreversible damage to subcutaneous fat, was first reported in 1999 and linked to antibody formation. In [...] Read more.
Background: Recombinant growth hormone (rhGH) has been used since 1985 to treat growth hormone (GH)-induced short stature, typically associated with transient adverse events. However, lipoatrophy, characterized by irreversible damage to subcutaneous fat, was first reported in 1999 and linked to antibody formation. In 2021, localized lipoatrophy was observed in 14.5% of patients receiving daily rhGH, with repeated injections at the same sites being a common contributing factor. Long-acting rhGH (LAGH) preparation offers the advantage of weekly injections, enhancing patient comfort and adherence to treatment. Methods: This case report discusses a 5.5-year-old girl born at 40 weeks of gestation with a birth weight of 2300 g, diagnosed with idiopathic short stature and borderline GH secretion, along with a history of mild intrauterine growth retardation. Results: After initiating treatment with somatrogon, a non-pegylated fusion protein formulation of LAGH at the standard dose of 0.66 mg/kg body weight weekly, administered by her family, she developed localized lipoatrophy at the injection site within eleven weeks. The injections were performed consistently in the same area of the right upper arm, where lipoatrophy emerged. Following the onset of this adverse effect, her treatment was adjusted to daily rhGH, with strict instructions to rotate injection sites. Despite these clear instructions, follow-up revealed that the parents continued to administer injections with the non-pegylated LAGH fusion protein formulation, this time in the left upper arm, leading to a recurrence of lipoatrophy within eight weeks. Conclusions: The recurrence underscores the importance of proper injection techniques, particularly site rotation, in preventing localized adverse effects. Given the limitations of this case, where the recommended adjustments were not followed by the parents, it is crucial to emphasize that the administration of the preparation should be discontinued immediately upon the appearance of side effects such as lipoatrophy. Individual reactions to drugs are always possible, and this highlights the need for clinician vigilance in monitoring and addressing adverse effects promptly during treatments with LAGH. Full article
(This article belongs to the Section Pediatric Endocrinology & Diabetes)
Show Figures

Figure 1

Figure 1
<p>Lipoatrophy in the right upper arm after 11 weeks of weekly injections with the non-pegylated LAGH fusion protein formulation. The patient, a 5.5-year-old girl diagnosed with idiopathic short stature and growth retardation, developed lipoatrophy at the injection site due to repeated injections in the same localized area. The right upper arm mid-circumference measured 14 cm, compared to 16 cm in the left upper arm.</p>
Full article ">Figure 2
<p>Progression of lipoatrophy in the left upper arm after an additional 8 weeks of using the non-pegylated LAGH fusion protein formulation during follow-up. Despite instructions to switch to daily hGH injections, the parents continued administering the non-pegylated LAGH fusion protein formulation in the left upper arm, resulting in further lipoatrophy. At the 8-week follow-up, the left upper arm mid-circumference had decreased to 14 cm, while the right upper arm showed slight recovery, with a mid-circumference of 14.5 cm.</p>
Full article ">
12 pages, 388 KiB  
Article
Therapeutic Drug Monitoring of Vancomycin in Hemodialysis Patients in a Hospital in North-East Romania
by Aurelia Crețu, Luanda Irina Mititiuc, Iulia-Daniela Lungu, Mihaela Mihaila, Irina Dima, Adrian Covic and Cristina Mihaela Ghiciuc
Antibiotics 2025, 14(1), 34; https://doi.org/10.3390/antibiotics14010034 - 4 Jan 2025
Viewed by 566
Abstract
Background/Objectives: Vancomycin is a reserve antibiotic that is frequently prescribed for central venous catheter (CVC)-associated infections in hemodialysis patients. Hemodialysis patients are very fragile patients and the presence of CVCs increases the risk of sepsis. We conducted a prospective study, evaluating the [...] Read more.
Background/Objectives: Vancomycin is a reserve antibiotic that is frequently prescribed for central venous catheter (CVC)-associated infections in hemodialysis patients. Hemodialysis patients are very fragile patients and the presence of CVCs increases the risk of sepsis. We conducted a prospective study, evaluating the needs of changes in vancomycin dosing for treatment based on the use of the new 2020 vancomycin dosing guidelines, to increase drug safety (preventing subtherapeutic or supratherapeutic doses and offering therapeutic concentrations of the drug) in a particular group of patients with sepsis caused by catheter infections and being on intermittent hemodialysis. Methods: This prospective study included patients with sepsis caused by catheter infections and being on intermittent hemodialysis, treated with vancomycin, admitted in the nephrology department and intensive care unit (ICU). Vancomycin levels were adjusted according to the 2020 vancomycin guidelines. Results: In our study, nine (45%) patients had a vancomycin AUC between 400 and 600 mcg × h/mL, five (25%) patients had a subtherapeutic AUC, and six (30%) patients had a supratherapeutic AUC. It is important to mention that in 10 (50%) of the patients included in the study, the loading and maintenance doses mentioned in the protocol were respected, but 50% of them had a supratherapeutic AUC. We observed that a supratherapeutic AUC occurred when the loading dose was 1500 mg or 2000 mg, and in one case at 1000 mg with a low BMI. Conclusions: a therapeutic level of vancomycin can often be difficult to achieve because of different reasons, mainly in hemodialysis patients. Full article
(This article belongs to the Section Antibiotic Therapy in Infectious Diseases)
Show Figures

Figure 1

Figure 1
<p>Patients’ selection.</p>
Full article ">
28 pages, 7774 KiB  
Article
Chemoresistance in Pancreatic Cancer: The Role of Adipose-Derived Mesenchymal Stem Cells and Key Resistance Genes
by Shahram Parvaneh, Vanda Miklós, Zoltán Gábor Páhi, Diána Szűcs, Tamás Monostori, Szilárd Póliska, Viktória Venglovecz, Tibor Pankotai, Lajos Kemény and Zoltán Veréb
Int. J. Mol. Sci. 2025, 26(1), 390; https://doi.org/10.3390/ijms26010390 - 4 Jan 2025
Viewed by 612
Abstract
Drug resistance is a significant challenge in pancreatic ductal adenocarcinoma (PDAC), where stromal elements such as adipose-derived mesenchymal stem cells (ASCs) contribute to a chemoresistant tumor microenvironment (TME). This study explored the effects of oxaliplatin (OXP) and 5-fluorouracil (5-FU) on PDAC cells (Capan-1) [...] Read more.
Drug resistance is a significant challenge in pancreatic ductal adenocarcinoma (PDAC), where stromal elements such as adipose-derived mesenchymal stem cells (ASCs) contribute to a chemoresistant tumor microenvironment (TME). This study explored the effects of oxaliplatin (OXP) and 5-fluorouracil (5-FU) on PDAC cells (Capan-1) and ASCs to investigate the mechanisms of chemoresistance. While OXP and 5-FU reduced Capan-1 viability in a dose- and time-dependent manner, ASCs demonstrated high resistance, maintaining > 90% viability even at cytotoxic doses. Transcriptomic analyses revealed OXP-induced transcriptional reprogramming in ASCs, with over 7000 differentially expressed genes, highlighting the pathways related to DNA damage response, cell cycle regulation, and stress-related signaling. In contrast, 5-FU elicited limited transcriptional changes, affecting only 192 genes. Cytokine proteome profiling revealed that OXP-treated ASCs significantly influenced the tumor microenvironment by promoting immune evasion (via IL-4, GM-CSF, IP-10, and GROα) and driving extracellular matrix remodeling (through EMMPRIN and DPPIV). In contrast, 5-FU induced comparatively weaker effects, primarily limited to hypoxia-related pathways. Although OXP reduced angiogenic factors, it paradoxically activated pro-survival pathways, thereby enhancing ASC-mediated tumor support. These findings underscore ASCs as modulators of chemoresistance via secretome alterations and stress adaptation. Therefore, future strategies should prioritize the precise targeting of tumor cells while also focusing on the development of personalized treatments to achieve durable therapeutic responses in PDAC. Full article
(This article belongs to the Special Issue The Roles of Mesenchymal Stem/Stromal Cells in Tumor Therapies)
Show Figures

Figure 1

Figure 1
<p>Evaluating the cytotoxic effects of OXP and 5-FU on Capan-1 cells and ASCs using MTT assay. The cells were treated for 48 h with concentrations (ranging from 6.25 to 1000 μM) of (<b>a</b>) OXA and (<b>b</b>) 5-FU. Cell viability is expressed as a percentage of untreated control (Ctrl). Mitomycin (Mito) 20 μg/mL was used as the positive control. (mean ± SD, <span class="html-italic">n</span> = 3). ** <span class="html-italic">p</span> &lt; 0.01 and # <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>The overall results of RNA sequencing analysis comparing 5-FU and OXP-treated ASCs with the untreated control are presented in (<b>A</b>) PCA plot, (<b>B</b>) heatmap, and (<b>C</b>) Venn diagrams. D25, D39, and D45 represent three biological donor codes (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 3
<p>Significantly differentially expressed genes (DEGs) in ASCs treated with 5-FU and OXP compared with untreated controls. (<b>A</b>) The heatmap shows distinct clustering of control, 5-fluorouracil, and oxaliplatin treatments when only differentially expressed genes (DEGs) are plotted. Venn diagrams of (<b>B</b>) upregulated genes and (<b>C</b>) downregulated genes. D25, D39, and D45 represent three different biological donor codes (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 4
<p>Differentially expressed gene set enrichment analysis (GSEA) results for ASCs treated with OXP: (<b>A</b>) Hallmark gene sets, (<b>B</b>) KEGG pathways, and (<b>C</b>) GO term gene sets pathways.</p>
Full article ">Figure 5
<p>GSEA results from the Hallmark and GO Pathway databases for ASCs treated with 5-FU. (<b>A</b>) Hallmark gene sets, (<b>B</b>) GO term gene sets.</p>
Full article ">Figure 6
<p>Cytokine expression profile of ASCs following 5-FU and OXP treatment. Compared with untreated controls, the cytokine secretion profile of ASCs at 48 h post-treatment were assessed using the proteome profiler human cytokine array (mean ± SD, <span class="html-italic">n</span> = 3). <b>FGF-19</b>: Fibroblast Growth Factor 19, <b>GM-CSF</b>: Granulocyte–Macrophage Colony-Stimulating Factor, <b>GROα</b>: Growth-Regulated Oncogene Alpha (CXCL1), <b>IL-4</b>: Interleukin 4, <b>IP-10</b>: Interferon Gamma-Induced Protein 10 (CXCL10), <b>DPPIV</b>: Dipeptidyl Peptidase IV (CD26), <b>MIP-α</b>: Macrophage Inflammatory Protein-alpha (CCL3), <b>SDF-1α</b>: Stromal Cell-Derived Factor 1 (CXCL12), <b>uPAR</b>: Urokinase Plasminogen Activator Receptor, <b>VEGF</b>: Vascular Endothelial Growth Factor. Although twofold changes were measured for several cytokines, the significance values ranged from <span class="html-italic">p</span> = 0.05 to <span class="html-italic">p</span> = 0.06. (* <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01,).</p>
Full article ">Figure 7
<p>Schematic illustration of the effects of 5-FU and OXP treatments on ASCs, their potential role in the PDAC tumor microenvironment, and the development of chemotherapy resistance. <b>CAFs</b>: cancer-associated fibroblasts, <b>ECM</b>: extracellular matrix, <b>FGF-19</b>: Fibroblast Growth Factor 19, <b>DPPIV</b>: Dipeptidyl Peptidase IV (CD26), <b>IP-10</b>: Interferon Gamma-Induced Protein 10 (CXCL10), <b>IL-4</b>: Interleukin 4, <b>GM-CSF</b>: Granulocyte–Macrophage Colony-Stimulating Factor, <b>GROα</b>: Growth-Regulated Oncogene Alpha (CXCL1), <b>VEGF</b>: Vascular Endothelial Growth Factor, <b>uPAR</b>: Urokinase Plasminogen Activator Receptor, <b>SDF-1α</b>: Stromal Cell-Derived Factor 1 (CXCL12), <b>HIF-1α</b>: Hypoxia-Inducible Factor 1, <b>IL-6</b>: Interleukin 6, <b>IFNγ</b>: Interferon Gamma, <b>NF-κB</b>: Nuclear Factor Kappa B. Created in BioRender.com.</p>
Full article ">
20 pages, 5057 KiB  
Article
Chemoprotective Mechanism of Sodium Thiosulfate Against Cisplatin-Induced Nephrotoxicity Is via Renal Hydrogen Sulfide, Arginine/cAMP and NO/cGMP Signaling Pathways
by George J. Dugbartey, Karl K. Alornyo, Ismaila Adams, Samuel Adjei, Daniel Amoah and Richard Obeng-Kyeremeh
Int. J. Mol. Sci. 2025, 26(1), 384; https://doi.org/10.3390/ijms26010384 - 4 Jan 2025
Viewed by 342
Abstract
Cisplatin is a common and highly effective chemotherapeutic agent whose nephrotoxic side effect is well-characterized. Sodium thiosulfate (STS), an FDA-approved hydrogen sulfide (H2S) donor drug, is emerging as a chemoprotective agent against cisplatin-induced nephrotoxicity (CIN). In this study, we investigated the [...] Read more.
Cisplatin is a common and highly effective chemotherapeutic agent whose nephrotoxic side effect is well-characterized. Sodium thiosulfate (STS), an FDA-approved hydrogen sulfide (H2S) donor drug, is emerging as a chemoprotective agent against cisplatin-induced nephrotoxicity (CIN). In this study, we investigated the chemoprotective mechanism of STS in a rat model of CIN. Twenty-five male Sprague Dawley rats were randomly assigned to the following groups: HC: Healthy control (received 10 mL/kg/day of 0.9% saline intraperitoneally (ip), [n = 5]), CIN: Cisplatin (received single dose of 7 mg/kg cisplatin ip [n = 5]); CIN + PAG: Cisplatin and daily ip administration of 40 mg/kg of the H2S inhibitor, DL-propargylglycine (PAG) for 28 days (n = 5); CIN + PAG + STS: Cisplatin and daily PAG and STS (150 µM) ip injection for 28 days; CIN + STS: Cisplatin and daily STS ip administration for 28 days (n = 5). Rats in each group were kept in metabolic cages for 24 h on day 0, 14 and 29 after cisplatin administration for urine collection. Rats were then euthanized, and kidney and blood samples were collected for analysis. Histologically, CIN was characterized by glomerular and tubular injury and significant macrophage influx and tubular apoptosis, as well as markedly increased levels of plasma and renal IL-1β, IL-6 and TNF-α and impaired renal antioxidant status compared to HC rats (p < 0.001). These pathological changes were exacerbated in CIN + PAG rats and were strongly reduced in CIN + PAG + STS rats relative to CIN + PAG rats (p < 0.01), while superior renal protection was observed in CIN + STS rats. Functionally, CIN was evidenced by markedly increased levels of serum creatinine and BUN, and significantly decreased urine creatinine, renal creatinine clearance, as well as electrolyte imbalance and urinary concentrating defect in comparison with HC (p < 0.01). These functional changes worsened significantly in CIN + PAG rats (p < 0.05) but improved in CIN + PAG + STS rats, with further improvement in CIN + STS rats to levels comparable to HC rats. Mechanistically, STS increased renal and plasma levels of H2S, arginine, cAMP, nitric oxide (NO) and cGMP as well as SIRT3 and PGC-1α. We have shown for the first time that STS provides chemoprotection against CIN by activating renal arginine/cAMP and NO/cGMP signaling pathways and their downstream mechanisms through increased renal H2S production. Full article
Show Figures

Figure 1

Figure 1
<p>Histopathology and quantification of kidney tissue sections. (<b>A</b>) Representative images of kidney sections showing H and E, KIM-1, ED-1 and TUNEL stains. (<b>B</b>–<b>F</b>) Quantification of immune (histochemical) staining of (<b>B</b>) H and E, (<b>C</b>) KIM-1, (<b>D</b>) ED-1, (<b>E</b>) TUNEL and (<b>F</b>) glomerular size. HC = Healthy control group; CIN = Cisplatin group; CIN + PAG = Cisplatin group treated with propargylglycine; CIN + PAG + STS = Cisplatin group treated with propargylglycine and sodium thiosulfate; CIN + STS = Cisplatin group treated with sodium thiosulfate. All images were taken at 40× magnification. Values are mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>Parameters of kidney function. Levels of (<b>A</b>) serum creatinine, (<b>B</b>) blood urea nitrogen (BUN), (<b>C</b>) plasma Na<sup>+</sup>, (<b>D</b>) plasma K<sup>+</sup>, (<b>E</b>) urine creatinine, (<b>F</b>) creatinine clearance on day 1 after cisplatin administration, (<b>G</b>) creatinine clearance on day 15 after cisplatin administration, (<b>H</b>) creatinine clearance on day 30 after cisplatin administration, (<b>I</b>) relative kidney weight, (<b>J</b>) change in body weight, and (<b>K</b>) urinary albumin. HC = Healthy control group; CIN = Cisplatin group; CIN + PAG = Cisplatin group treated with propargylglycine; CIN + PAG + STS = Cisplatin group treated with propargylglycine and sodium thiosulfate; CIN + STS = Cisplatin group treated with sodium thiosulfate. All images were taken at 40× magnification. Values are mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Water intake and urine output. Volumes of (<b>A</b>) water intake on day 1 after cisplatin administration, (<b>B</b>) water intake on day 15 after cisplatin administration, (<b>C</b>) water intake on day 30 after cisplatin administration, (<b>D</b>) urine output on day 1 after cisplatin administration, (<b>E</b>) urine output on day 15 after cisplatin administration, and (<b>F</b>) urine output on day 30 after cisplatin administration. HC = Healthy control group; CIN = Cisplatin group; CIN + PAG = Cisplatin group treated with propargylglycine; CIN + PAG + STS = Cisplatin group treated with propargylglycine and sodium thiosulfate; CIN + STS = Cisplatin group treated with sodium thiosulfate. All images were taken at 40× magnification. Values are mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Anti-inflammatory and antioxidant effects of STS. Levels of (<b>A</b>) plasma interleukin-1beta (IL-1β), (<b>B</b>) plasma interleukin-6 (IL-6), (<b>C</b>) plasma tumor necrosis factor-alpha (TNF-α), (<b>D</b>) renal IL-1β, (<b>E</b>) renal IL-6, (<b>F</b>) renal TNF-α, (<b>G</b>) renal malondialdehyde (MDA), (<b>H</b>) renal glutathione (GSH), and (<b>I</b>) renal superoxide dismutase (SOD). HC = Healthy control group; CIN = Cisplatin group; CIN + PAG = Cisplatin group treated with propargylglycine; CIN + PAG + STS = Cisplatin group treated with propargylglycine and sodium thiosulfate; CIN + STS = Cisplatin group treated with sodium thiosulfate. All images were taken at 40× magnification. Values are mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Mechanism of STS chemoprotection. (<b>A</b>) Plasma H<sub>2</sub>S level, (<b>B</b>) renal H<sub>2</sub>S content, (<b>C</b>) plasma arginine level, (<b>D</b>) renal cyclic adenosine monophosphate (cAMP) content, (<b>E</b>) plasma nitric oxide (NO) level, and (<b>F</b>) renal cyclic guanosine monophosphate (cGMP) content. Immunohistochemical staining (<b>G</b>), and quantification of (<b>H</b>) SIRT3 and (<b>I</b>) PGC-1α. HC = Healthy control group; CIN = Cisplatin group; CIN + PAG = Cisplatin group treated with propargylglycine; CIN + PAG + STS = Cisplatin group treated with propargylglycine and sodium thiosulfate; CIN + STS = Cisplatin group treated with sodium thiosulfate. All images were taken at 40× magnification. Values are mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
11 pages, 1375 KiB  
Article
Azole Antifungal Consumption in Community Pharmacy Sales in Mainland Portugal: Trend Analysis from 2014 to 2023
by Sofia Moura, Paulo Duarte, Ana Sofia Oliveira, José Martinez-de-Oliveira, Ana Palmeira-de-Oliveira and Joana Rolo
Antibiotics 2025, 14(1), 33; https://doi.org/10.3390/antibiotics14010033 - 4 Jan 2025
Viewed by 395
Abstract
Background/Objectives: Excessive or inadequate use of antimicrobial drugs may lead to the emergence of resistant strains. For this reason, it is important to monitor consumption indicators to assess drugs’ utilization over time. This study aimed to analyze the consumption of medically prescribed [...] Read more.
Background/Objectives: Excessive or inadequate use of antimicrobial drugs may lead to the emergence of resistant strains. For this reason, it is important to monitor consumption indicators to assess drugs’ utilization over time. This study aimed to analyze the consumption of medically prescribed azole antifungal drugs in mainland Portugal from 2014 to 2023, focusing on those directed to genital infections: fluconazole, isoconazole, itraconazole, and sertaconazole. Methods: For each drug, the evaluated parameters were the total number of packages, number of packages per 1000 inhabitants, defined daily dose (DDD) per 1000 inhabitants per day, and total costs. For this purpose, we used data from community pharmacies’ sales, which are available through INFARMED (the Portuguese national authority on medicines and health products). Results: Several trends emerged from data analysis. The COVID-19 pandemic negatively affected the consumption of all azole antifungal drugs included in this study. However, after 2020, fluconazole and sertaconazole consumption has been increasing. In the specific case of fluconazole, there was an increase in expenditure, although the total number of packages suffered a decrease over the 10-year study period. Additionally, the defined daily dose (DDD) per 1000 inhabitants per day for fluconazole and itraconazole was lower compared to estimates from the last available survey (2009). Conclusions: Although our findings represent a lesser pressure on fungi, further monitoring is needed to better understand the evolution of fluconazole and itraconazole consumption over time, particularly due to the trends observed in this study. Full article
(This article belongs to the Special Issue The Worldwide Problem of Antifungal Resistance: From Basic to Clinic)
Show Figures

Figure 1

Figure 1
<p>Consumption of fluconazole, isoconazole, itraconazole, and sertaconazole by number of packages and by Regional Health Administration (<b>A</b>—North, <b>B</b>—Centre, <b>C</b>—Lisbon and Tagus Valley, <b>D</b>—Alentejo, and <b>E</b>—Algarve) between 2014 and 2023.</p>
Full article ">Figure 2
<p>Consumption of fluconazole, isoconazole, itraconazole, and sertaconazole by number of packages per 1000 inhabitants in community pharmacies in mainland Portugal between 2014 and 2023.</p>
Full article ">Figure 3
<p>Total costs of fluconazole, isoconazole, itraconazole, and sertaconazole from community pharmacies’ sales in mainland Portugal between 2014 and 2023.</p>
Full article ">
Back to TopTop