Pub Date : 2025-11-11DOI: 10.3390/pharmaceutics17111454
Yeonhee Jeong, Jaejeong Kim, Changyu Kang, Yunjin Jung
Background/Objectives: Probenecid (PBN) is a uricosuric agent that facilitates the excretion of uric acid and is used to treat gout. Here, a colon-targeted prodrug of PBN was designed to facilitate repositioning as a treatment for inflammatory bowel disease (IBD). Methods: The carboxylic group in PBN was amide-conjugated with the amine groups of acidic amino acids to yield aspartic acid-conjugated PBN (PBN-AA) and glutamic acid-conjugated PBN (PBN-GA). Conjugation with amino acids increased the hydrophilicity of PBN and decreased cell permeability across the Caco-2 cell monolayer. While remaining intact in buffers (pH 1.2, 6.8) and in the small intestinal contents of rats, the conjugates were cleaved to release PBN from the cecal contents of rats, with a significant difference in the maximal conversion percentage between PBN-AA (12%) and PBN-GA (74%). Results: Upon oral gavage, PBN-GA accumulated a much greater amount of PBN in the cecum than PBN alone, thus verifying the in vitro colon specificity of PBN-GA. Oral PBN-GA enhanced the anticolitis effectiveness in dinitrobenzene sulfonic acid-induced rat colitis and limited the systemic absorption of PBN, thus reducing the risk of systemic adverse effects ascribed to PBN. Moreover, PBN-GA therapeutically surpassed sulfasalazine, a currently used anti-IBD drug, in rat colitis. Conclusions: These results suggest that amide conjugation with GA can be used to design a colon-targeting prodrug for PBN. Colon-targeted PBN may not only enhance therapeutic effectiveness but also improve the safety of PBN repositioned for the treatment of IBD and may be a pharmacological alternative for current small-molecule anti-IBD drugs with low efficacy or serious adverse effects with long-term use.
{"title":"Colon-Specific Delivery of Probenecid Enhances Therapeutic Activity of the Uricosuric Agent Against Rat Colitis.","authors":"Yeonhee Jeong, Jaejeong Kim, Changyu Kang, Yunjin Jung","doi":"10.3390/pharmaceutics17111454","DOIUrl":"https://doi.org/10.3390/pharmaceutics17111454","url":null,"abstract":"<p><p><b>Background/Objectives</b>: Probenecid (PBN) is a uricosuric agent that facilitates the excretion of uric acid and is used to treat gout. Here, a colon-targeted prodrug of PBN was designed to facilitate repositioning as a treatment for inflammatory bowel disease (IBD). <b>Methods</b>: The carboxylic group in PBN was amide-conjugated with the amine groups of acidic amino acids to yield aspartic acid-conjugated PBN (PBN-AA) and glutamic acid-conjugated PBN (PBN-GA). Conjugation with amino acids increased the hydrophilicity of PBN and decreased cell permeability across the Caco-2 cell monolayer. While remaining intact in buffers (pH 1.2, 6.8) and in the small intestinal contents of rats, the conjugates were cleaved to release PBN from the cecal contents of rats, with a significant difference in the maximal conversion percentage between PBN-AA (12%) and PBN-GA (74%). <b>Results</b>: Upon oral gavage, PBN-GA accumulated a much greater amount of PBN in the cecum than PBN alone, thus verifying the in vitro colon specificity of PBN-GA. Oral PBN-GA enhanced the anticolitis effectiveness in dinitrobenzene sulfonic acid-induced rat colitis and limited the systemic absorption of PBN, thus reducing the risk of systemic adverse effects ascribed to PBN. Moreover, PBN-GA therapeutically surpassed sulfasalazine, a currently used anti-IBD drug, in rat colitis. <b>Conclusions</b>: These results suggest that amide conjugation with GA can be used to design a colon-targeting prodrug for PBN. Colon-targeted PBN may not only enhance therapeutic effectiveness but also improve the safety of PBN repositioned for the treatment of IBD and may be a pharmacological alternative for current small-molecule anti-IBD drugs with low efficacy or serious adverse effects with long-term use.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"17 11","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12655446/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145637197","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-11DOI: 10.3390/pharmaceutics17111452
Su Guan, Mei-Juan Tu, Ai-Ming Yu
Integrated pharmacokinetic (PK) and pharmacodynamic (PD) models are essential for the understanding of quantitative relationship between drug exposure and response towards the identification of optimal dosing regimens in drug development and clinical therapy. This article summarizes the common PK-PD models being established in oncology, with a focus on combination therapies. Among them, the PK models include those used for practical non-compartmental and compartmental analyses, as well as those for physiologically based modeling that describe and predict exposure to various chemotherapy, targeted therapy, and immunotherapy drugs. Built on proper natural disease progression models, such as the empirical logistic growth curve, the Gompertzian growth model, and their modifications, the integrated PK-PD models recapitulate and predict antitumor drug efficacy, in which the PD models include practical indirect response model and various tumor growth inhibition models, as driven by the mechanistic actions of the drugs administered. Since anticancer drugs are usually co-administered, PK-PD modeling has been extended from monotherapy to combination therapy. However, relying on a single interaction factor or parameter to capitulate complex drug interactions, predict outcomes of different combinations, and determine possible synergism is problematic. Considering the apparent contributions from individual drugs following mutual interactions, a new PK-PD model has been developed for combination therapy, which may be integrated with proper algorism (e.g., the Combination Index method) to critically define combination effects, synergism, additivity, or antagonism. As drug combinations become more complex and individual drug actions are variable, these models should be optimized further to advance the understanding of PK-PD relationships and facilitate the development of improved therapies.
{"title":"Practical Pharmacokinetic-Pharmacodynamic Models in Oncology.","authors":"Su Guan, Mei-Juan Tu, Ai-Ming Yu","doi":"10.3390/pharmaceutics17111452","DOIUrl":"https://doi.org/10.3390/pharmaceutics17111452","url":null,"abstract":"<p><p>Integrated pharmacokinetic (PK) and pharmacodynamic (PD) models are essential for the understanding of quantitative relationship between drug exposure and response towards the identification of optimal dosing regimens in drug development and clinical therapy. This article summarizes the common PK-PD models being established in oncology, with a focus on combination therapies. Among them, the PK models include those used for practical non-compartmental and compartmental analyses, as well as those for physiologically based modeling that describe and predict exposure to various chemotherapy, targeted therapy, and immunotherapy drugs. Built on proper natural disease progression models, such as the empirical logistic growth curve, the Gompertzian growth model, and their modifications, the integrated PK-PD models recapitulate and predict antitumor drug efficacy, in which the PD models include practical indirect response model and various tumor growth inhibition models, as driven by the mechanistic actions of the drugs administered. Since anticancer drugs are usually co-administered, PK-PD modeling has been extended from monotherapy to combination therapy. However, relying on a single interaction factor or parameter to capitulate complex drug interactions, predict outcomes of different combinations, and determine possible synergism is problematic. Considering the apparent contributions from individual drugs following mutual interactions, a new PK-PD model has been developed for combination therapy, which may be integrated with proper algorism (e.g., the Combination Index method) to critically define combination effects, synergism, additivity, or antagonism. As drug combinations become more complex and individual drug actions are variable, these models should be optimized further to advance the understanding of PK-PD relationships and facilitate the development of improved therapies.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"17 11","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12655065/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145637322","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-10DOI: 10.3390/pharmaceutics17111451
Cristina Maderuelo, Roberto Arévalo-Pérez, José M Lanao
Introduction: Drug development has traditionally used mathematical models to predict formulation behavior. Objective: Building artificial neural networks for the drug release evaluation of drug delivery systems using sustained-release metronidazole-coated colonic hydrophilic matrices as a model. Methods: The technological factors associated with the biopharmaceutical performance of hydrophilic metronidazole matrices were evaluated using a quality by design approach (QbD). The developed neural network includes variables related to the technological process for producing the matrices. These are related to the materials used, such as the type and viscosity of core polymers, the type of coating agent, or the matrix production process, such as the mixing time of core materials or the percentage of the coating agent. The output variables of the neural network were the percentages of drug released in vitro at 1, 6, 12, and 24 h and the mean dissolution time of the matrix. An iterative quasi-Newton method was used to train the artificial neural network. Results: A neural network with excellent prediction capacity allows selecting the technological variables with the greatest influence on the % of drug dissolved: the type of coating agent used and the percentage of the total weight increase after coating for 1 h and 6 h of drug release and also the viscosity of the HPMC for 12 and 24 h. Conclusions: The optimized neural network demonstrated an excellent predictive capacity for in vitro drug dissolution profiles, allowing the use of this type of methodology based on artificial intelligence methods in the optimization of drug delivery systems.
{"title":"Building Artificial Neural Networks for the Optimization of Sustained-Release Kinetics of Metronidazole from Colonic Hydrophilic Matrices.","authors":"Cristina Maderuelo, Roberto Arévalo-Pérez, José M Lanao","doi":"10.3390/pharmaceutics17111451","DOIUrl":"https://doi.org/10.3390/pharmaceutics17111451","url":null,"abstract":"<p><p><b>Introduction</b>: Drug development has traditionally used mathematical models to predict formulation behavior. <b>Objective</b>: Building artificial neural networks for the drug release evaluation of drug delivery systems using sustained-release metronidazole-coated colonic hydrophilic matrices as a model. <b>Methods</b>: The technological factors associated with the biopharmaceutical performance of hydrophilic metronidazole matrices were evaluated using a quality by design approach (QbD). The developed neural network includes variables related to the technological process for producing the matrices. These are related to the materials used, such as the type and viscosity of core polymers, the type of coating agent, or the matrix production process, such as the mixing time of core materials or the percentage of the coating agent. The output variables of the neural network were the percentages of drug released in vitro at 1, 6, 12, and 24 h and the mean dissolution time of the matrix. An iterative quasi-Newton method was used to train the artificial neural network. <b>Results</b>: A neural network with excellent prediction capacity allows selecting the technological variables with the greatest influence on the % of drug dissolved: the type of coating agent used and the percentage of the total weight increase after coating for 1 h and 6 h of drug release and also the viscosity of the HPMC for 12 and 24 h. <b>Conclusions</b>: The optimized neural network demonstrated an excellent predictive capacity for in vitro drug dissolution profiles, allowing the use of this type of methodology based on artificial intelligence methods in the optimization of drug delivery systems.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"17 11","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12655049/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145637207","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-10DOI: 10.3390/pharmaceutics17111450
Gyu Lin Kim, Yeon Ju Kang, Soo Hwa Seo, Jiwoon Jeon, Hyo-Kyung Han
Background: Although teriparatide is efficacious, its once-daily subcutaneous injections cause local adverse events, inconvenience, and higher cost, limiting long-term adherence. Therefore, this research aims to engineer a pH-responsive oral formulation of teriparatide for osteoporosis therapy. Methods: A layered silicate nanocomplex was obtained by spontaneous self-assembly of teriparatide (Teri) with 3-aminopropyl magnesium phyllosilicate (AC). The nanocomplex (AC-Teri) was then coated with a 1:1 blend of two polymethacrylic acid derivatives (Eudragit® L100 and Eudragit® S 100) to provide pH-triggered drug release along the gastrointestinal tract. Results: AC-Teri and the coated nanocomplex (EE/AC-Teri) displayed high encapsulation efficiency (>90%) with narrow size distributions. In a stepwise buffer transition system, EE/AC-Teri demonstrated pH-dependent release, with less than 25% drug liberated at pH 1.2, approximately 54% at pH 6.8, and 74% at pH 7.4 over 24 h. Particle size and ζ-potential of EE/AC-Teri shifted in parallel with dissolution of the outer polymer shell. EE/AC-Teri also protected the peptide against enzymatic degradation, preserving the secondary structure of encapsulated teriparatide in simulated intestinal fluids. Compared with free drug, EE/AC-Teri enhanced transcellular drug permeation 2.7-fold in Caco-2 cells. In dexamethasone-induced osteoporotic rats, oral EE/AC-Teri significantly stimulated bone formation while suppressing resorption; micro-CT and histology confirmed recovery of trabecular architecture. Conclusions: EE/AC-Teri represents a promising oral teriparatide formulation for the effective management of osteoporosis.
{"title":"Effective Oral Delivery of Teriparatide Using Organoclay-Polymethacrylate Nanocomposites for Osteoporosis Therapy.","authors":"Gyu Lin Kim, Yeon Ju Kang, Soo Hwa Seo, Jiwoon Jeon, Hyo-Kyung Han","doi":"10.3390/pharmaceutics17111450","DOIUrl":"https://doi.org/10.3390/pharmaceutics17111450","url":null,"abstract":"<p><p><b>Background</b>: Although teriparatide is efficacious, its once-daily subcutaneous injections cause local adverse events, inconvenience, and higher cost, limiting long-term adherence. Therefore, this research aims to engineer a pH-responsive oral formulation of teriparatide for osteoporosis therapy. <b>Methods</b>: A layered silicate nanocomplex was obtained by spontaneous self-assembly of teriparatide (Teri) with 3-aminopropyl magnesium phyllosilicate (AC). The nanocomplex (AC-Teri) was then coated with a 1:1 blend of two polymethacrylic acid derivatives (Eudragit<sup>®</sup> L100 and Eudragit<sup>®</sup> S 100) to provide pH-triggered drug release along the gastrointestinal tract. <b>Results</b>: AC-Teri and the coated nanocomplex (EE/AC-Teri) displayed high encapsulation efficiency (>90%) with narrow size distributions. In a stepwise buffer transition system, EE/AC-Teri demonstrated pH-dependent release, with less than 25% drug liberated at pH 1.2, approximately 54% at pH 6.8, and 74% at pH 7.4 over 24 h. Particle size and ζ-potential of EE/AC-Teri shifted in parallel with dissolution of the outer polymer shell. EE/AC-Teri also protected the peptide against enzymatic degradation, preserving the secondary structure of encapsulated teriparatide in simulated intestinal fluids. Compared with free drug, EE/AC-Teri enhanced transcellular drug permeation 2.7-fold in Caco-2 cells. In dexamethasone-induced osteoporotic rats, oral EE/AC-Teri significantly stimulated bone formation while suppressing resorption; micro-CT and histology confirmed recovery of trabecular architecture. <b>Conclusions</b>: EE/AC-Teri represents a promising oral teriparatide formulation for the effective management of osteoporosis.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"17 11","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12655526/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145637297","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-10DOI: 10.3390/pharmaceutics17111449
Hyo-Geun Lee, Habaragoda Dewage Tharushi Udayangani Wijerathne, Taeho Kim, Si-Hyeong Park, Won-Kyo Jung, Jae-Young Oh, Mi-Jin Yim, Jeong Min Lee, Seok-Chun Ko, Dae-Sung Lee, Hyun-Soo Kim
Background/Objectives: Marine-derived bioactive peptides have been reported to possess blood pressure-regulatory effects. However, most studies have focused on the antihypertensive effects after single-dose administration, and research on long-term administration and its protective effects against hypertension-induced tissue damage remains limited. Therefore, this study aimed to investigate the long-term antihypertensive efficacy of IGTGIPGIW, a bioactive peptide derived from Hippocampus abdominalis (H. abdominalis), and its protective effects on hypertension-related tissue damage. Methods: To evaluate the blood pressure-regulatory effects, spontaneously hypertensive rats (SHRs) were orally administered a high-dose (50 mg/kg) IGTGIPGIW peptide group (H-IGTGIPGIW) for 8 weeks. Systolic blood pressure (SBP), diastolic blood pressure (DBP), and mean arterial pressure (MAP) were monitored weekly. Serum levels of angiotensin II (Ang II), angiotensin-converting enzyme (ACE), and angiotensin-converting enzyme 2 (ACE2) were measured to assess the peptide's regulatory effects on the renin-angiotensin system. Histological analyses of the aorta and heart tissues were performed to evaluate the protective effects against hypertension-induced tissue damage. Results: After 8 weeks of treatment, H-IGTGIPGIW significantly reduced SBP, DBP, and MAP compared with SHRs. Serum Ang II and ACE levels were significantly decreased, while ACE2 levels were significantly increased. Histological analyses demonstrated that IGTGIPGIW alleviated aortic wall thickening and reduced renal and cardiac tissue damage in SHR. Conclusions: IGTGIPGIW, a bioactive peptide derived from H. abdominalis, effectively regulated blood pressure by modulating serum Ang II, ACE, and ACE2 levels. Moreover, it protected against hypertension-induced aortic, renal and cardiac tissue damage, suggesting its potential as a functional ingredient for managing hypertension.
{"title":"Therapeutic Potential of Big-Belly Seahorse Derived Peptide in Blood Pressure Regulation and Protection Against Aortic, Renal, and Cardiac Injuries on Spontaneously Hypertensive Rats.","authors":"Hyo-Geun Lee, Habaragoda Dewage Tharushi Udayangani Wijerathne, Taeho Kim, Si-Hyeong Park, Won-Kyo Jung, Jae-Young Oh, Mi-Jin Yim, Jeong Min Lee, Seok-Chun Ko, Dae-Sung Lee, Hyun-Soo Kim","doi":"10.3390/pharmaceutics17111449","DOIUrl":"https://doi.org/10.3390/pharmaceutics17111449","url":null,"abstract":"<p><p><b>Background/Objectives:</b> Marine-derived bioactive peptides have been reported to possess blood pressure-regulatory effects. However, most studies have focused on the antihypertensive effects after single-dose administration, and research on long-term administration and its protective effects against hypertension-induced tissue damage remains limited. Therefore, this study aimed to investigate the long-term antihypertensive efficacy of IGTGIPGIW, a bioactive peptide derived from <i>Hippocampus abdominalis</i> (<i>H. abdominalis</i>), and its protective effects on hypertension-related tissue damage. <b>Methods:</b> To evaluate the blood pressure-regulatory effects, spontaneously hypertensive rats (SHRs) were orally administered a high-dose (50 mg/kg) IGTGIPGIW peptide group (H-IGTGIPGIW) for 8 weeks. Systolic blood pressure (SBP), diastolic blood pressure (DBP), and mean arterial pressure (MAP) were monitored weekly. Serum levels of angiotensin II (Ang II), angiotensin-converting enzyme (ACE), and angiotensin-converting enzyme 2 (ACE2) were measured to assess the peptide's regulatory effects on the renin-angiotensin system. Histological analyses of the aorta and heart tissues were performed to evaluate the protective effects against hypertension-induced tissue damage. <b>Results:</b> After 8 weeks of treatment, H-IGTGIPGIW significantly reduced SBP, DBP, and MAP compared with SHRs. Serum Ang II and ACE levels were significantly decreased, while ACE2 levels were significantly increased. Histological analyses demonstrated that IGTGIPGIW alleviated aortic wall thickening and reduced renal and cardiac tissue damage in SHR. <b>Conclusions:</b> IGTGIPGIW, a bioactive peptide derived from <i>H. abdominalis</i>, effectively regulated blood pressure by modulating serum Ang II, ACE, and ACE2 levels. Moreover, it protected against hypertension-induced aortic, renal and cardiac tissue damage, suggesting its potential as a functional ingredient for managing hypertension.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"17 11","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12654982/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145637391","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background/Objectives: Propofol is frequently used as an intravenous anesthetic and is rapidly metabolized. Therefore, if it could be administered non-invasively (e.g., orally) as premedication, it might hasten emergence from anesthesia, thereby improving patient safety. However, it undergoes extensive first-pass metabolism in the liver and intestines, limiting the route for premedication. We evaluated whether intranasal delivery of a propofol-encapsulated liposome solution improves systemic exposure and bioavailability in rabbits. Methods: A propofol-encapsulated liposome solution was administered to rabbits via the intravenous, oral, and intranasal routes. Blood propofol concentrations were measured for up to 60 min after administration and the area under the concentration-time curve (AUC0-60) and bioavailability of the propofol-encapsulated liposome solution were compared with those of the non-encapsulated propofol formulation. The differences were tested by two-way analysis of variance (ANOVA) with Šidák's post hoc multiple-comparisons test and the Mann-Whitney test (α = 0.05). Results: The AUC0-60 for blood propofol concentrations after intravenous administration was significantly higher with the propofol-encapsulated liposome solution than with the non-encapsulated propofol formulation (3038.8 ± 661.5 vs. 1929.8 ± 58.2 ng·min/mL; p = 0.0286). By contrast, no increase in blood propofol concentrations was observed after oral administration, whereas intranasal administration increased blood propofol concentrations and yielded significantly higher bioavailability compared with the non-encapsulated propofol formulation (16.4 ± 7.3% vs. 2.0 ± 1.2%; p = 0.0286). Conclusions: The findings of the present study suggest that intranasal liposomal propofol increased systemic availability compared with a non-encapsulated formulation, supporting further evaluation as a candidate premedication approach for propofol.
背景/目的:异丙酚常被用作静脉麻醉剂,代谢迅速。因此,如果它可以无创地(例如口服)作为前用药,它可能会加速麻醉的恢复,从而提高患者的安全性。然而,它在肝脏和肠道中进行广泛的首过代谢,限制了预用药的途径。我们评估了异丙酚包封脂质体溶液是否可以改善兔的全身暴露和生物利用度。方法:采用异丙酚包封脂质体溶液经静脉、口服和鼻内给药。测定给药后60 min血液中异丙酚浓度,并比较异丙酚包封脂质体溶液与未包封的异丙酚制剂的浓度-时间曲线下面积(AUC0-60)和生物利用度。采用Šidák事后多重比较检验和Mann-Whitney检验的双向方差分析(ANOVA)进行差异检验(α = 0.05)。结果:静脉给药后异丙酚血药浓度AUC0-60值异丙酚包封脂质体溶液明显高于非包封异丙酚制剂(3038.8±661.5∶1929.8±58.2 ng·min/mL; p = 0.0286)。相比之下,口服给药后血液中异丙酚浓度未增加,而鼻内给药后血液中异丙酚浓度增加,生物利用度显著高于非胶囊化异丙酚制剂(16.4±7.3% vs. 2.0±1.2%;p = 0.0286)。结论:本研究的结果表明,与非胶囊化制剂相比,鼻内丙泊酚脂质体增加了全身可用性,支持进一步评估作为丙泊酚的候选用药前方法。
{"title":"Development of Propofol-Encapsulated Liposomes and the Effect of Intranasal Administration on Bioavailability in Rabbits.","authors":"Hitomi Ujita, Hitoshi Higuchi, Yukiko Nishioka, Saki Miyake, Riko Sato, Takuya Miyawaki","doi":"10.3390/pharmaceutics17111446","DOIUrl":"https://doi.org/10.3390/pharmaceutics17111446","url":null,"abstract":"<p><p><b>Background/Objectives:</b> Propofol is frequently used as an intravenous anesthetic and is rapidly metabolized. Therefore, if it could be administered non-invasively (e.g., orally) as premedication, it might hasten emergence from anesthesia, thereby improving patient safety. However, it undergoes extensive first-pass metabolism in the liver and intestines, limiting the route for premedication. We evaluated whether intranasal delivery of a propofol-encapsulated liposome solution improves systemic exposure and bioavailability in rabbits. <b>Methods:</b> A propofol-encapsulated liposome solution was administered to rabbits via the intravenous, oral, and intranasal routes. Blood propofol concentrations were measured for up to 60 min after administration and the area under the concentration-time curve (AUC<sub>0-60</sub>) and bioavailability of the propofol-encapsulated liposome solution were compared with those of the non-encapsulated propofol formulation. The differences were tested by two-way analysis of variance (ANOVA) with Šidák's post hoc multiple-comparisons test and the Mann-Whitney test (α = 0.05). <b>Results:</b> The AUC<sub>0-60</sub> for blood propofol concentrations after intravenous administration was significantly higher with the propofol-encapsulated liposome solution than with the non-encapsulated propofol formulation (3038.8 ± 661.5 vs. 1929.8 ± 58.2 ng·min/mL; <i>p</i> = 0.0286). By contrast, no increase in blood propofol concentrations was observed after oral administration, whereas intranasal administration increased blood propofol concentrations and yielded significantly higher bioavailability compared with the non-encapsulated propofol formulation (16.4 ± 7.3% vs. 2.0 ± 1.2%; <i>p</i> = 0.0286). <b>Conclusions:</b> The findings of the present study suggest that intranasal liposomal propofol increased systemic availability compared with a non-encapsulated formulation, supporting further evaluation as a candidate premedication approach for propofol.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"17 11","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12655750/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145637212","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-09DOI: 10.3390/pharmaceutics17111447
Joonhyuck Park, Tae-Ryong Riew
Ischemic stroke remains a major cause of mortality and long-term disability, yet current therapeutic strategies are largely limited to reperfusion approaches such as intravenous thrombolysis and thrombectomy, which are constrained by narrow treatment windows and the risk of complications. Moreover, the blood-brain barrier (BBB) severely restricts drug penetration into the injured brain, limiting the translation of promising neuroprotective agents into clinical success. Intranasal (IN) delivery has emerged as a compelling alternative route that bypasses the BBB and enables rapid access to the central nervous system through olfactory, trigeminal, and perivascular pathways. This narrative review highlights recent advances in preclinical research on IN therapeutics for ischemic stroke, ranging from small molecules and biologics to nucleic acids and cell-based therapies. Particular emphasis is placed on the application of nanotechnology, including extracellular vesicles, liposomes, and inorganic nanoparticles, which enhance drug stability, targeting, and bioavailability. Studies demonstrate that IN delivery of growth factors, cytokines, and engineered stem cells can promote neurogenesis, angiogenesis, white matter repair, and functional recovery, while nanocarriers further expand the therapeutic potential. Overall, intranasal delivery represents a promising and non-invasive strategy to overcome the limitations of conventional stroke therapies, offering new avenues for neuroprotection and regeneration that warrant further investigation toward clinical translation.
{"title":"Nanoparticle-Mediated Nose-to-Brain Delivery for Ischemic Stroke Therapy: Preclinical Insights.","authors":"Joonhyuck Park, Tae-Ryong Riew","doi":"10.3390/pharmaceutics17111447","DOIUrl":"https://doi.org/10.3390/pharmaceutics17111447","url":null,"abstract":"<p><p>Ischemic stroke remains a major cause of mortality and long-term disability, yet current therapeutic strategies are largely limited to reperfusion approaches such as intravenous thrombolysis and thrombectomy, which are constrained by narrow treatment windows and the risk of complications. Moreover, the blood-brain barrier (BBB) severely restricts drug penetration into the injured brain, limiting the translation of promising neuroprotective agents into clinical success. Intranasal (IN) delivery has emerged as a compelling alternative route that bypasses the BBB and enables rapid access to the central nervous system through olfactory, trigeminal, and perivascular pathways. This narrative review highlights recent advances in preclinical research on IN therapeutics for ischemic stroke, ranging from small molecules and biologics to nucleic acids and cell-based therapies. Particular emphasis is placed on the application of nanotechnology, including extracellular vesicles, liposomes, and inorganic nanoparticles, which enhance drug stability, targeting, and bioavailability. Studies demonstrate that IN delivery of growth factors, cytokines, and engineered stem cells can promote neurogenesis, angiogenesis, white matter repair, and functional recovery, while nanocarriers further expand the therapeutic potential. Overall, intranasal delivery represents a promising and non-invasive strategy to overcome the limitations of conventional stroke therapies, offering new avenues for neuroprotection and regeneration that warrant further investigation toward clinical translation.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"17 11","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12655175/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145637402","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-09DOI: 10.3390/pharmaceutics17111448
Chung-Hui Huang, Xuejia Kang, Lang Zhou, Junwei Wang, Shuai Wu, Peizhen Sun, Qi Wang, Adam B Keeton, Pengyu Chen, Gary A Piazza
Background/Objectives: Triple-negative breast cancer (TNBC) is among the most aggressive subtypes, lacking estrogen, progesterone, and HER2 receptors, which limits the efficacy of targeted therapies. Standard treatments often fail due to rapid drug resistance and poor long-term outcomes. Repurposing approved drugs with anticancer potential offers a promising alternative. Disulfiram (DSF), an FDA-approved alcohol-aversion drug, forms a copper complex [Cu(DDC)2] with potent anticancer activity, but its clinical translation is hindered by poor solubility, limited stability, and inefficient delivery. Methods: Here, we present an amphiphilic dendrimer-stabilized [Cu(DDC)2] nanoparticle (NP) platform synthesized via the stabilized metal ion ligand complex (SMILE) method. Results: The optimized nanocarrier achieved high encapsulation efficiency, enhanced serum stability, and potent cytotoxicity against TNBC cells. It induced immunogenic cell death (ICD) characterized by calreticulin exposure and ATP release, while modulating the tumor microenvironment by downregulating MMP-3, MMP-9, VEGF, and vimentin, and restoring epithelial markers. In a 4T1 TNBC mouse model, systemic [Cu(DDC)2] NP treatment significantly inhibited tumor growth without combinational chemo- or radiotherapy. Conclusions: This DSF-based metal-organic NP integrates drug repurposing, immune activation, and tumor microenvironment remodeling into a single platform, offering strong translational potential for treating aggressive breast cancers.
{"title":"Novel Disulfiram-Loaded Metal-Organic Nanoparticles Inhibit Tumor Growth and Induce Immunogenic Cell Death of Triple-Negative Breast Cancer Cells.","authors":"Chung-Hui Huang, Xuejia Kang, Lang Zhou, Junwei Wang, Shuai Wu, Peizhen Sun, Qi Wang, Adam B Keeton, Pengyu Chen, Gary A Piazza","doi":"10.3390/pharmaceutics17111448","DOIUrl":"https://doi.org/10.3390/pharmaceutics17111448","url":null,"abstract":"<p><p><b>Background/Objectives:</b> Triple-negative breast cancer (TNBC) is among the most aggressive subtypes, lacking estrogen, progesterone, and HER2 receptors, which limits the efficacy of targeted therapies. Standard treatments often fail due to rapid drug resistance and poor long-term outcomes. Repurposing approved drugs with anticancer potential offers a promising alternative. Disulfiram (DSF), an FDA-approved alcohol-aversion drug, forms a copper complex [Cu(DDC)<sub>2</sub>] with potent anticancer activity, but its clinical translation is hindered by poor solubility, limited stability, and inefficient delivery. <b>Methods:</b> Here, we present an amphiphilic dendrimer-stabilized [Cu(DDC)<sub>2</sub>] nanoparticle (NP) platform synthesized via the stabilized metal ion ligand complex (SMILE) method. <b>Results:</b> The optimized nanocarrier achieved high encapsulation efficiency, enhanced serum stability, and potent cytotoxicity against TNBC cells. It induced immunogenic cell death (ICD) characterized by calreticulin exposure and ATP release, while modulating the tumor microenvironment by downregulating MMP-3, MMP-9, VEGF, and vimentin, and restoring epithelial markers. In a 4T1 TNBC mouse model, systemic [Cu(DDC)<sub>2</sub>] NP treatment significantly inhibited tumor growth without combinational chemo- or radiotherapy. <b>Conclusions:</b> This DSF-based metal-organic NP integrates drug repurposing, immune activation, and tumor microenvironment remodeling into a single platform, offering strong translational potential for treating aggressive breast cancers.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"17 11","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12655402/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145637397","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-08DOI: 10.3390/pharmaceutics17111444
Husam M Younes, AlSayed A Sallam, Loai Ahmad Saifan, Aya M Ghanem, Enam A Khalil, Ehab A Abu-Basha, Ahmad Y Abuhelwa
Background/Objectives: Breast cancer remains the most prevalent malignancy among women worldwide, with letrozole (LZ) serving as a critical aromatase inhibitor for hormone receptor-positive cases. However, long-term oral administration of LZ is often associated with systemic adverse effects and poor patient compliance. To overcome these limitations, new non-aqueous nanoemulgels (NEMGs) were developed for transdermal delivery of LZ. Methods: The NEMGs were formulated using glyceryl monooleate (GMO), Sepineo P600®, Transcutol, propylene glycol, and penetration enhancers propylene glycol laurate (PGL), propylene glycol monocaprylate (PGMC), and Captex®. Physicochemical characterization, solubility, stability, and in vitro permeation studies were conducted using Strat-M® membranes, while in vivo pharmacokinetics were evaluated in rat models. Results: The optimized GMO/PGMC-based NEMG demonstrated significantly enhanced drug flux, higher permeability coefficients, and shorter lag times compared with other NEMGs and suspension emulgels. In vivo, transdermal application of the GMO/PGMC-based NEMG over an area of 2.55 cm2 produced dual plasma absorption peaks, with 57% of the LZ dose absorbed relative to oral administration over 12 days. Shelf-life and accelerated stability assessments confirmed excellent physicochemical stability with negligible crystallization. Conclusions: The developed LZ NEMG formulations offer a stable, effective, and patient-friendly transdermal drug delivery platform for breast cancer therapy. This system demonstrates potential to improve patient compliance and reduce systemic toxicity compared to conventional oral administration.
{"title":"Development and Pharmacokinetic Evaluation of Newly Formulated Letrozole Non-Aqueous Nanoemulgel Transdermal Systems for Hormone-Dependent Breast Cancer Therapy.","authors":"Husam M Younes, AlSayed A Sallam, Loai Ahmad Saifan, Aya M Ghanem, Enam A Khalil, Ehab A Abu-Basha, Ahmad Y Abuhelwa","doi":"10.3390/pharmaceutics17111444","DOIUrl":"https://doi.org/10.3390/pharmaceutics17111444","url":null,"abstract":"<p><p><b>Background/Objectives:</b> Breast cancer remains the most prevalent malignancy among women worldwide, with letrozole (LZ) serving as a critical aromatase inhibitor for hormone receptor-positive cases. However, long-term oral administration of LZ is often associated with systemic adverse effects and poor patient compliance. To overcome these limitations, new non-aqueous nanoemulgels (NEMGs) were developed for transdermal delivery of LZ. <b>Methods:</b> The NEMGs were formulated using glyceryl monooleate (GMO), Sepineo P600<sup>®</sup>, Transcutol, propylene glycol, and penetration enhancers propylene glycol laurate (PGL), propylene glycol monocaprylate (PGMC), and Captex<sup>®</sup>. Physicochemical characterization, solubility, stability, and in vitro permeation studies were conducted using Strat-M<sup>®</sup> membranes, while in vivo pharmacokinetics were evaluated in rat models. <b>Results:</b> The optimized GMO/PGMC-based NEMG demonstrated significantly enhanced drug flux, higher permeability coefficients, and shorter lag times compared with other NEMGs and suspension emulgels. In vivo, transdermal application of the GMO/PGMC-based NEMG over an area of 2.55 cm<sup>2</sup> produced dual plasma absorption peaks, with 57% of the LZ dose absorbed relative to oral administration over 12 days. Shelf-life and accelerated stability assessments confirmed excellent physicochemical stability with negligible crystallization. <b>Conclusions:</b> The developed LZ NEMG formulations offer a stable, effective, and patient-friendly transdermal drug delivery platform for breast cancer therapy. This system demonstrates potential to improve patient compliance and reduce systemic toxicity compared to conventional oral administration.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"17 11","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12655107/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145637276","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-08DOI: 10.3390/pharmaceutics17111445
Pauline Kramp, Aydin Özmaldar, Gloria Ruiz-Gómez, M Teresa Pisabarro
Background: The binding of glycosaminoglycans (GAG) to Wnt signaling components plays a key regulatory role in bone formation and regeneration. We previously reported de novo designed chemically modified hyaluronan derivatives, named REGAG (Rationally Engineered GAG), which demonstrated bone-regenerative properties in a mouse calvaria defect model. To gain initial insights into the pharmacological profile of two REGAG currently under preclinical investigation in mice, we performed a comprehensive in silico investigation of their binding to human and murine serum albumin (HSA and MSA), as it might influence their ADME properties. Furthermore, we evaluated whether REGAG binding might impact the recognition of well-characterized HSA-binding drugs. Methods: State-of-the-art in silico ADMET tools, docking and molecular dynamics simulations were used to predict and characterize the interaction of REGAG with HSA and MSA, and to investigate the molecular mechanisms involved at the atomic level. Results: The investigated REGAG molecules show a consistent binding preference for the FA1 site in both proteins, and an additional preference for the FA7 site in HSA. Their recognition might induce protein conformational changes and alter the functional state. Furthermore, REGAG's conformational adaptability is predicted to influence their binding to the FA5/6 and FA8/9 sites of HSA, and to the FA3/4 and FA7 sites of MSA. Conclusions: Our investigations predict the binding of two hyaluronan derivatives to HSA and MSA. The mechanistic insights gained into the molecular recognition of these two REGAG molecules offer valuable information for their potential clinical application and serve as a rational basis for future molecular design aimed at improving pharmacokinetic properties.
{"title":"<i>In Silico</i> Analysis of Serum Albumin Binding by Bone-Regenerative Hyaluronan-Based Molecules.","authors":"Pauline Kramp, Aydin Özmaldar, Gloria Ruiz-Gómez, M Teresa Pisabarro","doi":"10.3390/pharmaceutics17111445","DOIUrl":"https://doi.org/10.3390/pharmaceutics17111445","url":null,"abstract":"<p><p><b>Background:</b> The binding of glycosaminoglycans (GAG) to Wnt signaling components plays a key regulatory role in bone formation and regeneration. We previously reported <i>de novo</i> designed chemically modified hyaluronan derivatives, named <sub>RE</sub>GAG (Rationally Engineered GAG), which demonstrated bone-regenerative properties in a mouse calvaria defect model. To gain initial insights into the pharmacological profile of two <sub>RE</sub>GAG currently under preclinical investigation in mice, we performed a comprehensive <i>in silico</i> investigation of their binding to human and murine serum albumin (HSA and MSA), as it might influence their ADME properties. Furthermore, we evaluated whether <sub>RE</sub>GAG binding might impact the recognition of well-characterized HSA-binding drugs. <b>Methods</b>: State-of-the-art <i>in silico</i> ADMET tools, docking and molecular dynamics simulations were used to predict and characterize the interaction of <sub>RE</sub>GAG with HSA and MSA, and to investigate the molecular mechanisms involved at the atomic level. <b>Results</b>: The investigated <sub>RE</sub>GAG molecules show a consistent binding preference for the FA1 site in both proteins, and an additional preference for the FA7 site in HSA. Their recognition might induce protein conformational changes and alter the functional state. Furthermore, <sub>RE</sub>GAG's conformational adaptability is predicted to influence their binding to the FA5/6 and FA8/9 sites of HSA, and to the FA3/4 and FA7 sites of MSA. <b>Conclusions</b>: Our investigations predict the binding of two hyaluronan derivatives to HSA and MSA. The mechanistic insights gained into the molecular recognition of these two <sub>RE</sub>GAG molecules offer valuable information for their potential clinical application and serve as a rational basis for future molecular design aimed at improving pharmacokinetic properties.</p>","PeriodicalId":19894,"journal":{"name":"Pharmaceutics","volume":"17 11","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12655611/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145637553","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}