Pub Date : 2024-11-19DOI: 10.1016/j.ejps.2024.106966
Álvaro González-Garcinuño, Antonio Tabernero, Celia Nieto, Eva Martín Del Valle, Sasa Kenjeres
Several studies have recommended the use of hydrogels for localized targeted delivery of chemotherapeutic drugs following tumor removal surgery. This approach aims to both fill the cavity and prevent cancer recurrence. The use of Multiphysics-based simulation emerges as a valuable strategy for minimizing experimental work, providing detailed insights into how drug release occurs in the tissue, and enabling the optimization of the design. In this study, we introduced a mathematical model, utilizing experimental data, to investigate the transport of liposomes carrying MZ1 from a thermosensitive hydrogel and their impact on the viability of breast cancer cells. The proposed comprehensive model considers not just the transport within the interstitial tissue, represented as a porous medium, but also the uptake by cells and its influence on cell viability, along with the potential lymphatic drainage. The six real patient-specific tumor shapes extracted from MRI scans were used to investigate how the size and form of the tumor can modify the transport pattern. The computational results revealed that the concentration of liposomes in the tissue is significantly influenced by their release from the hydrogel, which proved to be the limiting step. Liposome concentrations of approximately 0.1% weight were found to be sufficient in ensuring minimal cell survival in the vicinity of the tumor.
{"title":"Multiphysics Simulation of Liposome Release from Hydrogels for Cavity Filling Following Patient-Specific Breast Tumor Surgery.","authors":"Álvaro González-Garcinuño, Antonio Tabernero, Celia Nieto, Eva Martín Del Valle, Sasa Kenjeres","doi":"10.1016/j.ejps.2024.106966","DOIUrl":"https://doi.org/10.1016/j.ejps.2024.106966","url":null,"abstract":"<p><p>Several studies have recommended the use of hydrogels for localized targeted delivery of chemotherapeutic drugs following tumor removal surgery. This approach aims to both fill the cavity and prevent cancer recurrence. The use of Multiphysics-based simulation emerges as a valuable strategy for minimizing experimental work, providing detailed insights into how drug release occurs in the tissue, and enabling the optimization of the design. In this study, we introduced a mathematical model, utilizing experimental data, to investigate the transport of liposomes carrying MZ1 from a thermosensitive hydrogel and their impact on the viability of breast cancer cells. The proposed comprehensive model considers not just the transport within the interstitial tissue, represented as a porous medium, but also the uptake by cells and its influence on cell viability, along with the potential lymphatic drainage. The six real patient-specific tumor shapes extracted from MRI scans were used to investigate how the size and form of the tumor can modify the transport pattern. The computational results revealed that the concentration of liposomes in the tissue is significantly influenced by their release from the hydrogel, which proved to be the limiting step. Liposome concentrations of approximately 0.1% weight were found to be sufficient in ensuring minimal cell survival in the vicinity of the tumor.</p>","PeriodicalId":12018,"journal":{"name":"European Journal of Pharmaceutical Sciences","volume":" ","pages":"106966"},"PeriodicalIF":4.3,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142686340","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-19DOI: 10.1016/j.ejps.2024.106967
Farnaz Shokraneh, Anne M Filppula, Aleksi Tornio, Jaan Aruväli, Urve Paaver, Niklas Sandler Topelius
The exploration of three-dimensional (3D) printing inspired technologies in pharmaceutical compounding reveals a promising frontier in personalized medicine manufacture. This study focuses on the development of clopidogrel bisulphate tablets, with doses ranging from 2 mg to 20 mg per tablet, suitable for pediatric use. The study explored a semi-solid extrusion-based deposition technology already being used in compounding pharmacies across several European locations. The investigation explored various properties of two formulations of 1% and 2% clopidogrel gel tablets, with a specific focus on mass variation, drug content uniformity, in vitro drug release profiles, disintegration time, and stability. The mean weights of the smallest printed 200 mg tablets with 1% and 2% clopidogrel concentrations were 199.1 ± 4.6 mg and 201.0 ± 3.2 mg, respectively. For the largest printed 500 mg tablets with 1% and 2% concentrations, the mean weights were 499.3 ± 7.7 mg and 501.7 ± 6.5 mg, respectively. The mean clopidogrel content uniformity for 1% clopidogrel 200 mg and 500 mg tablets were 102.0 ± 1.8%and 96.6 ± 2.6%, respectively, and for 2% clopidogrel 200 mg and 500 mg were 102.6 ± 3.9% and 101.2 ± 1.6%, respectively, well within the acceptable acceptance value (AV) range of 3 to 12. Both 1% and 2% formulations of clopidogrel tablets exhibited rapid drug release, meeting the USP pharmacopeial target of 85% release in 15 minutes. All tablet sizes formulated at 1% and 2% concentrations met specified disintegration specifications. The stability assessment over three months revealed consistent pH values and assay results within target specifications for both clopidogrel formulations (93.5% for 1% formulation and 93.6% for 2% formulation). At three months, X-ray Diffraction (XRD) and Fourier Transform Infrared Spectroscopy (FTIR) results demonstrated stability in clopidogrel tablets. In conclusion, a comprehensive evaluation of our developed clopidogrel tablets demonstrate their suitability for clinical use in an extemporaneous setting using the presented semi-solid extrusion-based automation technology.
{"title":"Automated Extrusion-Based Dispensing: Personalized Dosing and Quality Control of Clopidogrel Tablets for Pediatric Care.","authors":"Farnaz Shokraneh, Anne M Filppula, Aleksi Tornio, Jaan Aruväli, Urve Paaver, Niklas Sandler Topelius","doi":"10.1016/j.ejps.2024.106967","DOIUrl":"https://doi.org/10.1016/j.ejps.2024.106967","url":null,"abstract":"<p><p>The exploration of three-dimensional (3D) printing inspired technologies in pharmaceutical compounding reveals a promising frontier in personalized medicine manufacture. This study focuses on the development of clopidogrel bisulphate tablets, with doses ranging from 2 mg to 20 mg per tablet, suitable for pediatric use. The study explored a semi-solid extrusion-based deposition technology already being used in compounding pharmacies across several European locations. The investigation explored various properties of two formulations of 1% and 2% clopidogrel gel tablets, with a specific focus on mass variation, drug content uniformity, in vitro drug release profiles, disintegration time, and stability. The mean weights of the smallest printed 200 mg tablets with 1% and 2% clopidogrel concentrations were 199.1 ± 4.6 mg and 201.0 ± 3.2 mg, respectively. For the largest printed 500 mg tablets with 1% and 2% concentrations, the mean weights were 499.3 ± 7.7 mg and 501.7 ± 6.5 mg, respectively. The mean clopidogrel content uniformity for 1% clopidogrel 200 mg and 500 mg tablets were 102.0 ± 1.8%and 96.6 ± 2.6%, respectively, and for 2% clopidogrel 200 mg and 500 mg were 102.6 ± 3.9% and 101.2 ± 1.6%, respectively, well within the acceptable acceptance value (AV) range of 3 to 12. Both 1% and 2% formulations of clopidogrel tablets exhibited rapid drug release, meeting the USP pharmacopeial target of 85% release in 15 minutes. All tablet sizes formulated at 1% and 2% concentrations met specified disintegration specifications. The stability assessment over three months revealed consistent pH values and assay results within target specifications for both clopidogrel formulations (93.5% for 1% formulation and 93.6% for 2% formulation). At three months, X-ray Diffraction (XRD) and Fourier Transform Infrared Spectroscopy (FTIR) results demonstrated stability in clopidogrel tablets. In conclusion, a comprehensive evaluation of our developed clopidogrel tablets demonstrate their suitability for clinical use in an extemporaneous setting using the presented semi-solid extrusion-based automation technology.</p>","PeriodicalId":12018,"journal":{"name":"European Journal of Pharmaceutical Sciences","volume":" ","pages":"106967"},"PeriodicalIF":4.3,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142686337","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-17DOI: 10.1016/j.ejps.2024.106964
Letao Li, Annelienke M van Hulst, Emma J Verwaaijen, M Marry van den Heuvel-Eibrink, E L T Erica van den Akker, W Wim J R Rietdijk, B C P Birgit Koch, S D T Sebastiaan Sassen
Dexamethasone is crucial in pediatric acute lymphoblastic leukemia (ALL) treatment, however, studies regarding the pharmacokinetics of dexamethasone and its metabolites are scarce. Our study conducted a comprehensive pharmacokinetic-pharmacodynamic analysis of dexamethasone and metabolite, examining their association with dexamethasone-induced toxicity. Peak and trough concentrations were collected during the maintenance phase from pediatric ALL patients who received oral dexamethasone (6mg/m2/day). NONMEM was used to study the population pharmacokinetics including covariates. Pharmacokinetic (PK) and pharmacodynamic (PD) correlations between drug and its active metabolite exposure and adverse effects were examined. 382 samples (dexamethasone: n=191; 6β-hydroxydexamethasone: n=191) from 104 children (age range 3.0 -18.8 years) were collected. A one-compartment model described the data best. The estimated apparent dexamethasone total clearance was 26 L/h/70kg with 18% inter-individual variability, and an apparent volume of distribution of 123 L/70kg, yielding a half-life of 3.3 hours. Covariate analysis demonstrated that when asparaginase was co-administered, there was a 50% reduction in both the clearance of dexamethasone and the extent to which dexamethasone was metabolized to 6β-hydroxydexamethasone. A statistically significant but weak positive correlation was observed between dexamethasone drug exposure and fasting hunger scores. Dexamethasone exposure significantly increased with asparaginase co-administration by inhibition of the CYP3A4 pathway. Our study found a statistically significant but weak positive correlation between dexamethasone exposure and increased hunger. These results support the need for more studies on how to personalize dexamethasone dosing in pediatric ALL treatment and adjust doses to limit side effects, especially in case of co-medication.
{"title":"Towards optimization of dexamethasone therapy in the maintenance phase of pediatric acute lymphoblastic leukemia: a population pharmacokinetic and pharmacodynamic study of dexamethasone and metabolite.","authors":"Letao Li, Annelienke M van Hulst, Emma J Verwaaijen, M Marry van den Heuvel-Eibrink, E L T Erica van den Akker, W Wim J R Rietdijk, B C P Birgit Koch, S D T Sebastiaan Sassen","doi":"10.1016/j.ejps.2024.106964","DOIUrl":"https://doi.org/10.1016/j.ejps.2024.106964","url":null,"abstract":"<p><p>Dexamethasone is crucial in pediatric acute lymphoblastic leukemia (ALL) treatment, however, studies regarding the pharmacokinetics of dexamethasone and its metabolites are scarce. Our study conducted a comprehensive pharmacokinetic-pharmacodynamic analysis of dexamethasone and metabolite, examining their association with dexamethasone-induced toxicity. Peak and trough concentrations were collected during the maintenance phase from pediatric ALL patients who received oral dexamethasone (6mg/m2/day). NONMEM was used to study the population pharmacokinetics including covariates. Pharmacokinetic (PK) and pharmacodynamic (PD) correlations between drug and its active metabolite exposure and adverse effects were examined. 382 samples (dexamethasone: n=191; 6β-hydroxydexamethasone: n=191) from 104 children (age range 3.0 -18.8 years) were collected. A one-compartment model described the data best. The estimated apparent dexamethasone total clearance was 26 L/h/70kg with 18% inter-individual variability, and an apparent volume of distribution of 123 L/70kg, yielding a half-life of 3.3 hours. Covariate analysis demonstrated that when asparaginase was co-administered, there was a 50% reduction in both the clearance of dexamethasone and the extent to which dexamethasone was metabolized to 6β-hydroxydexamethasone. A statistically significant but weak positive correlation was observed between dexamethasone drug exposure and fasting hunger scores. Dexamethasone exposure significantly increased with asparaginase co-administration by inhibition of the CYP3A4 pathway. Our study found a statistically significant but weak positive correlation between dexamethasone exposure and increased hunger. These results support the need for more studies on how to personalize dexamethasone dosing in pediatric ALL treatment and adjust doses to limit side effects, especially in case of co-medication.</p>","PeriodicalId":12018,"journal":{"name":"European Journal of Pharmaceutical Sciences","volume":" ","pages":"106964"},"PeriodicalIF":4.3,"publicationDate":"2024-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142675517","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-16DOI: 10.1016/j.ejps.2024.106962
Mai Bay Stie , Dirk Fennema Galparsoro , Xin Zhou , Vito Foderà
Protein self-assemblies in the form of ordered supramolecular structures such as particulates hold great potential as new biomaterials. However, research in this field is rarely conducted under physiologically relevant conditions but such studies are crucially needed to unravel the potential use of particulates and other amyloid structures in health sciences. In this study, particulates of α-lactalbumin (ALA) were prepared at different stages of maturation by thermal incubation. Disassembly of particulates in isotonic buffer, neutral pH and at 37 °C was investigated by simultaneously measuring Thioflavin T fluorescence intensity and light scattering. Freshly formed particulates quickly disassembled and displayed complete release of soluble ALA within 1 h. Mature particulates displayed slower disassembly kinetics with incomplete release of ALA within 1 h. The biocompatibility of particulates at different maturation stages to epithelial lung and fibroblast cells was assessed in vitro. Good cell compatibility was observed in the presence of the particulates and their released species. Our findings display protein particulates as biodegradable and highly tunable particles, promoting them as good candidates for drug delivery purposes.
有序超分子结构形式的蛋白质自组装(如微粒)具有作为新型生物材料的巨大潜力。然而,该领域的研究很少在生理相关条件下进行,但要揭示微粒和其他淀粉样结构在健康科学中的潜在用途,此类研究是非常必要的。本研究通过热孵育法制备了处于不同成熟阶段的α-乳白蛋白(ALA)微粒。通过同时测量硫黄素 T 荧光强度和光散射,研究了微粒在等渗缓冲液、中性 pH 和 37 °C条件下的分解情况。在体外评估了不同成熟阶段的微粒对肺上皮细胞和成纤维细胞的生物相容性。在微粒及其释放物存在的情况下,观察到了良好的细胞相容性。我们的研究结果表明,蛋白质微粒具有生物可降解性和高度可调性,是药物输送的理想候选材料。
{"title":"Disassembly and in vitro cell compatibility of α-lactalbumin particulates under physiologically relevant conditions","authors":"Mai Bay Stie , Dirk Fennema Galparsoro , Xin Zhou , Vito Foderà","doi":"10.1016/j.ejps.2024.106962","DOIUrl":"10.1016/j.ejps.2024.106962","url":null,"abstract":"<div><div>Protein self-assemblies in the form of ordered supramolecular structures such as particulates hold great potential as new biomaterials. However, research in this field is rarely conducted under physiologically relevant conditions but such studies are crucially needed to unravel the potential use of particulates and other amyloid structures in health sciences. In this study, particulates of α-lactalbumin (ALA) were prepared at different stages of maturation by thermal incubation. Disassembly of particulates in isotonic buffer, neutral pH and at 37 °C was investigated by simultaneously measuring Thioflavin T fluorescence intensity and light scattering. Freshly formed particulates quickly disassembled and displayed complete release of soluble ALA within 1 h. Mature particulates displayed slower disassembly kinetics with incomplete release of ALA within 1 h. The biocompatibility of particulates at different maturation stages to epithelial lung and fibroblast cells was assessed <em>in vitro</em>. Good cell compatibility was observed in the presence of the particulates and their released species. Our findings display protein particulates as biodegradable and highly tunable particles, promoting them as good candidates for drug delivery purposes.</div></div>","PeriodicalId":12018,"journal":{"name":"European Journal of Pharmaceutical Sciences","volume":"204 ","pages":"Article 106962"},"PeriodicalIF":4.3,"publicationDate":"2024-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142647192","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 : 2024-11-15DOI: 10.1016/j.ejps.2024.106963
Zarah Schaal, Pieter-Jan van Bockstal, Joris Lammens, Julian H Lenger, Adrian P Funke, Stefan C Schneid, Hristo L Svilenov, Thomas De Beer
Continuous spin-freeze-drying is an innovative pharmaceutical manufacturing approach offering real-time monitoring and control at the individual vial level, unlike conventional batch lyophilization. A central feature of this technology is spin-freezing, which involves rapidly spinning liquid-filled vials under a precisely controlled cold gas flow, resulting in a thin, uniform frozen product layer. Using a model peptide formulation, we investigated the impact of different cooling and crystallization rates on quality attributes (QA) and primary drying duration. Key QAs included monomer content, peptide assay, moisture content, and pore structure. The monomer content, peptide content, and primary drying duration remained consistent across all spin-freezing conditions. However, scanning electron microscopy (SEM) and Karl Fischer titration revealed that freezing parameters significantly influenced pore structure and residual moisture content. Samples with smaller pores displayed lower residual moisture, as larger surface areas facilitate moisture desorption. Variations in freezing parameters also significantly impacted desorption kinetics during secondary drying. Slower crystallization rates led to more cracks and less shrinkage in the cake structure, while faster rates resulted in more uniform, stable cakes. Although specific to the product under study, these findings highlight the crucial role of spin-freezing in enhancing freeze-drying efficiency and product quality of biopharmaceuticals.
{"title":"Optimization of Continuous Spin-Freeze-Drying: The Role of Spin-Freezing on Quality Attributes and Drying Efficiency of a Model Peptide Formulation.","authors":"Zarah Schaal, Pieter-Jan van Bockstal, Joris Lammens, Julian H Lenger, Adrian P Funke, Stefan C Schneid, Hristo L Svilenov, Thomas De Beer","doi":"10.1016/j.ejps.2024.106963","DOIUrl":"https://doi.org/10.1016/j.ejps.2024.106963","url":null,"abstract":"<p><p>Continuous spin-freeze-drying is an innovative pharmaceutical manufacturing approach offering real-time monitoring and control at the individual vial level, unlike conventional batch lyophilization. A central feature of this technology is spin-freezing, which involves rapidly spinning liquid-filled vials under a precisely controlled cold gas flow, resulting in a thin, uniform frozen product layer. Using a model peptide formulation, we investigated the impact of different cooling and crystallization rates on quality attributes (QA) and primary drying duration. Key QAs included monomer content, peptide assay, moisture content, and pore structure. The monomer content, peptide content, and primary drying duration remained consistent across all spin-freezing conditions. However, scanning electron microscopy (SEM) and Karl Fischer titration revealed that freezing parameters significantly influenced pore structure and residual moisture content. Samples with smaller pores displayed lower residual moisture, as larger surface areas facilitate moisture desorption. Variations in freezing parameters also significantly impacted desorption kinetics during secondary drying. Slower crystallization rates led to more cracks and less shrinkage in the cake structure, while faster rates resulted in more uniform, stable cakes. Although specific to the product under study, these findings highlight the crucial role of spin-freezing in enhancing freeze-drying efficiency and product quality of biopharmaceuticals.</p>","PeriodicalId":12018,"journal":{"name":"European Journal of Pharmaceutical Sciences","volume":" ","pages":"106963"},"PeriodicalIF":4.3,"publicationDate":"2024-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142647191","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-15DOI: 10.1016/j.ejps.2024.106957
Justus Johann Lange, Lukas Enzner, Martin Kuentz, Patrick J O'Dwyer, Wiebke Saal, Brendan T Griffin, Nicole Wyttenbach
Preclinical testing of new drug candidates frequently necessitates high-dose solution formulations to support robust testing in rodent models. This study aimed to expand the range of high solubilisation capacity formulations by exploring the solubilisation effects of the polymeric surfactant Soluplus® in combination with ionic surfactants. The interactions between Soluplus® and three ionic surfactants, sodium dodecyl sulfate, dioctyl sodium succinate, and sodium oleate, with a primary focus on solubility enhancement were investigated over a range of ionic surfactant concentrations. The solubilisation profiles for seven model drugs were obtained, and the vehicles were characterized by their visual characteristics, dynamic light scattering, and viscosity measurements. The solubilisation profiles were non-linear, indicating the formation of different colloidal species with individual solubilisation strengths depending on surfactant type and concentration, demonstrating substantial solubility enhancement. For certain drugs more than additive solubilisation, facilitated by synergistic interactions between Soluplus® and the ionic surfactants, was obtained. Overall, the solubility increase provided by the excipient combinations resulted in non-linear and drug specific solubilisation profiles. The non-linearities observed were reflected in visual observations of the vehicles appearance, DLS and viscosity measurements, which collectively indicated a change in polymer aggregation with increasing concentration of anionic surfactant. This investigation highlights that already low quantities of ionic surfactants introduced to Soluplus® may substantially enhance solubility, which offers a promising approach for further exploration in preclinical drug development where more conventional solubilising formulation strategies may fall short.
{"title":"Exploration of solubilisation effects facilitated by the combination of Soluplus® with ionic surfactants.","authors":"Justus Johann Lange, Lukas Enzner, Martin Kuentz, Patrick J O'Dwyer, Wiebke Saal, Brendan T Griffin, Nicole Wyttenbach","doi":"10.1016/j.ejps.2024.106957","DOIUrl":"https://doi.org/10.1016/j.ejps.2024.106957","url":null,"abstract":"<p><p>Preclinical testing of new drug candidates frequently necessitates high-dose solution formulations to support robust testing in rodent models. This study aimed to expand the range of high solubilisation capacity formulations by exploring the solubilisation effects of the polymeric surfactant Soluplus® in combination with ionic surfactants. The interactions between Soluplus® and three ionic surfactants, sodium dodecyl sulfate, dioctyl sodium succinate, and sodium oleate, with a primary focus on solubility enhancement were investigated over a range of ionic surfactant concentrations. The solubilisation profiles for seven model drugs were obtained, and the vehicles were characterized by their visual characteristics, dynamic light scattering, and viscosity measurements. The solubilisation profiles were non-linear, indicating the formation of different colloidal species with individual solubilisation strengths depending on surfactant type and concentration, demonstrating substantial solubility enhancement. For certain drugs more than additive solubilisation, facilitated by synergistic interactions between Soluplus® and the ionic surfactants, was obtained. Overall, the solubility increase provided by the excipient combinations resulted in non-linear and drug specific solubilisation profiles. The non-linearities observed were reflected in visual observations of the vehicles appearance, DLS and viscosity measurements, which collectively indicated a change in polymer aggregation with increasing concentration of anionic surfactant. This investigation highlights that already low quantities of ionic surfactants introduced to Soluplus® may substantially enhance solubility, which offers a promising approach for further exploration in preclinical drug development where more conventional solubilising formulation strategies may fall short.</p>","PeriodicalId":12018,"journal":{"name":"European Journal of Pharmaceutical Sciences","volume":" ","pages":"106957"},"PeriodicalIF":4.3,"publicationDate":"2024-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142647190","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
4-Bromo-N-(thiazol-2-yl)benzenesulfonamide (1) is enriched with bioactive components and is highlighted for its pharmacological properties. However, its pharmacokinetic characteristics are yet to be reported. The interaction of compound 1 with carrier proteins in the bloodstream is an important factor that affects its potential therapeutic efficacy. This study aimed to elucidate the pharmacokinetic mechanisms of compound 1 in relation to human serum albumin (HSA) using multi-spectroscopic and computational techniques. Its predicted drug-like properties revealed no mutagenicity, although potential hepatotoxicity and interactions with certain cytochrome P450 enzymes were observed. Spectroscopic analyses extensively provided the interaction between HSA and 1 through a static fluorescence quenching mechanism with spontaneous hydrophobic interactions and hydrogen bonding. The binding constant of the HSA‒1 complex was relatively moderate to strong at a level of 106 M-1. Various spectroscopic techniques including ultraviolet-visible, Fourier transform infrared, and circular dichroism spectroscopies indicated that its binding induced alteration in the α-helix content of HSA. Competitive binding and molecular docking studies designated the preferential binding of 1 to sub-structural domain IIA binding site I of HSA. Molecular dynamic simulations further illustrated the formation of a stable complex between 1 and HSA, accompanied by conformational changes in the protein. Importantly, esterase capacity of the HSA‒1 complex increased compared to the free HSA. Therefore, elucidation of the HSA‒1 binding mechanism provides valuable insights into the pharmacokinetics, suggesting potential benefits for the further development of 1 as a therapeutic agent.
{"title":"Biophysical insight into the interaction mechanism of 4-bromo-N-(thiazol-2-yl)benzenesulfonamide and human serum albumin using multi-spectroscopic and computational studies.","authors":"Francis Ayimbila, Kamonrat Phopin, Waralee Ruankham, Ratchanok Pingaew, Supaluk Prachayasittikul, Virapong Prachayasittikul, Tanawut Tantimongcolwat","doi":"10.1016/j.ejps.2024.106961","DOIUrl":"10.1016/j.ejps.2024.106961","url":null,"abstract":"<p><p>4-Bromo-N-(thiazol-2-yl)benzenesulfonamide (1) is enriched with bioactive components and is highlighted for its pharmacological properties. However, its pharmacokinetic characteristics are yet to be reported. The interaction of compound 1 with carrier proteins in the bloodstream is an important factor that affects its potential therapeutic efficacy. This study aimed to elucidate the pharmacokinetic mechanisms of compound 1 in relation to human serum albumin (HSA) using multi-spectroscopic and computational techniques. Its predicted drug-like properties revealed no mutagenicity, although potential hepatotoxicity and interactions with certain cytochrome P450 enzymes were observed. Spectroscopic analyses extensively provided the interaction between HSA and 1 through a static fluorescence quenching mechanism with spontaneous hydrophobic interactions and hydrogen bonding. The binding constant of the HSA‒1 complex was relatively moderate to strong at a level of 10<sup>6</sup> M<sup>-1</sup>. Various spectroscopic techniques including ultraviolet-visible, Fourier transform infrared, and circular dichroism spectroscopies indicated that its binding induced alteration in the α-helix content of HSA. Competitive binding and molecular docking studies designated the preferential binding of 1 to sub-structural domain IIA binding site I of HSA. Molecular dynamic simulations further illustrated the formation of a stable complex between 1 and HSA, accompanied by conformational changes in the protein. Importantly, esterase capacity of the HSA‒1 complex increased compared to the free HSA. Therefore, elucidation of the HSA‒1 binding mechanism provides valuable insights into the pharmacokinetics, suggesting potential benefits for the further development of 1 as a therapeutic agent.</p>","PeriodicalId":12018,"journal":{"name":"European Journal of Pharmaceutical Sciences","volume":" ","pages":"106961"},"PeriodicalIF":4.3,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142617210","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-08DOI: 10.1016/j.ejps.2024.106960
Larissa Keßler, Rashmi Mishra, Sami Hietala, Manon Lammens, Leena Peltonen, Thomas Rades, Bert van Veen, Anne Juppo, Timo Laaksonen, Clare Strachan, Robert Luxenhofer
Improving the solubility of poorly water-soluble drugs is essential for enhancing bioavailability, formulation flexibility and reducing patient-to-patient variability. The preparation of amorphous solid dispersions (ASDs) is an attractive strategy to formulate such drugs, leading to higher apparent water solubility and therefore higher bioavailability. For such ASDs, water-soluble polymer excipients, such as poly(vinyl pyrrolidone) (PVP) or poly(vinyl pyrrolidone-co-vinyl acetate) (P(VP-co-VA)), are employed to solubilize and stabilize the drug against crystallization. We posit that polymers bearing tertiary amides are particularly well suited to stabilizing drugs containing H-bond donors, as they offer strong H-bonding potential between the polymer and drug. The aim of this study was to compare new and established polymers with tertiary amides as excipients for ASDs. Experimental amphiphilic ABA triblock copolymers comprising poly(2-methyl-2-oxazoline) (pMeOx), poly(2-butyl-2-oxazoline) (pBuOx) and poly(2-butyl-2-oxazine) (pBuOzi) blocks, were compared with the established excipients, PVP and P(VP-co-VA). ASDs with indomethacin as the model drug were prepared at high drug loadings via hot melt extrusion. The extrudates were studied with DSC and PXRD, revealing the ASDs to be fully amorphous up to 75wt% indomethacin, independent of the polymer used. 13C CPMAS NMR provided insights into intermolecular associations as a function of drug loading, and suggested the presence of drug dimers at 75wt% drug loading in pMeOx-pBuOzi-pMeOx and pMeOx-pBuOx-pMeOx, which could affect physical stability. Independent of the polymers, the solid-state form of the drug in the ASD was found to affect the dissolution profile of the samples, insofar as the samples containing crystalline indomethacin showed slower dissolution than the fully amorphous ones. This study shows that the polymers comprising poly(2-oxazoline) and poly(2-oxazine) are effective polymers for ASD preparation, similar to PVP and P(VP-co-VA) which merits further investigations into these novel polymers for formulating ASDs.
{"title":"Amorphous solid dispersions of amphiphilic polymer excipients and indomethacin prepared by hot melt extrusion.","authors":"Larissa Keßler, Rashmi Mishra, Sami Hietala, Manon Lammens, Leena Peltonen, Thomas Rades, Bert van Veen, Anne Juppo, Timo Laaksonen, Clare Strachan, Robert Luxenhofer","doi":"10.1016/j.ejps.2024.106960","DOIUrl":"https://doi.org/10.1016/j.ejps.2024.106960","url":null,"abstract":"<p><p>Improving the solubility of poorly water-soluble drugs is essential for enhancing bioavailability, formulation flexibility and reducing patient-to-patient variability. The preparation of amorphous solid dispersions (ASDs) is an attractive strategy to formulate such drugs, leading to higher apparent water solubility and therefore higher bioavailability. For such ASDs, water-soluble polymer excipients, such as poly(vinyl pyrrolidone) (PVP) or poly(vinyl pyrrolidone-co-vinyl acetate) (P(VP-co-VA)), are employed to solubilize and stabilize the drug against crystallization. We posit that polymers bearing tertiary amides are particularly well suited to stabilizing drugs containing H-bond donors, as they offer strong H-bonding potential between the polymer and drug. The aim of this study was to compare new and established polymers with tertiary amides as excipients for ASDs. Experimental amphiphilic ABA triblock copolymers comprising poly(2-methyl-2-oxazoline) (pMeOx), poly(2-butyl-2-oxazoline) (pBuOx) and poly(2-butyl-2-oxazine) (pBuOzi) blocks, were compared with the established excipients, PVP and P(VP-co-VA). ASDs with indomethacin as the model drug were prepared at high drug loadings via hot melt extrusion. The extrudates were studied with DSC and PXRD, revealing the ASDs to be fully amorphous up to 75wt% indomethacin, independent of the polymer used. <sup>13</sup>C CPMAS NMR provided insights into intermolecular associations as a function of drug loading, and suggested the presence of drug dimers at 75wt% drug loading in pMeOx-pBuOzi-pMeOx and pMeOx-pBuOx-pMeOx, which could affect physical stability. Independent of the polymers, the solid-state form of the drug in the ASD was found to affect the dissolution profile of the samples, insofar as the samples containing crystalline indomethacin showed slower dissolution than the fully amorphous ones. This study shows that the polymers comprising poly(2-oxazoline) and poly(2-oxazine) are effective polymers for ASD preparation, similar to PVP and P(VP-co-VA) which merits further investigations into these novel polymers for formulating ASDs.</p>","PeriodicalId":12018,"journal":{"name":"European Journal of Pharmaceutical Sciences","volume":" ","pages":"106960"},"PeriodicalIF":4.3,"publicationDate":"2024-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142617207","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-08DOI: 10.1016/j.ejps.2024.106959
Elena Markova , Camilla Wolowczyk , Aly Mohamed , Alexandros Marios Sofias , Montserrat Martin-Armas , Rune Sundset , Jens Berndtsson , Sjoerd Hak , Nataša Škalko-Basnet
Cancer immunotherapy has evolved significantly over the last decade, with therapeutics targeting the adaptive immune system showing exciting effects in clinics. Yet, the modulation of the innate immune system, particularly the tumor-associated innate immune cells which are an integral part of immune responses in cancer, remains less understood. The arginase 1 (Arg1) pathway is a pivotal metabolic pathway that tumor-associated innate immune cells exploit to create an immunosuppressive tumor microenvironment, leading to the evasion of immune surveillance. The inhibition of Arg1 presents a therapeutic opportunity to reverse this immunosuppression, and Nω‑hydroxy-l-norarginine (nor-NOHA) has emerged as a potent arginase inhibitor with promising in vivo efficacy. However, the rapid systemic clearance of nor-NOHA poses a significant challenge for its therapeutic application. This study pioneers the encapsulation of nor-NOHA in liposomes, aiming to enhance its bioavailability and prolong its inhibitory activity against Arg1. Historically, the extensive interaction between innate immune cells and nanoparticles has been one of the biggest drawbacks in nanomedicine. Here we seek to utilize this effect and deliver liposomal nor-NOHA to the arginase 1 expressing innate immune cells. We systematically investigated the effect of lipid composition, acyl chain length, manufacturing and loading methodology on the encapsulation efficiency (EE%) and release profile of nor-NOHA. Our results indicate that while the manufacturing method and lipid acyl chain length do not significantly impact EE%, they crucially influence the release kinetics of nor-NOHA, with longer acyl chains demonstrating a more sustained release of nor-NOHA from liposomes enabling continuous inhibition of Arg1. Our findings suggest that liposomal nor-NOHA retains its functional inhibitory activity and could offer improved pharmacokinetic properties, making it a compelling base for iterations for further innovative cancer immunotherapeutic strategies in preclinical and clinical evaluations.
{"title":"Liposomal Nω-hydroxy-l-norarginine, a proof-of-concept: Arginase inhibitors can be incorporated in liposomes while retaining their therapeutic activity ex vivo","authors":"Elena Markova , Camilla Wolowczyk , Aly Mohamed , Alexandros Marios Sofias , Montserrat Martin-Armas , Rune Sundset , Jens Berndtsson , Sjoerd Hak , Nataša Škalko-Basnet","doi":"10.1016/j.ejps.2024.106959","DOIUrl":"10.1016/j.ejps.2024.106959","url":null,"abstract":"<div><div>Cancer immunotherapy has evolved significantly over the last decade, with therapeutics targeting the adaptive immune system showing exciting effects in clinics. Yet, the modulation of the innate immune system, particularly the tumor-associated innate immune cells which are an integral part of immune responses in cancer, remains less understood. The arginase 1 (Arg1) pathway is a pivotal metabolic pathway that tumor-associated innate immune cells exploit to create an immunosuppressive tumor microenvironment, leading to the evasion of immune surveillance. The inhibition of Arg1 presents a therapeutic opportunity to reverse this immunosuppression, and Nω‑hydroxy-<span>l</span>-norarginine (nor-NOHA) has emerged as a potent arginase inhibitor with promising in vivo efficacy. However, the rapid systemic clearance of nor-NOHA poses a significant challenge for its therapeutic application. This study pioneers the encapsulation of nor-NOHA in liposomes, aiming to enhance its bioavailability and prolong its inhibitory activity against Arg1. Historically, the extensive interaction between innate immune cells and nanoparticles has been one of the biggest drawbacks in nanomedicine. Here we seek to utilize this effect and deliver liposomal nor-NOHA to the arginase 1 expressing innate immune cells. We systematically investigated the effect of lipid composition, acyl chain length, manufacturing and loading methodology on the encapsulation efficiency (EE%) and release profile of nor-NOHA. Our results indicate that while the manufacturing method and lipid acyl chain length do not significantly impact EE%, they crucially influence the release kinetics of nor-NOHA, with longer acyl chains demonstrating a more sustained release of nor-NOHA from liposomes enabling continuous inhibition of Arg1. Our findings suggest that liposomal nor-NOHA retains its functional inhibitory activity and could offer improved pharmacokinetic properties, making it a compelling base for iterations for further innovative cancer immunotherapeutic strategies in preclinical and clinical evaluations.</div></div>","PeriodicalId":12018,"journal":{"name":"European Journal of Pharmaceutical Sciences","volume":"204 ","pages":"Article 106959"},"PeriodicalIF":4.3,"publicationDate":"2024-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142617222","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 : 2024-11-07DOI: 10.1016/j.ejps.2024.106958
Shiyao Ma , Shanhui Yi , Hui Zou , Shasha Fan , Yin Xiao
Protein Arginine Methyltransferase 1 (PRMT1), a primary protein arginine methyltransferase, plays a pivotal role in cellular regulation, influencing processes such as gene expression, signal transduction, and cell differentiation. Dysregulation of PRMT1 has been linked to the development of various cancers, establishing it as a key target for therapeutic intervention. This review synthesizes the biochemical characteristics, structural domains, and functional mechanisms of PRMT1, focusing on its involvement in tumorigenesis. Additionally, the development and efficacy of emerging PRMT1 inhibitors as potential cancer therapies are examined. By employing molecular modeling and insights from existing literature, this review posits that targeting PRMT1’s methyltransferase activity could disrupt cancer progression, providing valuable insights for future drug development.
{"title":"The role of PRMT1 in cellular regulation and disease: Insights into biochemical functions and emerging inhibitors for cancer therapy","authors":"Shiyao Ma , Shanhui Yi , Hui Zou , Shasha Fan , Yin Xiao","doi":"10.1016/j.ejps.2024.106958","DOIUrl":"10.1016/j.ejps.2024.106958","url":null,"abstract":"<div><div>Protein Arginine Methyltransferase 1 (PRMT1), a primary protein arginine methyltransferase, plays a pivotal role in cellular regulation, influencing processes such as gene expression, signal transduction, and cell differentiation. Dysregulation of PRMT1 has been linked to the development of various cancers, establishing it as a key target for therapeutic intervention. This review synthesizes the biochemical characteristics, structural domains, and functional mechanisms of PRMT1, focusing on its involvement in tumorigenesis. Additionally, the development and efficacy of emerging PRMT1 inhibitors as potential cancer therapies are examined. By employing molecular modeling and insights from existing literature, this review posits that targeting PRMT1’s methyltransferase activity could disrupt cancer progression, providing valuable insights for future drug development.</div></div>","PeriodicalId":12018,"journal":{"name":"European Journal of Pharmaceutical Sciences","volume":"204 ","pages":"Article 106958"},"PeriodicalIF":4.3,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142617227","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}