Pub Date : 2025-12-01Epub Date: 2025-06-16DOI: 10.1080/09205063.2025.2519868
Jianfei Cao, Chao Peng, Yan Lei, Haoming Wu, Shaojuan Xu, Qiyu Liu, Yi Liu, Mengjue Li, Yue Lu
Poly(lacticacid-ε-caprolactone) (PLCL) scaffolds face significant challenges in bone regeneration due to excessively slow degradation kinetics and inherent hydrophobicity. To overcome these limitations, we developed a novel ternary 3D-printed scaffold composed of PLCL, poly(p-dioxanone) (PPDO), and hydroxyapatite (HA) via fused deposition modeling (FDM) for the first time. The incorporation of PPDO would accelerate and enable tunable degradation of PLCL to match the bone healing timeline, while HA was aimed to enhance osteoinductivity and regulated the pH level to reduce adverse immune reactions of the acidic degradation products. The results demonstrated that degradation rate of the scaffolds was found to be modulated by PPDO and HA effectively. Moreover, the 3D printing extrusion enabled the porous scaffolds with customizability, diverse shapes, adjustable porosity and uniform pore sizes. In addition, proliferation and adhesion of bone marrow mesenchymal stem cells (BMSCs) as well as the expression of various osteogenic genes (ALP, Col-Ι, OCN, BMP-2, OPN) were also upregulated on the PLCL/PPDO/HA scaffolds. Therefore, these low-cost 3D-printed scaffolds may serve as an optimal bone graft for applications in bone tissue engineering.
{"title":"PPDO-induced tunable degradation and HA-enhanced osteogenesis in PLCL scaffolds for bone regeneration.","authors":"Jianfei Cao, Chao Peng, Yan Lei, Haoming Wu, Shaojuan Xu, Qiyu Liu, Yi Liu, Mengjue Li, Yue Lu","doi":"10.1080/09205063.2025.2519868","DOIUrl":"10.1080/09205063.2025.2519868","url":null,"abstract":"<p><p>Poly(lacticacid-ε-caprolactone) (PLCL) scaffolds face significant challenges in bone regeneration due to excessively slow degradation kinetics and inherent hydrophobicity. To overcome these limitations, we developed a novel ternary 3D-printed scaffold composed of PLCL, poly(p-dioxanone) (PPDO), and hydroxyapatite (HA) <i>via</i> fused deposition modeling (FDM) for the first time. The incorporation of PPDO would accelerate and enable tunable degradation of PLCL to match the bone healing timeline, while HA was aimed to enhance osteoinductivity and regulated the pH level to reduce adverse immune reactions of the acidic degradation products. The results demonstrated that degradation rate of the scaffolds was found to be modulated by PPDO and HA effectively. Moreover, the 3D printing extrusion enabled the porous scaffolds with customizability, diverse shapes, adjustable porosity and uniform pore sizes. In addition, proliferation and adhesion of bone marrow mesenchymal stem cells (BMSCs) as well as the expression of various osteogenic genes (ALP, Col-Ι, OCN, BMP-2, OPN) were also upregulated on the PLCL/PPDO/HA scaffolds. Therefore, these low-cost 3D-printed scaffolds may serve as an optimal bone graft for applications in bone tissue engineering.</p>","PeriodicalId":15195,"journal":{"name":"Journal of Biomaterials Science, Polymer Edition","volume":" ","pages":"3108-3124"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144309959","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-06-25DOI: 10.1080/09205063.2025.2522744
Huimin Wang, Kanran Ling, Qingting Hu, Hanchen Ding, Ran Li, Miao Li, Si Xu, Yangyang Cao
As surfactants, amphiphilic molecules form micelles in aqueous solution to load hydrophobic medicines to increase their solubility and absorbability. TPGS, i.e. VE-PEG conjugate, is a commonly used effective surfactant suffering immune effects in human bodies with reduced biocompatibility and stealth property. Among the potential alternatives of PEG, polysarcosine (pSar) is the most promising one due to its outstanding property and effectiveness. Herein, we propose two strategies to polymerize Sar-NPC, direct initialization and post-polymerization chain end modification to conjugate hydrophobic building blocks onto pSar. Direct initial-ization applies amino-group-containing lipids 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) as initiators to produce DSPE-pSar and DOPE-pSar, respectively. Post-polymerization chain end modification changes chain end amino group of pSar to carboxyl group for esterification with the hydroxyl groups on vitamin E (VE) and 1,2-dimyristoyl-sn-glycerol (DMG) to produce VE-pSar and DMG-pSar. The degrees of polymerization of pSar blocks are designed to be 14, 25 and 28 precisely, and the CMC values of the amphiphilic products are between 0.28 and 5.63 µg/mL. VE-pSar samples have extremely strong ability to increase the solubility of paclitaxel (PTX), 30 times more than TPGS. It also exhibits high cytocompatibility and low hemolysis rate below 5%, much less than TPGS. The two preparations of pSar-containing surfactants are efficient and versatile, and the products have high probability to become a new generation of clinical hydrophobic medicine solubilizer.
{"title":"Polysarcosine-based surfactants: syntheses and properties.","authors":"Huimin Wang, Kanran Ling, Qingting Hu, Hanchen Ding, Ran Li, Miao Li, Si Xu, Yangyang Cao","doi":"10.1080/09205063.2025.2522744","DOIUrl":"10.1080/09205063.2025.2522744","url":null,"abstract":"<p><p>As surfactants, amphiphilic molecules form micelles in aqueous solution to load hydrophobic medicines to increase their solubility and absorbability. TPGS, i.e. VE-PEG conjugate, is a commonly used effective surfactant suffering immune effects in human bodies with reduced biocompatibility and stealth property. Among the potential alternatives of PEG, polysarcosine (pSar) is the most promising one due to its outstanding property and effectiveness. Herein, we propose two strategies to polymerize Sar-NPC, <i>direct initialization</i> and <i>post-polymerization chain end modification</i> to conjugate hydrophobic building blocks onto pSar. <i>Direct initial-ization</i> applies amino-group-containing lipids 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) as initiators to produce DSPE-pSar and DOPE-pSar, respectively. <i>Post-polymerization chain end modification</i> changes chain end amino group of pSar to carboxyl group for esterification with the hydroxyl groups on vitamin E (VE) and 1,2-dimyristoyl-sn-glycerol (DMG) to produce VE-pSar and DMG-pSar. The degrees of polymerization of pSar blocks are designed to be 14, 25 and 28 precisely, and the CMC values of the amphiphilic products are between 0.28 and 5.63 µg/mL. VE-pSar samples have extremely strong ability to increase the solubility of paclitaxel (PTX), 30 times more than TPGS. It also exhibits high cytocompatibility and low hemolysis rate below 5%, much less than TPGS. The two preparations of pSar-containing surfactants are efficient and versatile, and the products have high probability to become a new generation of clinical hydrophobic medicine solubilizer.</p>","PeriodicalId":15195,"journal":{"name":"Journal of Biomaterials Science, Polymer Edition","volume":" ","pages":"3173-3184"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144496731","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-06-10DOI: 10.1080/09205063.2025.2515944
Sruthi Laakshmi Mugundhan, Mothilal Mohan
Breast cancer therapy with Axitinib (AXT), a potent tyrosine kinase inhibitor, is limited by poor aqueous solubility, rapid clearance, and off-target toxicity. To address these challenges, we developed AXT-loaded nanomicelles (AXT-M) composed of Triton X-100, Brij-35, and Vitamin-E TPGS, with surface functionalization using hyaluronic acid (HA) for CD44 receptor-mediated targeting (HA-AXT-M). This formulation aims to enhance solubility, improve tumour specificity, and reduce systemic toxicity. The formulation was characterized for particle size, zeta potential, and encapsulation efficiency. Additionally, in vitro drug release, cytotoxicity using the MTT assay in MCF-7 cells, and stability studies under various storage and dilution conditions were performed. The optimized HA-AXT-M showed a particle size of 244.27 ± 1.04 nm, zeta potential of -27.3 mV, and encapsulation efficiency of 87.92%. Drug release studies demonstrated a biphasic release pattern with sustained release over 24 h. Cytotoxicity assays revealed enhanced anticancer activity of HA-AXT-M compared to uncoated micelles and free Axitinib. Stability studies confirmed the physical and chemical stability of the formulation. Moreover, HA functionalization substantially improved cellular uptake and selective targeting of CD44-overexpressing breast cancer cells, minimizing off-target effects. These findings highlight the promise of HA-AXT-M as a targeted nanocarrier platform that enhances the therapeutic potential of Axitinib. This novel delivery system offers improved efficacy, tumour selectivity, and translational relevance for breast cancer therapy.
{"title":"Hyaluronic acid-functionalized Axitinib nanomicelles for targeted drug delivery in breast cancer therapy.","authors":"Sruthi Laakshmi Mugundhan, Mothilal Mohan","doi":"10.1080/09205063.2025.2515944","DOIUrl":"10.1080/09205063.2025.2515944","url":null,"abstract":"<p><p>Breast cancer therapy with Axitinib (AXT), a potent tyrosine kinase inhibitor, is limited by poor aqueous solubility, rapid clearance, and off-target toxicity. To address these challenges, we developed AXT-loaded nanomicelles (AXT-M) composed of Triton X-100, Brij-35, and Vitamin-E TPGS, with surface functionalization using hyaluronic acid (HA) for CD44 receptor-mediated targeting (HA-AXT-M). This formulation aims to enhance solubility, improve tumour specificity, and reduce systemic toxicity. The formulation was characterized for particle size, zeta potential, and encapsulation efficiency. Additionally<i>, in vitro</i> drug release, cytotoxicity using the MTT assay in MCF-7 cells, and stability studies under various storage and dilution conditions were performed. The optimized HA-AXT-M showed a particle size of 244.27 ± 1.04 nm, zeta potential of -27.3 mV, and encapsulation efficiency of 87.92%. Drug release studies demonstrated a biphasic release pattern with sustained release over 24 h. Cytotoxicity assays revealed enhanced anticancer activity of HA-AXT-M compared to uncoated micelles and free Axitinib. Stability studies confirmed the physical and chemical stability of the formulation. Moreover, HA functionalization substantially improved cellular uptake and selective targeting of CD44-overexpressing breast cancer cells, minimizing off-target effects. These findings highlight the promise of HA-AXT-M as a targeted nanocarrier platform that enhances the therapeutic potential of Axitinib. This novel delivery system offers improved efficacy, tumour selectivity, and translational relevance for breast cancer therapy.</p>","PeriodicalId":15195,"journal":{"name":"Journal of Biomaterials Science, Polymer Edition","volume":" ","pages":"2948-2976"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144266325","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The article provides an in-depth overview of the mechanical, chemical, and biological properties of engineering materials used for orthopedic bone plates, along with their designs and fabrication methods. This review addresses the benefits and drawbacks of various materials that have been utilized as bone plates for the treatment of fractures and bone abnormalities. Due to their excellent mechanical properties, metallic bone plates have traditionally been employed for bone fracture fixation. However, the mismatch in mechanical properties and high density of metallic bone plates can lead to stress shielding and non-union, often requiring revision surgeries. These challenges are highlighted in the review, which then explores the potential of polymeric plates to overcome such issues. Nevertheless, the insufficient mechanical performance of polymeric bone plates often necessitates the development of composite bone plates that are patient-specific, biocompatible, and easily tailorable. Emerging research initiatives in this area are discussed. The article further elaborates on various fabrication processes and their impact on the surface properties of bone plates. Both conventional machining processes for internal fixation devices and 3D printing methods for fabricating patient-specific, customized bone plates are reviewed. The paper concludes by evaluating current advancements and anticipated developments related to bone plate technology.
{"title":"A critical review on bone plates: state of the art and future directions.","authors":"Gourav Sardana, Subrata Bandhu Ghosh, Sanchita Bandyopadhyay-Ghosh","doi":"10.1080/09205063.2025.2517717","DOIUrl":"10.1080/09205063.2025.2517717","url":null,"abstract":"<p><p>The article provides an in-depth overview of the mechanical, chemical, and biological properties of engineering materials used for orthopedic bone plates, along with their designs and fabrication methods. This review addresses the benefits and drawbacks of various materials that have been utilized as bone plates for the treatment of fractures and bone abnormalities. Due to their excellent mechanical properties, metallic bone plates have traditionally been employed for bone fracture fixation. However, the mismatch in mechanical properties and high density of metallic bone plates can lead to stress shielding and non-union, often requiring revision surgeries. These challenges are highlighted in the review, which then explores the potential of polymeric plates to overcome such issues. Nevertheless, the insufficient mechanical performance of polymeric bone plates often necessitates the development of composite bone plates that are patient-specific, biocompatible, and easily tailorable. Emerging research initiatives in this area are discussed. The article further elaborates on various fabrication processes and their impact on the surface properties of bone plates. Both conventional machining processes for internal fixation devices and 3D printing methods for fabricating patient-specific, customized bone plates are reviewed. The paper concludes by evaluating current advancements and anticipated developments related to bone plate technology.</p>","PeriodicalId":15195,"journal":{"name":"Journal of Biomaterials Science, Polymer Edition","volume":" ","pages":"3039-3068"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144505818","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
This work presents antibacterial wound dressing membranes based on a nonisocyanate polyurethane-siloxane framework. These membranes protect wounded skin by providing mechanical strength, maintaining a moist environment, and ensuring hygiene through chemically anchored antibacterial moieties. Methoxysilane-functionalized soybean oil-based polyhydroxyurethane with quaternary ammonium groups was synthesized and combined with GPTMS and TEOS. Hydrolysis-condensation reactions formed membranes with siloxane domains and pendant epoxy groups. Gelatin was incorporated to enhance biocompatibility and mechanical strength. The resulting films demonstrated tensile strengths of 7.9 MPa (dry) and 0.61 MPa (swelled). Fluid handling capacities were 2.66-2.81 g/10 cm2/day (serum) and 0.79-1.10 g/10 cm2/day (serum vapor), making them suitable for light to moderately exuding wounds. Cytocompatibility was confirmed by MTT assays, showing over 80% fibroblast viability on dressings and over 90% viability in leachate-containing media. The blood compatibility of the dressing was confirmed by standard methods. The dressings also exhibited strong antibacterial activity, with 82% killing of Staphylococcus aureus and 52% killing of Escherichia coli. These results highlight the potential of these membranes for advanced wound care applications.
{"title":"Gelatin modified nonisocyanate polyurethane/siloxane functionalized with quaternary ammonium groups as antibacterial wound dressing membrane.","authors":"Parsa Mousavi, Hamid Yeganeh, Ismail Omrani, Masoud Babaahmadi","doi":"10.1080/09205063.2025.2518305","DOIUrl":"10.1080/09205063.2025.2518305","url":null,"abstract":"<p><p>This work presents antibacterial wound dressing membranes based on a nonisocyanate polyurethane-siloxane framework. These membranes protect wounded skin by providing mechanical strength, maintaining a moist environment, and ensuring hygiene through chemically anchored antibacterial moieties. Methoxysilane-functionalized soybean oil-based polyhydroxyurethane with quaternary ammonium groups was synthesized and combined with GPTMS and TEOS. Hydrolysis-condensation reactions formed membranes with siloxane domains and pendant epoxy groups. Gelatin was incorporated to enhance biocompatibility and mechanical strength. The resulting films demonstrated tensile strengths of 7.9 MPa (dry) and 0.61 MPa (swelled). Fluid handling capacities were 2.66-2.81 g/10 cm<sup>2</sup>/day (serum) and 0.79-1.10 g/10 cm<sup>2</sup>/day (serum vapor), making them suitable for light to moderately exuding wounds. Cytocompatibility was confirmed by MTT assays, showing over 80% fibroblast viability on dressings and over 90% viability in leachate-containing media. The blood compatibility of the dressing was confirmed by standard methods. The dressings also exhibited strong antibacterial activity, with 82% killing of Staphylococcus aureus and 52% killing of Escherichia coli. These results highlight the potential of these membranes for advanced wound care applications.</p>","PeriodicalId":15195,"journal":{"name":"Journal of Biomaterials Science, Polymer Edition","volume":" ","pages":"3069-3088"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144309958","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-05-26DOI: 10.1080/09205063.2025.2504711
Samridhi Kurl, Gurpreet Kaur
This study aimed to develop a novel polymeric complex composed of Mimosa pudica gum (MMG) and chitosan (CH) and to explore its potential as a delivery system for targeting drugs to the colon. The method of extraction of MMG was optimized, resulting in a maximum yield of 12.41%. The molecular weight of the gum was determined to be 5.07 × 106 Da, and it was characterized for its physicochemical and rheological properties. A species distribution profile was constructed using the pKa values of both polymers, and polyelectrolyte complexes (PECs) were prepared at a pH value of 5.25 ± 0.10. The 40:60 (MMG: CH) PECs exhibited the highest yield (99%), minimal viscosity, and near-neutral zeta potential. Microflora biodegradation studies of PECs in pH 6.8 buffer containing rat cecal contents showed a pH decrease, likely due to degradation products of the PECs. In vitro drug release studies revealed 16.6% capecitabine release (model drug) from PECs without rat cecal contents, compared to 88.5% release after 24h with rat cecal contents. These findings suggest that MMG-CH PECs could serve as promising vehicles for microbially triggered, colon-targeted drug delivery systems.
{"title":"Development, optimization, and characterization of microbially triggered <i>Mimosa pudica</i> gum-chitosan polyelectrolyte complex for colon-targeted drug delivery.","authors":"Samridhi Kurl, Gurpreet Kaur","doi":"10.1080/09205063.2025.2504711","DOIUrl":"10.1080/09205063.2025.2504711","url":null,"abstract":"<p><p>This study aimed to develop a novel polymeric complex composed of <i>Mimosa pudica</i> gum (MMG) and chitosan (CH) and to explore its potential as a delivery system for targeting drugs to the colon. The method of extraction of MMG was optimized, resulting in a maximum yield of 12.41%. The molecular weight of the gum was determined to be 5.07 × 10<sup>6</sup> Da, and it was characterized for its physicochemical and rheological properties. A species distribution profile was constructed using the pKa values of both polymers, and polyelectrolyte complexes (PECs) were prepared at a pH value of 5.25 ± 0.10. The 40:60 (MMG: CH) PECs exhibited the highest yield (99%), minimal viscosity, and near-neutral zeta potential. Microflora biodegradation studies of PECs in pH 6.8 buffer containing rat cecal contents showed a pH decrease, likely due to degradation products of the PECs. <i>In vitro</i> drug release studies revealed 16.6% capecitabine release (model drug) from PECs without rat cecal contents, compared to 88.5% release after 24h with rat cecal contents. These findings suggest that MMG-CH PECs could serve as promising vehicles for microbially triggered, colon-targeted drug delivery systems.</p>","PeriodicalId":15195,"journal":{"name":"Journal of Biomaterials Science, Polymer Edition","volume":" ","pages":"2559-2581"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144150392","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
This work aimed to improve the peroral bioavailability of capecitabine (CPB) by developing and assessing solid lipid nanoparticles (SLNs). SLNs were made using the modified nanoprecipitation method. Particle size, zeta potential, entrapment efficiency, drug loading, in-vitro drug release, TEM, in-vivo pharmacokinetic study, stability study, histopathological evaluation and cytotoxicity study were assessed. The TEM revealed that the SLNs were transparent, with a mean particle size ranging from 13.06 ± 0.09 to 86.10 ± 0.15 nm. The F-3 formulation demonstrated the highest drug entrapment efficiency at 45.49 ± 0.28. The zeta potential and polydispersity index of all SLNs ranged from -15.53 ± 0.17 to 17.55 ± 0.18 mV and from 0.1356 ± 0.11 to 0.2678 ± 0.13, respectively. The drug entrapment efficiency and drug loading of all SLNs ranged from 18.45 ± 0.36 to 45.49 ± 0.28 and from 21.75 ± 0.64 to 59.49 ± 0.38, respectively. The CPB-SLNs showed sustained drug release with prolonged plasma retention, delayed Tmax, and extended half-life compared to raw CPB. In vivo pharmacokinetic studies suggest that developed SLNs may enhance therapeutic efficacy by maintaining drug concentrations in plasma for longer periods. Toxicity was observed at 200 mg/kg/day, indicated by changes in clinical biochemistry, organ weights, and histopathology, particularly affecting the liver and kidneys. Therefore, it can be said that these developed SLNs may be among the best preparations for the delivery of anti-cancer drugs for improved therapeutic efficacy.
{"title":"Development and evaluation of solid lipid nanoparticles for enhanced peroral bioavailability of capecitabine.","authors":"Mayukh Jana, Chandra Sekhar Patro, Suraj Sharma, Sweet Naskar, Ujjwal Kumar Biswas, Biplab Debnath","doi":"10.1080/09205063.2025.2505344","DOIUrl":"10.1080/09205063.2025.2505344","url":null,"abstract":"<p><p>This work aimed to improve the peroral bioavailability of capecitabine (CPB) by developing and assessing solid lipid nanoparticles (SLNs). SLNs were made using the modified nanoprecipitation method. Particle size, zeta potential, entrapment efficiency, drug loading, <i>in-vitro</i> drug release, TEM, <i>in-vivo</i> pharmacokinetic study, stability study, histopathological evaluation and cytotoxicity study were assessed. The TEM revealed that the SLNs were transparent, with a mean particle size ranging from 13.06 ± 0.09 to 86.10 ± 0.15 nm. The F-3 formulation demonstrated the highest drug entrapment efficiency at 45.49 ± 0.28. The zeta potential and polydispersity index of all SLNs ranged from -15.53 ± 0.17 to 17.55 ± 0.18 mV and from 0.1356 ± 0.11 to 0.2678 ± 0.13, respectively. The drug entrapment efficiency and drug loading of all SLNs ranged from 18.45 ± 0.36 to 45.49 ± 0.28 and from 21.75 ± 0.64 to 59.49 ± 0.38, respectively. The CPB-SLNs showed sustained drug release with prolonged plasma retention, delayed Tmax, and extended half-life compared to raw CPB. <i>In vivo</i> pharmacokinetic studies suggest that developed SLNs may enhance therapeutic efficacy by maintaining drug concentrations in plasma for longer periods. Toxicity was observed at 200 mg/kg/day, indicated by changes in clinical biochemistry, organ weights, and histopathology, particularly affecting the liver and kidneys. Therefore, it can be said that these developed SLNs may be among the best preparations for the delivery of anti-cancer drugs for improved therapeutic efficacy.</p>","PeriodicalId":15195,"journal":{"name":"Journal of Biomaterials Science, Polymer Edition","volume":" ","pages":"2582-2607"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144078106","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-05-26DOI: 10.1080/09205063.2025.2509028
Qi Yang, Xi Xiang, Han Wang, Yue Liao, Xinzhi Li
Oral administration, owing to its high patient compliance and favorable controllability, is widely employed in clinical settings; however, the efficacy is often constrained by the gastrointestinal environment's impact on bioavailability. As the demand for biocompatibility and biodegradability in biomedical applications intensifies, natural hydrogel-based oral drug delivery systems have gained substantial attention as promising carriers. In this study, we introduce a variety of natural materials, revealing their advantages in enhancing drug bioavailability and targeting capabilities. Through both physical and chemical crosslinking mechanisms, we successfully demonstrate hydrogels exhibiting excellent mechanical properties and biocompatibility. Furthermore, we analyze the potential applications of diverse natural oral hydrogels across fields such as gastrointestinal, metabolic, oncological, and immunotherapeutic diseases. By synthesizing recent advances in this area, we aim to elucidate the critical role these systems can play in biomedicine. Our findings suggest that natural materials possess broad prospects in drug delivery, advocating for continued exploration of their clinical application to facilitate the development and optimization of novel oral therapeutic modalities. This work provides a vital theoretical foundation and practical guidance for future innovations in drug delivery technologies.
{"title":"Oral natural material hydrogels: a new strategy for enhancing oral drug delivery efficiency.","authors":"Qi Yang, Xi Xiang, Han Wang, Yue Liao, Xinzhi Li","doi":"10.1080/09205063.2025.2509028","DOIUrl":"10.1080/09205063.2025.2509028","url":null,"abstract":"<p><p>Oral administration, owing to its high patient compliance and favorable controllability, is widely employed in clinical settings; however, the efficacy is often constrained by the gastrointestinal environment's impact on bioavailability. As the demand for biocompatibility and biodegradability in biomedical applications intensifies, natural hydrogel-based oral drug delivery systems have gained substantial attention as promising carriers. In this study, we introduce a variety of natural materials, revealing their advantages in enhancing drug bioavailability and targeting capabilities. Through both physical and chemical crosslinking mechanisms, we successfully demonstrate hydrogels exhibiting excellent mechanical properties and biocompatibility. Furthermore, we analyze the potential applications of diverse natural oral hydrogels across fields such as gastrointestinal, metabolic, oncological, and immunotherapeutic diseases. By synthesizing recent advances in this area, we aim to elucidate the critical role these systems can play in biomedicine. Our findings suggest that natural materials possess broad prospects in drug delivery, advocating for continued exploration of their clinical application to facilitate the development and optimization of novel oral therapeutic modalities. This work provides a vital theoretical foundation and practical guidance for future innovations in drug delivery technologies.</p>","PeriodicalId":15195,"journal":{"name":"Journal of Biomaterials Science, Polymer Edition","volume":" ","pages":"2758-2785"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144150398","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-05-29DOI: 10.1080/09205063.2025.2510445
Gunjan Adwani, Sharda Bharti, Awanish Kumar
Millions of individuals worldwide suffer from a chronic metabolic disorder, diabetes, defined as a reduction in insulin production or sensitivity, which raises blood glucose levels, weakens the immune system, and results in irregularities in the metabolism of carbohydrates, fats, and proteins. Therefore, there is still a high demand for non-invasive ways to administer insulin and other antidiabetics to treat diabetes and suitable therapeutics for wound healing. This generates a need for novel biomaterials that effectively use diabetes-associated therapy. This article emphasized that some special -features of Polyhydroxyalkanoates (PHAs) are biocompatible, biodegradable thermoplastic polyesters used in biomedical applications, expanding the options for bioresorbable polymers having antidiabetic and antimicrobial activities. PHAs can be synthesized into scaffolds and nanomaterials that release insulin and other antidiabetic medications in a sustained and controlled way that could improve treatment results. Research analysis on the application of PHAs as scaffold materials for bioartificial pancreas development offers a biocompatible and structurally supportive environment to encapsulate pancreatic cells. Further, challenges including excessive production costs, requirement for additional clinical setting optimization, and the current status of PHAs in the market are emphasized in this review. Further research is needed to explore the therapeutic potential of PHAs exhaustively in diabetes therapeutics and management.
{"title":"Exploring multi-functional biopolymer polyhydroxyalkanoates in diabetes treatment.","authors":"Gunjan Adwani, Sharda Bharti, Awanish Kumar","doi":"10.1080/09205063.2025.2510445","DOIUrl":"10.1080/09205063.2025.2510445","url":null,"abstract":"<p><p>Millions of individuals worldwide suffer from a chronic metabolic disorder, diabetes, defined as a reduction in insulin production or sensitivity, which raises blood glucose levels, weakens the immune system, and results in irregularities in the metabolism of carbohydrates, fats, and proteins. Therefore, there is still a high demand for non-invasive ways to administer insulin and other antidiabetics to treat diabetes and suitable therapeutics for wound healing. This generates a need for novel biomaterials that effectively use diabetes-associated therapy. This article emphasized that some special -features of Polyhydroxyalkanoates (PHAs) are biocompatible, biodegradable thermoplastic polyesters used in biomedical applications, expanding the options for bioresorbable polymers having antidiabetic and antimicrobial activities. PHAs can be synthesized into scaffolds and nanomaterials that release insulin and other antidiabetic medications in a sustained and controlled way that could improve treatment results. Research analysis on the application of PHAs as scaffold materials for bioartificial pancreas development offers a biocompatible and structurally supportive environment to encapsulate pancreatic cells. Further, challenges including excessive production costs, requirement for additional clinical setting optimization, and the current status of PHAs in the market are emphasized in this review. Further research is needed to explore the therapeutic potential of PHAs exhaustively in diabetes therapeutics and management.</p>","PeriodicalId":15195,"journal":{"name":"Journal of Biomaterials Science, Polymer Edition","volume":" ","pages":"2859-2890"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144173937","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Podophyllotoxin (PPT), a bioactive compound, shows promise as a potential cancer treatment drug. Nevertheless, low solubility and bioavailability of PPT necessitate a drug delivery system to improve its effectiveness. PPT was extracted from Linum album and delivered into HepG2 cancer cells using mPEG-PCL nanoparticles. Copolymers were synthesized and confirmed by UV-Vis, FTIR, 1HNMR, XRD, FESEM analyses, and the other physicochemical properties were also characterized. The critical micelle concentration of the copolymers was calculated, and the ratio of 1:10 with a CMC of 0.055 µg. mL-1 was selected as the optimal ratio. The average size and surface charge of micelles were 186 ± 12 nm and -5.13 ± 0.61 mV, respectively. FESEM analysis showed a uniform and spherical structure of nanoparticles. PPT was loaded into mPEG-PCL micelles in various ratios (w/w) of drug: copolymer using the nanoprecipitation method, and the ratio of 1:1 was selected as the optimal ratio with encapsulation and loading efficiency of 79.89 ± 1.28% and 10.15 ± 2.16%, respectively. The PPT release profile demonstrated a significant difference between the sustained release of PPT from the nanoparticles and the rapid release of free PPT. Cellular uptake studies revealed that the polymersomes effectively deliver the PPT to the HepG2 cells. The in vitro cytotoxicity assay showed increased cytotoxicity of PPT/mPEG-PCL NPs compared to the free drug. Based on the overall results, these nanoparticles show promise as a delivery system for controlled release of PPT in cancer therapy.
{"title":"Enhanced delivery of podophyllotoxin for hepatocellular carcinoma therapy using polymersome as an anticancer delivery platform.","authors":"Parvaneh Peyvand, Zahra Vaezi, Mohsen Sharifi, Hossein Naderi-Manesh","doi":"10.1080/09205063.2025.2520687","DOIUrl":"10.1080/09205063.2025.2520687","url":null,"abstract":"<p><p>Podophyllotoxin (PPT), a bioactive compound, shows promise as a potential cancer treatment drug. Nevertheless, low solubility and bioavailability of PPT necessitate a drug delivery system to improve its effectiveness. PPT was extracted from Linum album and delivered into HepG2 cancer cells using mPEG-PCL nanoparticles. Copolymers were synthesized and confirmed by UV-Vis, FTIR, <sup>1</sup>HNMR, XRD, FESEM analyses, and the other physicochemical properties were also characterized. The critical micelle concentration of the copolymers was calculated, and the ratio of 1:10 with a CMC of 0.055 µg. mL<sup>-1</sup> was selected as the optimal ratio. The average size and surface charge of micelles were 186 ± 12 nm and -5.13 ± 0.61 mV, respectively. FESEM analysis showed a uniform and spherical structure of nanoparticles. PPT was loaded into mPEG-PCL micelles in various ratios (w/w) of drug: copolymer using the nanoprecipitation method, and the ratio of 1:1 was selected as the optimal ratio with encapsulation and loading efficiency of 79.89 ± 1.28% and 10.15 ± 2.16%, respectively. The PPT release profile demonstrated a significant difference between the sustained release of PPT from the nanoparticles and the rapid release of free PPT. Cellular uptake studies revealed that the polymersomes effectively deliver the PPT to the HepG2 cells. The <i>in vitro</i> cytotoxicity assay showed increased cytotoxicity of PPT/mPEG-PCL NPs compared to the free drug. Based on the overall results, these nanoparticles show promise as a delivery system for controlled release of PPT in cancer therapy.</p>","PeriodicalId":15195,"journal":{"name":"Journal of Biomaterials Science, Polymer Edition","volume":" ","pages":"3150-3172"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144325862","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}