{"title":"Ginsenoside compound K-based multifunctional liposomes for the treatment of rheumatoid arthritis.","authors":"Meng Zhang, Ru Zhang, Chunbo Feng, Xinnan Jiang, Xinchun Xu, Jianxin Wang","doi":"10.1080/10717544.2025.2464190","DOIUrl":"10.1080/10717544.2025.2464190","url":null,"abstract":"<p><p>The clinical treatment of rheumatoid arthritis (RA) with first-line therapeutic drugs is hindered by the poor solubility, low bioavailability, off-target toxicity, and insufficient accumulation in inflamed joints. Liposomes have been shown to mitigate some of these limitations in drug delivery systems. However, the use of cholesterol to stabilize liposomal structures remains controversial due to its potential association with cardiovascular diseases. Here, we developed a novel liposome based on ginsenoside compound K (CK), which not only serves as an effective therapeutic agent for RA but also replaces cholesterol as a membrane stabilizer to address these challenges. Compared with conventional liposomes, ginsenoside CK Liposomes (CK@Lipo) are excellent nanoparticles, with CK stabilizing the liposomal structure and providing targeting functionality toward inflamed joints. When encapsulated with dexamethasone (Dex), CK@Lipo exhibits a synergistic anti-inflammatory effect, slowing the progression of RA. This study provides a theoretical basis for the future development of multifunctional novel ginsenoside CK@Lipo.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2464190"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11834820/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143432673","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-02-04DOI: 10.1080/10717544.2025.2459775
Soomin Lee, Mi-Young Jung, Choul Yong Park
{"title":"Development of a conjunctival contact-type drug delivery device for latanoprost using hyaluronic acid.","authors":"Soomin Lee, Mi-Young Jung, Choul Yong Park","doi":"10.1080/10717544.2025.2459775","DOIUrl":"10.1080/10717544.2025.2459775","url":null,"abstract":"<p><p>Effective topical drug delivery is crucial for glaucoma treatment, necessitating more convenient methods to enhance patient compliance. This study evaluates the efficacy and safety of using freeze-dried hyaluronic acid (HA) as a carrier for a novel conjunctival-contact drug delivery system. We developed HA tablets loaded with latanoprost (HA-latanoprost) and verified the concentration using high-performance liquid chromatography. Twenty mice (C57BL6) were divided into four groups (<i>n</i> = 5 per group): normal saline (group 1), control HA tablet (group 2), Xalatan™ (group 3), and HA-latanoprost tablet (group 4). Treatments were administered to the right eyes, with the left eyes serving as no-treatment controls. Intraocular pressure (IOP) and irritation (measured by scratching motions) were monitored for 10 days. On day 10, we quantified gene expression of inflammatory cytokines and IOP-affecting proteins using polymerase chain reaction, and performed histological and immunohistochemical analyses. Results showed that IOP was significantly lower in groups 3 and 4 compared to the other groups, with group 4 exhibiting the greatest reduction by day 10. Group 4 also experienced less irritation. Additionally, group 4 had lower expression of inflammatory cytokine genes and higher expression of IOP-lowering protein genes compared to group 3. No significant side effects were observed in any group. Overall, HA-latanoprost effectively lowered IOP and reduced ocular irritation more than latanoprost eyedrops in mice. However, these results are based on animal testing, so further development is needed for clinical use.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2459775"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11795754/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143188641","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-02-01DOI: 10.1080/10717544.2025.2459772
Rushikesh Girase, Nayan A Gujarathi, Amey Sukhia, Sri Sai Nikitha Kota, Tulshidas S Patil, Abhijeet A Aher, Yogeeta O Agrawal, Shreesh Ojha, Charu Sharma, Sameer N Goyal
{"title":"Targeted nanoliposomes for precision rheumatoid arthritis therapy: a review on mechanisms and <i>in vivo</i> potential.","authors":"Rushikesh Girase, Nayan A Gujarathi, Amey Sukhia, Sri Sai Nikitha Kota, Tulshidas S Patil, Abhijeet A Aher, Yogeeta O Agrawal, Shreesh Ojha, Charu Sharma, Sameer N Goyal","doi":"10.1080/10717544.2025.2459772","DOIUrl":"10.1080/10717544.2025.2459772","url":null,"abstract":"<p><p>Rheumatoid arthritis (RA) is an inflammatory immune-triggered disease that causes synovitis, cartilage degradation, and joint injury. In nanotechnology, conventional liposomes were extensively investigated for RA. However, they frequently undergo rapid clearance, reducing circulation time and therapeutic efficacy. Additionally, their stability in the bloodstream is often compromised, resulting in premature drug release. The current review explores the potential of targeted liposomal-based nanosystems in the treatment of RA. It highlights the pathophysiology of RA, explores selective targeting sites, and elucidates diverse mechanisms of novel liposomal types and their applications. Furthermore, the targeting strategies of pH-sensitive, flexible, surface-modified, PEGylated, acoustic, ROS-mediated, and biofunctionalized liposomes are addressed. Targeted nanoliposomes showed potential in precisely delivering drugs to CD44, SR-A, FR-β, FLS, and toll-like receptors through the high affinity of ligands. <i>In vitro</i> studies interpreted stable release profiles and improved stability. <i>Ex vivo</i> studies on skin demonstrated that ultradeformable and glycerol-conjugated liposomes enhanced drug penetrability. <i>In vivo</i> experiments for liposomal types in the arthritis rat model depicted remarkable efficacy in reducing joint swelling, pro-inflammatory cytokines, and synovial hyperplasia. In conclusion, these targeted liposomes represented a significant leap forward in drug delivery, offering effective therapeutic options for RA. In the future, integrating these advanced liposomes with artificial intelligence, immunotherapy, and precision medicine holds great promise.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2459772"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11789225/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143074272","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-02-03DOI: 10.1080/10717544.2025.2460693
Maryana Salamah, Bence Sipos, Zsuzsanna Schelz, István Zupkó, Ágnes Kiricsi, Ágnes Szalenkó-Tőkés, László Rovó, Gábor Katona, György Tibor Balogh, Ildikó Csóka
{"title":"Development, <i>in vitro</i> and <i>ex vivo</i> characterization of lamotrigine-loaded bovine serum albumin nanoparticles using QbD approach.","authors":"Maryana Salamah, Bence Sipos, Zsuzsanna Schelz, István Zupkó, Ágnes Kiricsi, Ágnes Szalenkó-Tőkés, László Rovó, Gábor Katona, György Tibor Balogh, Ildikó Csóka","doi":"10.1080/10717544.2025.2460693","DOIUrl":"10.1080/10717544.2025.2460693","url":null,"abstract":"<p><p>The present study aimed to prepare and optimize lamotrigine-loaded bovine serum albumin nanoparticles (LAM-NP) using the Quality by Design (QbD) approach and to investigate both the <i>in vitro</i> and <i>ex vivo</i> effects of different cross-linking agents glutaraldehyde (GLUT), glucose (GLUC) and 1-(3-dimethylaminutesopropyl)-3-ethylcarbodiimide hydrochloride (EDC) on intranasal applicability. Cross-linked LAM-NP from EDC (NP-EDC-1) showed the lowest Z-average value (163.7 ± 1.9 nm) and drug encapsulation efficacy (EE%) of 97.31 ± 0.17%. The drug release of GLUC cross-linked LAM-NP (NP-GLUC-9), glutaraldehyde cross-linked LAM-NP (NP-GLUT-2), and NP-EDC-1 at blood circulation conditions was higher than the initial LAM. The results of the blood-brain barrier parallel artificial membrane permeability assay (BBB-PAMPA) showed an increase in the permeability of LAM through the BBB with NP-GLUC-9 and an increase in flux with all selected formulations. The <i>ex vivo</i> study showed that LAM diffusion from the selected formulations through the human nasal mucosa was higher than in case of initial LAM. The cytotoxicity study indicated that BSA-NP reduced LAM toxicity, and GLUC 9 mM and EDC 1 mg could be alternative cross-linking agents to avoid GLUT 2% v/v toxicity. Furthermore, permeability through Caco-2 cells showed that nasal epithelial transport/absorption of LAM was improved by using BSA-NPs. The use of BSA-NP may be a promising approach to enhance the solubility, permeability through BBB and decrease the frequency of dosing and adverse effects of LAM.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2460693"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11795762/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143122491","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-02-10DOI: 10.1080/10717544.2025.2463433
Kirsi Toivanen, Luna De Sutter, Agnieszka Wozniak, Karo Wyns, Nanna Merikoski, Sami Salmikangas, Jianmin Duan, Mikael Maksimow, Maria Lahtinen, Tom Böhling, Patrick Schöffski, Harri Sihto
{"title":"Pharmacokinetic profile and <i>in vivo</i> anticancer efficacy of anagrelide administered subcutaneously in rodents.","authors":"Kirsi Toivanen, Luna De Sutter, Agnieszka Wozniak, Karo Wyns, Nanna Merikoski, Sami Salmikangas, Jianmin Duan, Mikael Maksimow, Maria Lahtinen, Tom Böhling, Patrick Schöffski, Harri Sihto","doi":"10.1080/10717544.2025.2463433","DOIUrl":"10.1080/10717544.2025.2463433","url":null,"abstract":"<p><p>Anagrelide (ANA) is a phosphodiesterase 3A (PDE3A) inhibitor, commonly prescribed for essential thrombocythemia. It also functions as a molecular glue, inducing complex formation between PDE3A and Schlafen 12. This association either triggers apoptosis or inhibits proliferation in tumor cells, supporting its use in cancer therapy. Conventionally administered orally, ANA undergoes rapid metabolism and elimination, resulting in a short drug exposure time at the site of action. Here, we explored the pharmacokinetic profile of a subcutaneously (SC) injected ANA formulation in Sprague-Dawley rats by quantifying plasma ANA and metabolite concentrations using liquid-chromatography-tandem mass spectrometry. We further evaluated the <i>in vivo</i> tumor regression efficacy of orally and SC administered ANA in a patient-derived gastrointestinal stromal xenograft mouse model - UZLX-GIST2B - characterized by a <i>KIT</i> exon 9 driver mutation. The SC ANA exhibited extended-release plasma concentration-time profiles compared to intravenous and oral administrations. After a single administration in rats, plasma concentrations of ANA were detected up to 56 days later, and ANA metabolites up to 30 days later. The SC formulation also significantly reduced tumor volumes and demonstrated dose-dependent histological responses, nearly eradicating tumor tissue in 11 days with the highest dose. These findings suggest that the SC slow-release formulation maintains stable drug concentrations during treatment, potentially improving therapeutic efficacy at the target site.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2463433"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11816618/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143390246","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-02-06DOI: 10.1080/10717544.2025.2460666
Inga Jurgelane, Karina Egle, Andra Grava, Dana Galkina, Margarita Brante, Maksims Melnichuks, Marite Skrinda-Melne, Girts Salms, Arita Dubnika
{"title":"Exploring the effects of cannabidiol encapsulation in liposomes on their physicochemical properties and biocompatibility.","authors":"Inga Jurgelane, Karina Egle, Andra Grava, Dana Galkina, Margarita Brante, Maksims Melnichuks, Marite Skrinda-Melne, Girts Salms, Arita Dubnika","doi":"10.1080/10717544.2025.2460666","DOIUrl":"10.1080/10717544.2025.2460666","url":null,"abstract":"<p><p>Cannabidiol (CBD) is recognized for its therapeutic properties in various conditions. However, CBD's limited water solubility and sensitivity to environmental stresses hinder its efficacy and bioavailability. Encapsulation in drug delivery systems, particularly liposomes, offers a promising solution. This study aims to prepare CBD-containing liposomes using commercially used lipids distearoyl phosphatidylcholine (DSPC) and dipalmitoyl phosphatidylcholine (DPPC), and 1,2 distearoyl-sn-glycero-3 phosphoethanolamine-N-[carbonyl-amino(polyethylene glycol)-4300] (ammonium salt) (DSPE-PEG) and to perform <i>in vitro</i> studies - cell viability and CBD release. Liposomes were synthesized using thin-film hydration method, and characterized by Fourier-transform infrared (FT-IR) spectroscopy, dynamic light scattering (DLS), and scanning transmission electron microscopy (STEM). DLS analysis revealed that CBD incorporation reduced liposome size by 23-53%, depending on the liposomes. Encapsulation efficiency followed the order: DPPC CBD (63%) < DSPC CBD (74%) < DSPC DPPC CBD (81%) < DSPC DSPE-PEG CBD (87%). CBD release profiles indicated that DPPC CBD liposomes released the highest CBD amount initially, while DSPC DSPE-PEG CBD exhibited sustained release, achieving 79% release over 504 h. <i>In vitro</i> cell viability tests showed that blank liposomes were non-cytotoxic. However, CBD-loaded liposomes significantly reduced cell viability for defined type of CBD containing liposomes. The inclusion of DSPE-PEG improved encapsulation efficiency and liposome stability, making DSPC DSPE-PEG CBD liposomes more suitable for long-term CBD release. Compared to other studies, encapsulation of CBD in liposomes enhances its bioavailability, allowing lower concentrations of CBD to be directly delivered to cells, resulting in observable changes in cell viability.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2460666"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11809167/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143364061","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Drug DeliveryPub Date : 2025-12-01Epub Date: 2025-03-24DOI: 10.1080/10717544.2025.2476140
Shuting Xu, Yaming Zhang, Jia Li, Xinyu Zhang, Weiping Wang
{"title":"External stimuli-responsive drug delivery to the posterior segment of the eye.","authors":"Shuting Xu, Yaming Zhang, Jia Li, Xinyu Zhang, Weiping Wang","doi":"10.1080/10717544.2025.2476140","DOIUrl":"10.1080/10717544.2025.2476140","url":null,"abstract":"<p><p>Posterior segment eye diseases represent the leading causes of vision impairment and blindness globally. Current therapies still have notable drawbacks, including the need for frequent invasive injections and the associated risks of severe ocular complications. Recently, the utility of external stimuli, such as light, ultrasound, magnetic field, and electric field, has been noted as a promising strategy to enhance drug delivery to the posterior segment of the eye. In this review, we briefly summarize the main physiological barriers against ocular drug delivery, focusing primarily on the recent advancements that utilize external stimuli to improve treatment outcomes for posterior segment eye diseases. The advantages of these external stimuli-responsive drug delivery strategies are discussed, with illustrative examples highlighting improved tissue penetration, enhanced control over drug release, and targeted drug delivery to ocular lesions through minimally invasive routes. Finally, we discuss the challenges and future perspectives in the translational research of external stimuli-responsive drug delivery platforms, aiming to bridge existing gaps toward clinical use.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2476140"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11934192/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143691137","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Harnessing nanofibers for targeted delivery of phytoconstituents in age-related macular degeneration.","authors":"Ulia Andrades, Sahil Gaikar, Khushali Nathani, Sujata Sawarkar, Abdelwahab Omri","doi":"10.1080/10717544.2025.2489491","DOIUrl":"https://doi.org/10.1080/10717544.2025.2489491","url":null,"abstract":"<p><p>Age-related macular degeneration is a degenerative eye condition that affects the macula and results in central vision loss. Phytoconstituents show great promise in the treatment of AMD. AMD therapy can benefit from the advantages of phytoconstituents loaded nanofibers. There are opportunities to improve the effectiveness of phytoconstituents in the treatment of age-related macular degeneration (AMD) through the use of nanofiber-based delivery methods. These novel platforms encapsulate and distribute plant-derived bioactives by making use of the special qualities of nanofibers. These qualities include their high surface area-to-volume ratio, variable porosity, and biocompatibility. Exploring the use of nanofiber-based delivery methods to provide phytoconstituents in AMD treatment is a great choice for enhancing patient adherence, safety, and efficacy in managing this condition. This article explores the potential of nanofiber-based delivery methods to revolutionize AMD treatment, providing an innovative and effective approach to treat this condition.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2489491"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143794964","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Preparation, quality evaluation and preliminary pharmacokinetic-pharmacodynamic studies of synephrine dry powder inhaler.","authors":"Jiming Ke, Shenao Li, Miaomiao Zi, Jing Zhang, Shan Huang, Wenhui Luo, Hailun Han, Jiwen Zhang, Can Peng","doi":"10.1080/10717544.2025.2486346","DOIUrl":"https://doi.org/10.1080/10717544.2025.2486346","url":null,"abstract":"<p><p>Acute lung injury (ALI) is a lung disease characterized by pulmonary edema caused by an excessive inflammatory response within the lungs and disruption of the alveolar capillary barrier, with a high morbidity and mortality rate in critically ill patients. Dry powder inhalers (DPI) are an effective way of administering medication to improve efficacy, and inhalation administration not only improves efficacy but also increases the bioavailability of the drug. Synephrine, a natural ingredient derived from the fruit of the citrus plant in the <i>Brassicaceae family</i>, has anti-inflammatory and antioxidant properties. In the present study, we prepared a synephrine dry powder inhaler (SYN-DPI) by anti-solvent precipitation method and evaluated it <i>in vivo</i> and <i>in vitro</i>. The <i>in vitro</i> results show that SYN-DPI has low hygroscopicity and good aerodynamic properties. The <i>in vitro</i> and <i>in vivo</i> efficacy results showed that SYN-DPI not only had low toxicity but also possessed good anti-inflammatory and antioxidant capacity, which could significantly reduce inflammation, oxidative stress, and lung injury. Pharmacokinetic results showed that inhalation administration significantly increased SYN bioavailability. In conclusion, this study provides inhalation administration of synephrine as an inhalable formulation that can be used to improve ALI.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2486346"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143802832","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Biomimetic peptide conjugates as emerging strategies for controlled release from protein-based materials.","authors":"Juthatip Manissorn, Jaturong Promsuk, Kittikhun Wangkanont, Peerapat Thongnuek","doi":"10.1080/10717544.2025.2449703","DOIUrl":"10.1080/10717544.2025.2449703","url":null,"abstract":"<p><p>Biopolymers, such as collagens, elastin, silk fibroin, spider silk, fibrin, keratin, and resilin have gained significant interest for their potential biomedical applications due to their biocompatibility, biodegradability, and mechanical properties. This review focuses on the design and integration of biomimetic peptides into these biopolymer platforms to control the release of bioactive molecules, thereby enhancing their functionality for drug delivery, tissue engineering, and regenerative medicine. Elastin-like polypeptides (ELPs) and silk fibroin repeats, for example, demonstrate how engineered peptides can mimic natural protein domains to modulate material properties and drug release profiles. Recombinant spider silk proteins, fibrin-binding peptides, collagen-mimetic peptides, and keratin-derived structures similarly illustrate the ability to engineer precise interactions and to design controlled release systems. Additionally, the use of resilin-like peptides showcases the potential for creating highly elastic and resilient biomaterials. This review highlights current achievements and future perspectives in the field, emphasizing the potential of biomimetic peptides to transform biopolymer-based biomedical applications.</p>","PeriodicalId":11679,"journal":{"name":"Drug Delivery","volume":"32 1","pages":"2449703"},"PeriodicalIF":6.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11721625/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142946591","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}