1
|
Zhang M, Qiu H, Han Z, Ma Y, Hou J, Yuan J, Jia H, Zhou M, Lu H, Wu Y. Topical transdermal administration of lenalidomide nanosuspensions-based hydrogels against melanoma: In vitro and in vivo studies. Int J Pharm X 2025; 9:100316. [PMID: 39898009 PMCID: PMC11787432 DOI: 10.1016/j.ijpx.2025.100316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 02/04/2025] Open
Abstract
Percutaneous neoadjuvant therapy has proven effective in diminishing tumor size and the surgical intervention area, which couldeffectively mitigate the risk of tumor recurrence and enhance immunotherapy efficacy. Lenalidomide, an approved medication orally used to treat myeloma, was loaded into nanosuspensions-based hydrogels (Len-NBHs) for transdermal administration as a percutaneous neoadjuvant therapy. This study was designed to investigate the inhibitory effect and mechanism of Len-NBHs on melanoma. Network pharmacology and transcriptomic analyses identified key targets and signaling pathways. The effects of lenalidomide on melanoma were further verified through Western blotting, immunohistochemistry, immunofluorescence, and quantitative real-time polymerase chain reaction,using both in vitro cell experiments and in vivo melanoma mouse models. Lenalidomide could induce melanoma cells apoptosis, disrupt cell cycle progression, impede cell migration and invasion, and modify tumor microenvironment (TME). Mechanistically, lenalidomide reversed the abnormal activation of the PI3K-AKT signaling pathway and the overexpression of CD93, while also recruiting CD8+ T cells, CD4+ T cells, and dendritic cells to infiltrate the tumor site. Transdermal administration of Len-NBHs represents a promising adjuvant therapy for the treatment of malignant melanoma. Preoperative administration of Len-NBHs can inhibit the outward spread of melanoma, reduce tumor size, thereby decreasing the surgical excision area and improving patient survival rates and prognosis.
Collapse
Affiliation(s)
| | | | - Zheyi Han
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Yazhong Ma
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Jingjing Hou
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Jingwei Yuan
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Haiyan Jia
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Menglu Zhou
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Hongjie Lu
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Yan Wu
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| |
Collapse
|
2
|
Fladung L, Langwald SV, Kruse O, Patel A. A novel indicator-based visualisation method to investigate diffusion behaviour of dissolved CO 2 in hydrogels. MethodsX 2025; 14:103225. [PMID: 40093573 PMCID: PMC11910120 DOI: 10.1016/j.mex.2025.103225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/15/2025] [Indexed: 03/19/2025] Open
Abstract
Biocompatible hydrogels are versatile platforms for encapsulating living cells in biotechnology due to their unique physical, structural and mechanical properties. The diffusion of dissolved carbon dioxide (dCO2) into the hydrogel matrix is of great importance for the growth of immobilised photosynthetic cells like microalgae and cyanobacteria. However, non-invasive analysis methods for measuring the diffusion of dCO2 in hydrogels are limited. In this article, we describe an indirect method for the non-invasive measurement of diffusion rates for dCO2 in hydrogels. We visually tracked the diffusion along the axial direction of pH indicator-doped hydrogel monoliths by recording the interface position over time. We calculated the interface velocity and the pseudo diffusion coefficients (Dpseudo) over time. The obtained Dpseudo values are in a realistic range compared to literature values. Therefore, this novel analysis method for dCO2 diffusion gained valuable insights into diffusion dynamics in different hydrogels and can aid in the design of better immobilisation matrices for photosynthetic cells.•Non-invasive, rapid method for estimation of dissolved CO2 (dCO2) diffusion in hydrogels•Automatic analysis of colour interface formation due to acidification of hydrogels by diffusing dCO2•Agarose hydrogels exhibit an approximated 30x higher pseudo dCO2 diffusion coefficient than silica gel.
Collapse
Affiliation(s)
- Laura Fladung
- WG Fermentation and Formulation of Biologicals and Chemicals, Faculty of Engineering and Mathematics, Hochschule Bielefeld - University of Applied Sciences and Arts, Interaktion 1, 33619 Bielefeld, Germany
- WG Algae Biotechnology and Bioenergy, Center for Biotechnology, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Sarah Vanessa Langwald
- WG Fermentation and Formulation of Biologicals and Chemicals, Faculty of Engineering and Mathematics, Hochschule Bielefeld - University of Applied Sciences and Arts, Interaktion 1, 33619 Bielefeld, Germany
| | - Olaf Kruse
- WG Algae Biotechnology and Bioenergy, Center for Biotechnology, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Anant Patel
- WG Fermentation and Formulation of Biologicals and Chemicals, Faculty of Engineering and Mathematics, Hochschule Bielefeld - University of Applied Sciences and Arts, Interaktion 1, 33619 Bielefeld, Germany
| |
Collapse
|
3
|
Uchuwittayakul A, Thompson KD, Thangsunan P, Phaksopa J, Buncharoen W, Saenphet K, Kumwan B, Meachasompop P, Saenphet S, Wiratama N, Mahatnirunkul T, Kantha P, Suree N, Thangsunan P, Srisapoome P. Evaluation of a hydrogel platform for encapsulated multivalent Vibrio antigen delivery to enhance immune responses and disease protection against vibriosis in Asian seabass (Lates calcarifer). FISH & SHELLFISH IMMUNOLOGY 2025; 160:110230. [PMID: 40015493 DOI: 10.1016/j.fsi.2025.110230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/15/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
This study reports the development and evaluation of a novel multivalent oral hydrogel vaccine designed to protect Asian seabass (Lates calcarifer) against vibriosis caused by multiple Vibrio species. The hydrogel formulation, composed of alginate and bentonite, was engineered to encapsulate three Vibrio pathogens (V. harveyi, V. vulnificus, and P. damsela), and subsequently freeze-dried to yield stable, dry hydrogel beads. Although the process achieved a relatively low yield (approximately 10 %), the dry beads maintained their structural integrity, retained antigenic components with 2.4 ± 1.6 × 107 DNA copies of total cell antigens/mg dry beads, and resisted acidic degradation, ensuring antigen preservation during simulated gastric exposure. Physicochemical characterization (FTIR) confirmed that the encapsulation process preserved the structural and functional properties of alginate, bentonite, and bacterial antigens without introducing new chemical bonds. Scanning electron microscopy (SEM) revealed a porous, sponge-like internal architecture that supported antigen entrapment and controlled release. Under simulated gastric conditions (pH 2.0), the hydrogel exhibited remarkable stability for up to 8 h, preventing antigen loss. Upon transitioning to intestinal conditions (pH 7.2), the matrix gradually disintegrated, releasing the antigens in a controlled manner for up to 8 h. Oral vaccination trials demonstrated enhanced immune responses in Asian seabass. Fish receiving the multivalent Vibrio antigen-containing hydrogel vaccine (Hg-mVibrioAg) displayed elevated serum-specific IgM titers against all three Vibrio species, with significantly higher and more sustained antibody levels following a 7-day and 14-day vaccination regimen. Increased lysozyme and bactericidal activity further supported improved innate defense mechanisms. Subsequent pathogen challenge tests confirmed that vaccinated fish, particularly those following the 7-day and 14-day regimens, exhibited significantly higher survival rates and robust protection. Concurrently, immune-related gene expression in the head kidney, peripheral blood leukocytes, gills, and intestines was upregulated, indicating a broad immune activation associated with specific IgM responses. No significant alterations in blood biochemistry or tissue histology were observed, highlighting the vaccine's biocompatibility. Additionally, these findings underscore the potential of this multivalent oral hydrogel vaccine as a promising, safe, and effective prophylactic strategy against Vibrio infections in Asian seabass aquaculture.
Collapse
Affiliation(s)
- Anurak Uchuwittayakul
- Special Research Incubator Unit for Development and Application of Vaccine Delivery Systems for Aquatic Animals, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand.
| | - Kim D Thompson
- Moredun Research Institute, Pentlands Science Park, Penicuik, EH26 0PZ, United Kingdom.
| | - Pattanapong Thangsunan
- Division of Biochemistry and Biochemical Innovation, Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence for Innovation in Chemistry, Research Laboratory on Advanced Materials for Sensor and Biosensor Innovation, Material Science Research Center, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Jitraporn Phaksopa
- Department of Marine Science, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand.
| | - Wararut Buncharoen
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Kanokporn Saenphet
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Benchawan Kumwan
- Special Research Incubator Unit for Development and Application of Vaccine Delivery Systems for Aquatic Animals, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand.
| | - Pakapon Meachasompop
- Special Research Incubator Unit for Development and Application of Vaccine Delivery Systems for Aquatic Animals, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand.
| | - Supap Saenphet
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Nugroho Wiratama
- Special Research Incubator Unit for Development and Application of Vaccine Delivery Systems for Aquatic Animals, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand.
| | - Thanisorn Mahatnirunkul
- National Nanotechnology Center, National Science and Technology Development Agency, 111 Thailand Science Park, Phahonyothin Road, Khlong Luang, Pathum Thani, 12120, Thailand.
| | - Phunsin Kantha
- Special Research Incubator Unit for Development and Application of Vaccine Delivery Systems for Aquatic Animals, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand.
| | - Nuttee Suree
- Division of Biochemistry and Biochemical Innovation, Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Patcharapong Thangsunan
- Division of Biochemistry and Biochemical Innovation, Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand; Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Prapansak Srisapoome
- Special Research Incubator Unit for Development and Application of Vaccine Delivery Systems for Aquatic Animals, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand; Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok, 10900, Thailand.
| |
Collapse
|
4
|
Yourdkhani A, Esfandyari-Manesh M, Ranjbaran P, Amani M, Dinarvand R. Recent progress in topical and transdermal approaches for melanoma treatment. Drug Deliv Transl Res 2025; 15:1457-1495. [PMID: 39653958 DOI: 10.1007/s13346-024-01738-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2024] [Indexed: 04/04/2025]
Abstract
The global incidence of melanoma, the most lethal form of skin cancer, continues to escalate, emphasizing the urgent need for more effective therapeutic strategies. This review assesses the latest advancements in topical and transdermal drug delivery systems, positioning them as promising alternatives. These systems allow for the direct application of therapeutic agents to tumor sites, enhancing drug effectiveness, improving patient compliance, and reducing systemic toxicity. Specifically, innovations such as nanoparticles, microneedles, and vesicular systems are explored for their potential to optimize topical and localized drug delivery. By incorporating a graphical overview of these drug delivery vehicles, we visually underscore their roles in enhancing therapeutic outcomes across various treatment categories such as chemotherapy, immunotherapy, phototherapy, phytotherapy, and targeted therapy. This article critically evaluates recent breakthroughs, addresses the current challenges faced by researchers, and explores the future directions of topical and transdermal approaches in melanoma management. By presenting a summary of the latest research and predicting future trends, this review aims to inform ongoing developments and encourage further innovation in strategies for treating melanoma.
Collapse
Affiliation(s)
- Alaleh Yourdkhani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Esfandyari-Manesh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Paniz Ranjbaran
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdiyar Amani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Leicester School of Pharmacy, De Montfort University, Leicester, UK.
| |
Collapse
|
5
|
El-Adl K, Ghobashy MM, Ismail AFM, El-Morsy A, Shoman NA. Radiation-induced nanogel engineering based on pectin for pH-responsive rutin delivery for cancer treatment. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5249-5271. [PMID: 39540896 PMCID: PMC11985661 DOI: 10.1007/s00210-024-03573-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
This research investigates the formulation of a nanogel complex using pectin and poly(acrylic acid) (PAAc) to encapsulate rutin. The nanogel's pH-responsive behavior and its potential as a targeted drug delivery platform are investigated. The gamma irradiation-induced crosslinking mechanism is elucidated, highlighting its role in creating a stable three-dimensional network structure within the polymer matrix. Fourier transform infrared spectroscopy analysis sheds light on the molecular interactions within rutin and the nanogel-rutin complex. The pH-responsive behavior of the nanogel is explored, showcasing its ability to release rutin selectively in response to pH variations and displaying high physical and chemical stability. Transmission electron microscopy imaging provides visual insights into nanogel morphology and interactions. The cumulative drug content from the nanogel was 86.14 ± 2.61%. The pH-dependent release profile of the nanogel was examined, demonstrating selective rutin release in response to varying pH levels. Cytotoxicity studies were conducted against four human cancer cell lines-HepG2, A549, MCF-7, and HCT-116 showing significant reductions in IC50 values, indicating enhanced therapeutic efficacy. Additionally, molecular docking studies revealed strong binding interactions of rutin with VEGFR-2 and EGFRT790M. Our nanogel compound 5 significantly reduced the IC50 values for HepG2, A549, MCF-7, and HCT-116 cells by 58.19%, 81.29%, 71.81%, and 67.16%, respectively. Furthermore, it lowered the IC50 values for VEGFR-2 and EGFRT790M by 29.66% and 68.18%, respectively.
Collapse
Affiliation(s)
- Khaled El-Adl
- Chemistry Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt.
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11884, Cairo, Egypt.
| | - Mohamed M Ghobashy
- Radiation Research of Polymer Chemistry Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Nasr City, P.O. Cairo, Egypt
| | - Amel F M Ismail
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Ahmed El-Morsy
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11884, Cairo, Egypt
- Pharmaceutical Chemistry Department, College of Pharmacy, The Islamic University, Najaf, Iraq
| | - Nabil A Shoman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| |
Collapse
|
6
|
Chaudhari P, Ghate VM, Nampoothiri M, Lewis SA. Cyclosporine a Eluting Nano Drug Reservoir Film for the Management of Dry Eye Disease. AAPS PharmSciTech 2025; 26:109. [PMID: 40246763 DOI: 10.1208/s12249-025-03104-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/01/2025] [Indexed: 04/19/2025] Open
Abstract
Cyclosporine A (CsA) is widely used to treat dry eye disease (DED), and ocular morbidity is on the rise and is a growing concern globally. However, several drug and formulation challenges, such as poor drug solubility, short pre-corneal residence time, and poor patient compliance, have limited the ocular bioavailability of CsA to < 5%. A CsA cyclodextrin-based ternary complex loaded dissolvable nano drug reservoir films were developed to overcome these limitations and efficiently manage DED. Drug-loaded nano-reservoir films were fabricated via lithography using silicone and poly (dimethyl siloxane) (PDMS) molds. Different physicochemical characterizations were performed to confirm the formation of stable CsA-cyclodextrin-based ternary complexes. Formation of nanoreservoirs on the films was confirmed using SEM and AFM. Optimized CsA-complex-loaded nano-reservoir films were evaluated for in vitro drug release, ex vivo corneal permeation, and in vivo precorneal retention. Preclinical efficacy studies were performed to assess the efficacy of CsA-complex-loaded nano-reservoirs in an experimental dry-eye mouse model. Physicochemical characterization confirmed the formation of a stable complex and the improved solubility of CsA. In vitro release and ex vivo permeation studies indicated a controlled drug release and improved permeation, respectively. Furthermore, tear volume measurement and corneal damage assessment using slit-lamp imaging suggested decreased dry eye symptoms, significantly increasing tear volume in the drug-loaded nano-reservoir-treated group. Moreover, histopathological studies corroborated the tear volume and slit-lamp imaging results, with reduced inflammation and neovascularization. The poorly water-soluble drug with cyclodextrin complex incorporated nanoreservoir films presents a potential alternative for managing various ocular diseases.
Collapse
Affiliation(s)
- Pinal Chaudhari
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Madhavnagar, Manipal, Karnataka, 576104, India
| | - Vivek M Ghate
- Yenepoya Technology Incubator, Yenepoya (Deemed to Be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Madhavnagar, Manipal, Karnataka, India
| | - Shaila A Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Madhavnagar, Manipal, Karnataka, 576104, India.
| |
Collapse
|
7
|
Oh N, Hwang J, Kang MS, Yoo CY, Kwak M, Han DW. Versatile and Marvelous Potentials of Polydeoxyribonucleotide for Tissue Engineering and Regeneration. Biomater Res 2025; 29:0183. [PMID: 40231205 PMCID: PMC11994882 DOI: 10.34133/bmr.0183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 04/16/2025] Open
Abstract
Over the past decade, substantial focus has been placed on polydeoxyribonucleotide (PDRN) due to its promising pharmacological properties, making it a valuable candidate for tissue engineering applications. Accordingly, this paper aims to review and summarize the latest experimental research on PDRN in the context of tissue engineering and regeneration. The unique biochemical mechanisms of PDRN to promote cellular behavior and regeneration are summarized. We categorize commonly utilized PDRN-based tissue engineering fields as neuromuscular tissues, diabetic wound or skin, and bone regeneration. At the same time, we explore scaffold strategies for integrating PDRN into bioceramics, polymers, and cell/tissue-derived materials, along with its combination with photo/electromodulation techniques. Furthermore, we discuss potential opportunities and challenges in translating PDRN-based approaches into clinical practice. We expect future interdisciplinary research and clinical trials to evaluate the long-term efficacy and safety of PDRN while emphasizing standardization and quality control to ensure its consistency and effectiveness in regenerative applications.
Collapse
Affiliation(s)
- Nuri Oh
- Department of Chemistry and Biology,
Korea Science Academy of Korea Advanced Institute of Science and Technology, Busan 47162, Republic of Korea
| | - Juyoung Hwang
- Department of Chemistry,
Pukyong National University, Busan 48513, Republic of Korea
- Smart Gym-Based Translational Research Center for Active Senior’s Healthcare,
Pukyong National University, Busan 48513, Republic of Korea
- Ajou Energy Science Research Center,
Ajou University, Suwon 16499, Republic of Korea
| | - Moon Sung Kang
- Research Institute of Mechanical Technology,
Pusan National University, Busan 46241, Republic of Korea
| | - Chung-Yul Yoo
- Department of Energy Systems Research and Chemistry,
Ajou University, Suwon 16499, Republic of Korea
| | - Minseok Kwak
- Department of Chemistry,
Pukyong National University, Busan 48513, Republic of Korea
- Smart Gym-Based Translational Research Center for Active Senior’s Healthcare,
Pukyong National University, Busan 48513, Republic of Korea
- Industry 4.0 Convergence Bionics Engineering,
Pukyong National University, Busan 48513, Republic of Korea
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering,
Pusan National University, Busan 46241, Republic of Korea
- Institute of Nano-Bio Convergence,
Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
8
|
Kumwan B, Meachasompop P, Thompson KD, Thangsunan P, Buncharoen W, Thangsunan P, Srisapoome P, Uchuwittayakul A. Intestinal mucosal transcriptomic responses of Asian seabass (Lates calcarifer) vaccinated with an oral hydrogel-encapsulated multivalent Vibrio antigen following Vibrio spp. infection. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101512. [PMID: 40252617 DOI: 10.1016/j.cbd.2025.101512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/21/2025]
Abstract
This study examined the intestinal mucosal immune responses elicited by an oral hydrogel-encapsulated multivalent Vibrio vaccine in Asian seabass (Lates calcarifer) to protect against vibriosis caused by Vibrio harveyi, V. vulnificus, and Photobacterium damselae (formerly Vibrio damsela). Both 7-day and 14-day oral vaccination regimens effectively enhanced innate and adaptive immune responses while supporting gut recovery post-infection. Transcriptomic analyses of intestines from fish that received consecutive 7- and 14-day vaccination regimens, followed by co-infection with multistrain Vibrio spp., revealed significant upregulation of innate and specific immune markers at week 8 post-vaccination. These responses were further bolstered by a strong adaptive immune activation, characterized by T-cell and B-cell receptor signaling as well as antibody production. In addition, the vaccine also demonstrated cross-protective immunomodulatory effects, evidenced by elevated interferon-related pathways (e.g., IFNAR2 and IFN-induced proteins), suggesting its potential to protect against co-infecting pathogens, a critical advantage in aquaculture systems facing diverse pathogen pressures. Beyond immune activation, the involvement proteins of TGF-β family members, including BMP3 and BMP4, highlights the vaccine's role in tissue repair and remodeling. These responses likely mitigate epithelial damage and preserve gut barrier integrity post-infection, showcasing the dual benefits of immunoprotection and post-infection recovery. The findings highlight the oral hydrogel-encapsulated multivalent Vibrio vaccine's ability to enhance immunity against specific bacterial pathogens while offering broader immunomodulatory and tissue-repair benefits. Its cross-protective and recovery-supporting properties make it a promising solution for sustainable aquaculture practices, effectively addressing pathogen control and boosting host resilience.
Collapse
Affiliation(s)
- Benchawan Kumwan
- Special Research Incubator Unit for Development and Application of Vaccine Delivery Systems for Aquatic Animals, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand; Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand; Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand.
| | - Pakapon Meachasompop
- Special Research Incubator Unit for Development and Application of Vaccine Delivery Systems for Aquatic Animals, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand; Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand; Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand.
| | - Kim D Thompson
- Moredun Research Institute, Pentlands Science Park, Penicuik EH26 0PZ, United Kingdom.
| | - Pattanapong Thangsunan
- Division of Biochemistry and Biochemical Innovation, Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence for Innovation in Chemistry, and Research Laboratory on Advanced Materials for Sensor and Biosensor Innovation, Material Science Research Center, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Wararut Buncharoen
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Patcharapong Thangsunan
- Division of Biochemistry and Biochemical Innovation, Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand; Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Prapansak Srisapoome
- Special Research Incubator Unit for Development and Application of Vaccine Delivery Systems for Aquatic Animals, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand; Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand; Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand.
| | - Anurak Uchuwittayakul
- Special Research Incubator Unit for Development and Application of Vaccine Delivery Systems for Aquatic Animals, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand; Laboratory of Aquatic Animal Health Management, Department of Aquaculture, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand; Center of Excellence in Aquatic Animal Health Management, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand.
| |
Collapse
|
9
|
Xia Q, Zhou S, Zhou J, Zhao X, Saif MS, Wang J, Hasan M, Zhao M, Liu Q. Recent Advances and Challenges for Biological Materials in Micro/Nanocarrier Synthesis for Bone Infection and Tissue Engineering. ACS Biomater Sci Eng 2025; 11:1945-1969. [PMID: 40067283 DOI: 10.1021/acsbiomaterials.4c02118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Roughly 1.71 billion people worldwide suffer from large bone abnormalities, which are the primary cause of disability. Traditional bone grafting procedures have several drawbacks that impair their therapeutic efficacy and restrict their use in clinical settings. A great deal of work has been done to create fresh, more potent strategies. Under these circumstances, a crucial technique for the regeneration of major lesions has emerged: bone tissue engineering (BTE). BTE involves the use of biomaterials that can imitate the natural design of bone. To yet, no biological material has been able to fully meet the parameters of the perfect implantable material, even though several varieties have been created and investigated for bone regeneration. Against this backdrop, researchers have focused a great deal of interest over the past few years on the subject of nanotechnology and the use of nanostructures in regenerative medicine. The ability to create nanoengineered particles that can overcome the current constraints in regenerative strategies─such as decreased cell proliferation and differentiation, insufficient mechanical strength in biological materials, and insufficient production of extrinsic factors required for effective osteogenesis has revolutionized the field of bone and tissue engineering. The effects of nanoparticles on cell characteristics and the application of biological materials for bone regeneration are the main topics of our review, which summarizes the most recent in vitro and in vivo research on the application of nanotechnology in the context of BTE.
Collapse
Affiliation(s)
- Qipeng Xia
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
| | - Shuyan Zhou
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, PR China
| | - Jingya Zhou
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- College of Acupuncture and Massage, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, PR China
| | - Xia Zhao
- Faculty of Medicine, Dalian University of Technology, Dalian 116024, PR China
| | - Muhammad Saqib Saif
- Department of Biochemistry, Faculty of Chemical and Biological Sciences, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Jianping Wang
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
| | - Murtaza Hasan
- Department of Biotechnology, Faculty of Chemical and Biological Sciences, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Min Zhao
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
| | - Qiang Liu
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
- Faculty of Medicine, Dalian University of Technology, Dalian 116024, PR China
| |
Collapse
|
10
|
Esfanjani E, Ramezani A, Sanjarian F, Shafiei SS. Electrospun polycaprolactone coated with gum tragacanth containing layered double hydroxide/thymol nanohybrid for wound dressing application. Int J Biol Macromol 2025; 309:143049. [PMID: 40220807 DOI: 10.1016/j.ijbiomac.2025.143049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 04/06/2025] [Accepted: 04/08/2025] [Indexed: 04/14/2025]
Abstract
Incorporating a nanohybrid containing a natural antibacterial agent into nanofibrous scaffolds can synergistically affect wound healing. In this study, polycaprolactone (PCL) nanofibrous membranes were fabricated via electrospinning. Gum-tragacanth (GT)-containing thymol (Thy)-loaded layered double hydroxide (LDH) nanohybrids were coated on the nanofiber surface, which provided a sustained thymol release. The LDH/Thy nanohybrids were prepared using a co-precipitation method. LDH/Thy was added to the GT solution at different concentrations (0, 0.1, 1, and 10 % w/v, followed by dip-coating onto PCL nanofiber surfaces. The nanofibers were characterized using scanning electron microscopy (SEM), Fourier-transform infrared spectroscopy (FTIR), and energy-dispersive X-ray spectroscopy (EDX). These mats were evaluated for their hydrophilicity, in vitro drug release, mechanical and antibacterial properties, cytocompatibility, cell attachment, and in vivo wound healing performance. Results demonstrated that the inclusion of LDH/Thy significantly improved the hydrophilicity. The mechanical study showed that adding LDH (10 %) nanoparticles to PCL/GT nanofibers increased tensile strength from 0.27 to 1.74 MPa, respectively. In addition, the PCL/GT/LDH/Thy nanofibers exhibited superior antibacterial activity against Escherichia coli (Gram-negative), Staphylococcus aureus (Gram-positive), and methicillin-resistant Staphylococcus aureus (MRSA), which correlated with the sustained release of thymol (78 % over 110 h). Moreover, after five days, the nanofibers showed no cytotoxic activity against the L929 mouse fibroblast cell line and provided an appropriate microenvironment for cell attachment. The animal study results revealed that the average wound healing rate of PCL/GT/LDH/Thy nanofibers significantly increased compared to the control group on the 10th day post-treatment (from 80.33 % to 48.5 %, respectively), along with improved wound closure quality. The synergistic effect of PCL/GT/LDH/Thy could provide helpful information and implications for promoting their application in wound dressings.
Collapse
Affiliation(s)
- Elaheh Esfanjani
- Plant Bio-Product Group, Agricultural Biotechnology Institute, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Alireza Ramezani
- Stem Cell and Regenerative Medicine Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering & Biotechnology, Tehran, Iran
| | - Forough Sanjarian
- Plant Bio-Product Group, Agricultural Biotechnology Institute, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | - Seyedeh Sara Shafiei
- Stem Cell and Regenerative Medicine Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering & Biotechnology, Tehran, Iran.
| |
Collapse
|
11
|
Sathi Devi L, Gigliobianco MR, Gabrielli S, Agas D, Sabbieti MG, Morelli MB, Amantini C, Casadidio C, Di Martino P, Censi R. Localized Cancer Treatment Using Thiol-Ene Hydrogels for Dual Drug Delivery. Biomacromolecules 2025. [PMID: 40198273 DOI: 10.1021/acs.biomac.5c00387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Combinatorial cancer therapy benefits from injectable hydrogels for localized, controlled drug delivery. This study presents a thiol-ene conjugated hydrogel formed by cross-linking thiol-modified hyaluronic acid (HASH) with vinyl sulfone-modified β-cyclodextrin (CDVS). Four formulations (23Gel-16, 23Gel-33, 99Gel-16, 99Gel-33) were synthesized by varying HASH molecular weight (23 or 99 kDa) and CDVS modification (16% or 33%). Rheological analysis confirmed enhanced viscoelasticity with increasing molecular weight and modification (99Gel-33 > 99Gel-16 > 23Gel-33 > 23Gel-16). The system enabled combinatorial delivery of doxorubicin (DOX) and carvacrol (CRV), exhibiting tumor-responsive degradation and tunable release. DOX release accelerated under tumor-mimicking conditions (100% in 46 h vs 58.7% in PBS), while CRV showed an initial burst followed by sustained release. The hydrogel promoted mesenchymal stem cell proliferation and effectively inhibited triple-negative breast cancer cells. This injectable, tumor-responsive hydrogel system offers a promising platform for minimally invasive, personalized cancer therapy.
Collapse
Affiliation(s)
- Lakshmi Sathi Devi
- School of Pharmacy, ChIP Chemistry Interdisciplinary Project Research Centre, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, MC, Italy
| | - Maria Rosa Gigliobianco
- Department of Pharmacy, University of "G. D'Annunzio" Chieti and Pescara, Via dei Vestini 1, 66100 Chieti, CH, Italy
| | - Serena Gabrielli
- School of Science and Technology, ChIP Chemistry Interdisciplinary Project Research Centre, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, MC, Italy
| | - Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, MC, Italy
| | - Maria Giovanna Sabbieti
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, MC, Italy
| | - Maria Beatrice Morelli
- School of Pharmacy, Department of Experimental Medicine and Public Health, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, MC, Italy
| | - Consuelo Amantini
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, MC, Italy
| | - Cristina Casadidio
- School of Pharmacy, ChIP Chemistry Interdisciplinary Project Research Centre, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, MC, Italy
| | - Piera Di Martino
- Department of Pharmacy, University of "G. D'Annunzio" Chieti and Pescara, Via dei Vestini 1, 66100 Chieti, CH, Italy
| | - Roberta Censi
- School of Pharmacy, ChIP Chemistry Interdisciplinary Project Research Centre, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, MC, Italy
| |
Collapse
|
12
|
Said ZAS, Mohammed HS, Ibrahim S, Amer HH. Electrospun zinc oxide nanoscaffolds: a targeted and selective anticancer approach. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025; 36:689-710. [PMID: 39508689 DOI: 10.1080/09205063.2024.2422698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024]
Abstract
This study aims to prepare, characterize, and evaluate zinc oxide nanoscaffolds (ZnO NSs) as a potential anticancer drug that selectively targets malignant cells while remaining non-toxic to normal cells. Electrospun NSs were fabricated and loaded with varying concentrations of ZnO nanoparticles (NPs). The uniform morphology of the fabricated samples was confirmed through Field Emission Scanning Electron Microscope (FESEM) imaging. Elemental composition was investigated using Energy Dispersive X-ray spectroscopy (EDX), Fourier Transform Infrared (FTIR), and X-ray diffraction (XRD) analyses. Biocompatibility and cytotoxicity were assessed using the (3-(4.5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay) (MTT) assay and flow cytometry. The water uptake and degradation properties of the electrospun NSs were also examined. Furthermore, a cumulative release profile was generated to assess the release behavior of ZnO NSs. The prepared ZnO NSs demonstrated negligible toxicity toward normal human dermal cells. Conversely, the four used concentrations of ZnO NSs displayed substantial cytotoxicity and induced apoptosis in various cancer cell lines. The observed effects were concentration-dependent. Notably, ZnO NSs 8% exhibited the most significant reduction in cell viability against the MCF7 cell line. The findings from this study indicate the potential of ZnO NSs as an effective anticancer agent, with the ZnO NSs 8% demonstrating the most pronounced impact. This research introduces a novel application of electrospun zinc oxide nanoscaffolds, demonstrating their capacity for selective anticancer activity, particularly against breast carcinoma, while preserving normal cell viability. The study presents a significant advancement in the use of nanomaterial for targeted cancer therapy.
Collapse
Affiliation(s)
- Zeinab A S Said
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Haitham S Mohammed
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Sara Ibrahim
- Basic Medical Science Department, Faculty of Dentistry, Al Ryada University for Science and Technology, Menoufia, Egypt
| | - Hanan H Amer
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
13
|
Wu X, Hu Y, Sheng S, Yang H, Li Z, Han Q, Zhang Q, Su J. DNA-based hydrogels for bone regeneration: A promising tool for bone organoids. Mater Today Bio 2025; 31:101502. [PMID: 39911372 PMCID: PMC11795821 DOI: 10.1016/j.mtbio.2025.101502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 02/07/2025] Open
Abstract
DNA-based hydrogels stand out for bone regeneration due to their exceptional biocompatibility and programmability. These hydrogels facilitate the formation of spatial bone structures through bulk hydrogel fabricating, microsphere formatting, and 3D printing. Furthermore, the bone microenvironment can be finely tuned by leveraging the degradation products, nanostructure, targeting, and delivery capabilities inherent to DNA-based materials. In this review, we underscore the advantages of DNA-based hydrogels, detailing their composition, gelation techniques, and structure optimization. We then delineate three critical elements in the promotion of bone regeneration using DNA-based hydrogels: (i) osteogenesis driven by phosphate ions, plasmids, and oligodeoxynucleotides (ODNs) that enhance mineralization and promote gene and protein expression; (ii) vascularization facilitated by tetrahedral DNA nanostructures (TDNs) and aptamers, which boosts gene expression and targeted release; (iii) immunomodulation achieved through loaded factors, TDNs, and bound ions that stimulate macrophage polarization and exhibit antibacterial properties. With these advantages and properties, these DNA-based hydrogels can be used to construct bone organoids, providing an innovative tool for disease modeling and therapeutic applications in bone tissue engineering. Finally, we discuss the current challenges and future prospects, emphasizing the potential impacts and applications in regenerative medicine.
Collapse
Affiliation(s)
- Xiang Wu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Materials Science and Engineering, Shanghai University, Shanghai, 200444, China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Huijian Yang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Clinical Laboratory, Shanghai Zhongye Hospital, Shanghai, 201941, China
| | - Zuhao Li
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Qinglin Han
- Department of Orthopedics, The Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Qin Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Sanming Institute of Translational Medicine, Sanming, 365004, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
14
|
Field EH, Ratcliffe J, Johnson CJ, Binger KJ, Reynolds NP. Self-healing, 3D printed bioinks from self-assembled peptide and alginate hybrid hydrogels. BIOMATERIALS ADVANCES 2025; 169:214145. [PMID: 39675342 DOI: 10.1016/j.bioadv.2024.214145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/17/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
There is a pressing need for new cell-laden, printable, biomaterials that are rigid and highly biocompatible. These materials can mimic stiffer tissues such as cartilage, fibrotic tissue and cancer microenvironments, and thus have exciting applications in regenerative medicine, wound healing and cancer research. Self-assembled peptides (SAPs) functionalised with aromatic groups such as Fluorenyl-9-methoxycarbonyl (Fmoc) show promise as components of these biomaterials. However, the harsh basic conditions often used to solubilise SAPs leads to issues with toxicity and reproducibility. Here, we have designed a hybrid material comprised of self-assembled Fmoc-diphenylalanine (Fmoc-FF) assemblies dispersed throughout a sodium alginate matrix and investigated the influence of different organic solvents as peptide solubilising agents. Bioinks fabricated from peptides dissolved in 1,1,1,3,3,3-Hexafluoro-2-propanol (HFIP) showed improved biocompatibility compared to those made from Dimethyl Sulfoxide (DMSO) peptide stocks, due to the increased volatility and reduced surface tension of HFIP, allowing for more efficient expulsion from the system. Through optimisation of assembly and solvent conditions we can generate hybrid bioinks with stiffnesses up to 8 times greater than sodium alginate alone that remain highly printable, even when laden with high concentrations of cells. In addition, the shear-thinning nature of the self-assembled peptide assemblies gave the hybrid bioinks highly desirable self-healing capabilities. Our developed hybrid materials allow the bioprinting of materials previously considered too stiff to extrude without causing shear induced cytotoxicity with applications in tissue engineering and biosensing.
Collapse
Affiliation(s)
- Emily H Field
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Julian Ratcliffe
- La Trobe University Bioimaging platform, La Trobe University, Australia, Melbourne, Victoria 3086, Australia
| | - Chad J Johnson
- La Trobe University Bioimaging platform, La Trobe University, Australia, Melbourne, Victoria 3086, Australia
| | - Katrina J Binger
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria 3086, Australia; Centre for Cardiovascular Biology & Disease Research, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria 3086, Australia
| | - Nicholas P Reynolds
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria 3086, Australia; The Biomedical and Environmental Sensor Technology (BEST) Research Centre, Biosensors Program, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria 3086, Australia.
| |
Collapse
|
15
|
Ma DJ, Li TH, Yang SY, Yu JJ, Li ST, Yu Y, Liu Y, Zang J, Kong L, Li XT. Self-assembling Bletilla polysaccharide nanogels facilitate healing of acute and infected wounds via inflammation control and antibacterial activity. Int J Biol Macromol 2025; 299:140125. [PMID: 39842574 DOI: 10.1016/j.ijbiomac.2025.140125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/08/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
Wound healing is one of the fundamental problems faced by the medical profession. Thus, there is a need for the development of biomaterials that are safe, economically viable, possess anti-inflammatory and antibacterial characteristics, and enhance wound healing. In this study, we designed a nanomicelle of Bletilla striata polysaccharide (BSP) self-loaded with Azithromycin (AZI). The properties are improved by physically blending Carbomer 940 (CBM) with Gelatin (GEL) to serve as the hydrogel matrix. The preparation was made by combining the nanomicelle, used as the precursor solution, with the gel matrix. It was designed to treat wound infections and promote healing. Relevant experiments indicate its excellent biocompatibility. The hydrogel not only promotes cell migration, proliferation, angiogenesis, and collagen deposition associated with skin healing, but also regulates the polarization of macrophages from the M1 to M2 phenotype, as well as the expression of related factors. Additionally, in vitro experiments demonstrate its good antibacterial activity. In addition, we demonstrated the gel's anti-inflammatory, antibacterial, and pro-healing effects in acute wounds and methicillin-resistant Staphylococcus aureus (MRSA) wounds. Therefore, the nanomicellar gel enhances antibacterial activity and related immune regulation, offering a new direction in the treatment of acute and chronic wounds.
Collapse
Affiliation(s)
- De-Jin Ma
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Chinese Medicine Targeted Delivery Key laboratory, Shenyang 110847, China
| | - Tian-Hua Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Chinese Medicine Targeted Delivery Key laboratory, Shenyang 110847, China
| | - Su-Yu Yang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Chinese Medicine Targeted Delivery Key laboratory, Shenyang 110847, China
| | - Jun-Jie Yu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Chinese Medicine Targeted Delivery Key laboratory, Shenyang 110847, China
| | - Shu-Tong Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Chinese Medicine Targeted Delivery Key laboratory, Shenyang 110847, China
| | - Yang Yu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Chinese Medicine Targeted Delivery Key laboratory, Shenyang 110847, China
| | - Yang Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Chinese Medicine Targeted Delivery Key laboratory, Shenyang 110847, China
| | - Juan Zang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Chinese Medicine Targeted Delivery Key laboratory, Shenyang 110847, China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Chinese Medicine Targeted Delivery Key laboratory, Shenyang 110847, China.
| | - Xue-Tao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Chinese Medicine Targeted Delivery Key laboratory, Shenyang 110847, China.
| |
Collapse
|
16
|
Abbasi M, Aghamollaei H, Vaez A, Amani AM, Kamyab H, Chelliapan S, Jamalpour S, Zambrano-Dávila R. Bringing ophthalmology into the scientific world: Novel nanoparticle-based strategies for ocular drug delivery. Ocul Surf 2025; 37:140-172. [PMID: 40147816 DOI: 10.1016/j.jtos.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/03/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
The distinctive benefits and drawbacks of various drug delivery strategies to supply corneal tissue improvement for sense organs have been the attention of studies worldwide in recent decades. Static and dynamic barriers of ocular tissue prevent foreign chemicals from entering and inhibit the active absorption of therapeutic medicines. The distribution of different medications to ocular tissue is one of the most appealing and demanding tasks for investigators in pharmacology, biomaterials, and ophthalmology, and it is critical for cornea wound healing due to the controlled release rate and increased drug bioavailability. It should be mentioned that the transport of various types of medications into the different sections of the eye, particularly the cornea, is exceedingly challenging because of its distinctive structure and various barriers throughout the eye. Nanoparticles are being studied to improve medicine delivery strategies for ocular disease. Repetitive corneal drug delivery using biodegradable nanocarriers allows a medicine to remain in different parts of the cornea for extended periods of time and thus improve administration route effectiveness. In this review, we discussed eye anatomy, ocular delivery barriers, as well as the emphasis on the biodegradable nanomaterials ranging from organic nanostructures, such as nanomicelles, polymers, liposomes, niosomes, nanowafers, nanoemulsions, nanosuspensions, nanocrystals, cubosomes, olaminosomes, hybridized NPs, dendrimers, bilosomes, solid lipid NPs, nanostructured lipid carriers, and nanofiber to organic nanomaterials like silver, gold, and mesoporous silica nanoparticles. In addition, we describe the nanotechnology-based ophthalmic medications that are presently on the market or in clinical studies. Finally, drawing on current trends and therapeutic approaches, we discuss the challenges that innovative optical drug delivery systems confront and propose future research routes. We hope that this review will serve as a source of motivation and inspiration for developing innovative ophthalmic formulations.
Collapse
Affiliation(s)
- Milad Abbasi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hesam Kamyab
- Department of Biomaterials, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 600077, India; The KU-KIST Graduate School of Energy and Environment, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Republic of Korea; Universidad UTE, Quito, 170527, Ecuador.
| | - Shreeshivadasan Chelliapan
- Department of Smart Engineering and Advanced Technology, Faculty of Artificial Intelligence, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia.
| | - Sajad Jamalpour
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Renato Zambrano-Dávila
- Universidad UTE, Centro de Investigación en Salud Públicay Epidemiología Clínica (CISPEC), Quito, 170527, Ecuador
| |
Collapse
|
17
|
Lapmanee S, Bhubhanil S, Khongkow M, Namdee K, Yingmema W, Bhummaphan N, Wongchitrat P, Charoenphon N, Hutchison JA, Talodthaisong C, Kulchat S. Application of Gelatin/Vanillin/Fe 3+/AGP-AgNPs Hydrogels Promotes Wound Contraction, Enhances Dermal Growth Factor Expression, and Minimizes Skin Irritation. ACS OMEGA 2025; 10:10530-10545. [PMID: 40124024 PMCID: PMC11923657 DOI: 10.1021/acsomega.4c10648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025]
Abstract
This study further investigates the potential of gelatin-based hydrogel cross-linked with vanillin and ferric ion (GVF), combined with andrographolide (AGP) and silver nanoparticles (AgNPs), as an anti-infection biomaterial for wound dressing, aimed at exploring the mechanisms that attenuate inflammation, enhance wound healing rates, and address allergic skin irritation. AGP-AgNPs were evaluated for cytotoxicity in human adult epidermal keratinocytes (HEKa) and the murine macrophage cell line (RAW 264.7), as well as for nitric oxide (NO) production in response to lipopolysaccharide-induced inflammation in RAW 264.7 macrophage cells. Skin-wound specimens from male Wistar rats were histologically analyzed for epidermal thickness and inflammatory changes. The mRNA expression profiling of dermal growth factors was assessed using RT-qPCR, and skin irritation tests were conducted in female New Zealand rabbits. These AGP-AgNPs exhibited significantly lower toxicity in HEKa and no toxicity in RAW 264.7. Interestingly, AGP-AgNPs at specific concentrations produced NO in RAW 264.7 control cells but were more effective in reducing inflammatory NO levels in RAW 264.7 cells pretreated with lipopolysaccharides, suggesting that AGP-AgNP composites are safe and effectively diminish inflammation. Furthermore, a marked increase in epidermal thickness and a reduction in histological inflammatory cells at wound sites were observed in rats treated with AGP-AgNPs/GVF hydrogels over 21 days. Upregulation of dermal genes promoting wound healing, including collagen types I and III, epidermal growth factor, transforming growth factor-beta, fibronectin, and vascular endothelial growth factor, but not fibroblast growth factor, was observed in a time-dependent manner. These results suggest that the anti-inflammatory properties of GVF/AGP-AgNP hydrogels could promote epithelialization, enhance cellular proliferation, support extracellular matrix synthesis, and facilitate angiogenesis. Additionally, rabbit skin in contact with GVF/AGP-AgNP hydrogels consistently displayed reduced levels of erythema and edema, with no swelling, and a standardized scoring system yielded low primary dermal irritation indices for this hydrogel. These findings suggest that the novel GVF/AGP-AgNP hydrogels possess anti-inflammatory-like activity and can modulate dermal growth factors for wound healing. This leads to reduced dermal irritation, making the formulation potentially suitable for safe topical applications in skin and wound care. However, comprehensive human studies and clinical trials should be required in the future.
Collapse
Affiliation(s)
- Sarawut Lapmanee
- Chulabhorn
International College of Medicine, Thammasat
University, Pathumthani 10120, Thailand
- Department
of Basic Medical Sciences, Faculty of Medicine, Siam University, Bangkok 10160, Thailand
| | - Sakkarin Bhubhanil
- Department
of Basic Medical Sciences, Faculty of Medicine, Siam University, Bangkok 10160, Thailand
| | - Mattaka Khongkow
- National
Science and Technology Development Agency, National Nanotechnology Centre, Pathumthani 12120, Thailand
| | - Katawut Namdee
- National
Science and Technology Development Agency, National Nanotechnology Centre, Pathumthani 12120, Thailand
| | - Werayut Yingmema
- Laboratory
Animal Center, Thammasat University, Pathumthani 10120, Thailand
| | - Narumol Bhummaphan
- College
of Public Health Sciences, Chulalongkorn
University, Bangkok 10330, Thailand
| | - Prapimpun Wongchitrat
- Center
for
Research Innovation and Biomedical Informatics, Faculty of Medical
Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Natthawut Charoenphon
- Department
of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - James A. Hutchison
- School
of Chemistry and Centre of Excellence in Exciton Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Chanon Talodthaisong
- Department
of Chemistry and Center of Excellence for Innovation in Chemistry,
Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sirinan Kulchat
- Department
of Chemistry and Center of Excellence for Innovation in Chemistry,
Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
18
|
Boztepe T, Karp F, Cabrera S, Aleman J, Lamas DG, Huck-Iriart C, Islan GA, León IE. An oral delivery approach for riboflavin-targeted platinum(II)-loaded lipid nanoparticles into alginate-gelatin matrices against 2D and 3D colorectal carcinoma models. J Inorg Biochem 2025; 269:112900. [PMID: 40158497 DOI: 10.1016/j.jinorgbio.2025.112900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/01/2025] [Accepted: 03/16/2025] [Indexed: 04/02/2025]
Abstract
This study investigated the use of riboflavin-targeted Nanostructured Lipid Carriers (R-NLCs) to deliver a platinum-based anticancer drug [PtCl(8-O-quinolinate)(dmso)] (8-QO-Pt) to colorectal cancer cells. Three different R-8-QO-Pt-NLC formulations were prepared via hot-homogenization by ultrasonication method. The physicochemical characterizations of NLCs were analyzed by small- and wide-angle X-ray scattering (SAXS/WAXS) and fourier transformed infrared spectroscopy (FTIR). The cytotoxic effects and IC50 values of R-8-QO-Pt-NLC formulations were compared with those of the free 8-QO-Pt. Cellular uptake and apoptosis were evaluated towards HCT 116 cells in monolayer (2D). The liquid overlay technique was used to generate 3D multicellular tumor spheroids, MCTS. The anticancer and antimetastatic activities of the free 8-QO-Pt and R-8-QO-Pt-NLCs were determined in MCTS. The results revealed that R-8-QO-Pt-NLC exhibited greater cytotoxicity and lower IC50 values than free 8-QO-Pt in both 2D and 3D cell cultures. Furthermore, results showed that the volumes of the spheroids were reduced in response to increasing concentrations of R-8-QO-Pt-NLC, showing higher inhibition of cell migration in colorectal cancer spheroids at concentrations of 10.0, 15.0, and 25.0 μM than free 8-QO-Pt. To provide protection against gastric acid conditions, an additional drug delivery system based on alginate (Alg) and gelatin (Gel) beads for R-8-QO-Pt-NLC oral administration was developed. While free and R-NLC encapsulated 8-QO-Pt were practically inactivated at pH 1.2 and 37 °C, it was revealed that the Alg-Gel beads retain 5.7 times the initial activity of the R-8-QO-Pt-NLC. The findings of this research indicate that R-8-QO-Pt-NLC embedded in Alg-Gel beads are promising hydrogels for targeted colorectal delivery systems.
Collapse
Affiliation(s)
- Tugce Boztepe
- Centro de Investigación y Desarrollo en Fermentaciones Industriales (CINDEFI), Laboratorio de Nanobiomateriales, Departamento de Química, Facultad de Ciencias Exactas, CONICET-UNLP (CCT La Plata), La Plata, Buenos Aires, Argentina
| | - Federico Karp
- Centro de Investigación y Desarrollo en Fermentaciones Industriales (CINDEFI), Laboratorio de Nanobiomateriales, Departamento de Química, Facultad de Ciencias Exactas, CONICET-UNLP (CCT La Plata), La Plata, Buenos Aires, Argentina
| | - Silvia Cabrera
- Departamento de Química Inorgánica, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - José Aleman
- Departamento de Química Orgánica, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Diego G Lamas
- Instituto de Tecnologías Emergentes y Ciencias Aplicadas (ITECA), UNSAM-CONICET, Escuela de Ciencia y Tecnología (ECyT), Laboratorio de Cristalografía Aplicada (LCA), Campus Miguelete, (1650) San Martín, Buenos Aires, Argentina
| | - Cristián Huck-Iriart
- Instituto de Tecnologías Emergentes y Ciencias Aplicadas (ITECA), UNSAM-CONICET, Escuela de Ciencia y Tecnología (ECyT), Laboratorio de Cristalografía Aplicada (LCA), Campus Miguelete, (1650) San Martín, Buenos Aires, Argentina; ALBA Synchrotron Light Source, Carrer de la Llum 2-26, Cerdanyola del Vallès, 08290 Barcelona, Spain
| | - Germán A Islan
- Centro de Investigación y Desarrollo en Fermentaciones Industriales (CINDEFI), Laboratorio de Nanobiomateriales, Departamento de Química, Facultad de Ciencias Exactas, CONICET-UNLP (CCT La Plata), La Plata, Buenos Aires, Argentina; Children's Hospital, University Medical Center of the Johannes-Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Ignacio E León
- CEQUINOR (UNLP, CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata. Blvd., 120 N° 1465, La Plata 1900, Argentina; Cátedra de Fisiopatología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata 1900, Argentina.
| |
Collapse
|
19
|
El-Tanani M, Satyam SM, Rabbani SA, El-Tanani Y, Aljabali AAA, Al Faouri I, Rehman A. Revolutionizing Drug Delivery: The Impact of Advanced Materials Science and Technology on Precision Medicine. Pharmaceutics 2025; 17:375. [PMID: 40143038 PMCID: PMC11944361 DOI: 10.3390/pharmaceutics17030375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Recent progress in material science has led to the development of new drug delivery systems that go beyond the conventional approaches and offer greater accuracy and convenience in the application of therapeutic agents. This review discusses the evolutionary role of nanocarriers, hydrogels, and bioresponsive polymers that offer enhanced drug release, target accuracy, and bioavailability. Oncology, chronic disease management, and vaccine delivery are some of the applications explored in this paper to show how these materials improve the therapeutic results, counteract multidrug resistance, and allow for sustained and localized treatments. The review also discusses the translational barriers of bringing advanced materials into the clinical setting, which include issues of biocompatibility, scalability, and regulatory approval. Methods to overcome these challenges include surface modifications to reduce immunogenicity, scalable production methods such as microfluidics, and the harmonization of regulatory systems. In addition, the convergence of artificial intelligence (AI) and machine learning (ML) is opening new frontiers in material science and personalized medicine. These technologies allow for predictive modeling and real-time adjustments to optimize drug delivery to the needs of individual patients. The use of advanced materials can also be applied to rare and underserved diseases; thus, new strategies in gene therapy, orphan drugs development, and global vaccine distribution may offer new hopes for millions of patients.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates
| | - Shakta Mani Satyam
- Department of Pharmacology, RAK College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates
| | - Syed Arman Rabbani
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates
| | | | - Alaa A. A. Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan;
| | - Ibrahim Al Faouri
- RAK College of Nursing, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates
| | - Abdul Rehman
- Department of Pathology, RAK College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates;
| |
Collapse
|
20
|
Narayana S, Gowda BHJ, Hani U, Ahmed MG, Asiri ZA, Paul K. Smart Poly(N-isopropylacrylamide)-Based Hydrogels: A Tour D'horizon of Biomedical Applications. Gels 2025; 11:207. [PMID: 40136912 PMCID: PMC11942434 DOI: 10.3390/gels11030207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Hydrogels are innovative materials characterized by a water-swollen, crosslinked polymeric network capable of retaining substantial amounts of water while maintaining structural integrity. Their unique ability to swell or contract in response to environmental stimuli makes them integral to biomedical applications, including drug delivery, tissue engineering, and wound healing. Among these, "smart" hydrogels, sensitive to stimuli such as pH, temperature, and light, showcase reversible transitions between liquid and semi-solid states. Thermoresponsive hydrogels, exemplified by poly(N-isopropylacrylamide) (PNIPAM), are particularly notable for their sensitivity to temperature changes, transitioning near their lower critical solution temperature (LCST) of approximately 32 °C in water. Structurally, PNIPAM-based hydrogels (PNIPAM-HYDs) are chemically versatile, allowing for modifications that enhance biocompatibility and functional adaptability. These properties enable their application in diverse therapeutic areas such as cancer therapy, phototherapy, wound healing, and tissue engineering. In this review, the unique properties and behavior of smart PNIPAM are explored, with an emphasis on diverse synthesis methods and a brief note on biocompatibility. Furthermore, the structural and functional modifications of PNIPAM-HYDs are detailed, along with their biomedical applications in cancer therapy, phototherapy, wound healing, tissue engineering, skin conditions, ocular diseases, etc. Various delivery routes and patents highlighting therapeutic advancements are also examined. Finally, the future prospects of PNIPAM-HYDs remain promising, with ongoing research focused on enhancing their stability, responsiveness, and clinical applicability. Their continued development is expected to revolutionize biomedical technologies, paving the way for more efficient and targeted therapeutic solutions.
Collapse
Affiliation(s)
- Soumya Narayana
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India;
| | - B. H. Jaswanth Gowda
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India;
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia; (U.H.); (Z.A.A.)
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India;
| | - Zahrah Ali Asiri
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia; (U.H.); (Z.A.A.)
| | - Karthika Paul
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India;
| |
Collapse
|
21
|
Wei PS, Chou PY, Hsu HY, Chen M, Chen YJ, Tsai TH, Wen BY, Sheu MT, Chuang KH, Lin HL. Nonshrinkable Thermosensitive Hydrogels Combined with Bispecific Anti-PSMA/CD3 T-Cell Engager for Effective Against Tumors in Mice Model. Int J Nanomedicine 2025; 20:3083-3111. [PMID: 40098725 PMCID: PMC11911822 DOI: 10.2147/ijn.s496746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose CD3-based Bispecific T-cell engagers (BiTEs) are effective for solid tumors due to their tumor specificity and tissue penetration, but they face challenges like short half-lives and narrow therapeutic windows. Innovative delivery systems, like thermosensitive hydrogels, show the potential to enhance stability, sustained release, and therapeutic efficacy. Methods We developed PEGylated PLGA (PEG-PLGA) thermosensitive hydrogels with a nonshrinkable property (nsTPPgels) for effective controlled release and loaded them with bispecific anti-prostate surface membrane antigen (PSMA) Fab /anti-CD3 scFv T-cell engager (BiPTE) to form in situ drug deposits with a sustained-release profile after subcutaneous injection. Each group of hydrogels was first tested for differences in properties through rheological and in vitro drug release profiles. Meanwhile, in vivo pharmacokinetics, anti-tumor efficacy studies, and T-cell tracking studies were conducted to analyze the advantages of nsTPPgels included D2gel and DTgels. Results The cytotoxicity of BiPTE against PSMA-overexpressing tumor cells and the drug release functionality of nsTPPgels were validated in vitro. Rheological studies showed that both D2gel and DTgels remained in solution below 27 °C for easy injection and solidified at physiological temperatures to form localized depots for sustained BiPTE release. All nsTPPgels demonstrated a 5-day in vitro sustained release, prolonged elimination half-life, steady plasma BiPTE levels, and extended mean residence time. In an LNCaP-xenograft mouse model, tumor growth inhibition rates for BiPTE/DTgel-2, BiPTE/DTgel-2S, and BiPTE/D2gel were 74.3%, 96.1%, and 113.1%, respectively, compared to 35.6% for intravenous and 46% for subcutaneous BiPTE administration. Furthermore, all nsTPPgels effectively achieved T-cell recruitment to lymph nodes and tumor sites in tracking studies. Conclusion In conclusion, we developed relatively convenient injectable thermosensitive D2gel with a desirable gelation temperature window, which have the potential to be used for antibody drug delivery in several biomedical applications.
Collapse
Affiliation(s)
- Pu-Sheng Wei
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Po-Yu Chou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Hao-Yi Hsu
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Michael Chen
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jou Chen
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Tung-Han Tsai
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Bang-Yu Wen
- Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Ming-Thau Sheu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Program in Clinical Genomics and Proteomics, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Ph.D Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan
- The Ph.D. Program of Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hong-Liang Lin
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
22
|
Jeleriu RM, Hajaj RK, Trăilă IA, Zaharie M, Puiu M. Theoretical Models and Simulations of Gene Delivery with Polyurethane: The Importance of Polyurethane as a Vector in Personalized Therapy. Biomedicines 2025; 13:692. [PMID: 40149668 PMCID: PMC11939958 DOI: 10.3390/biomedicines13030692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Advancements in personalized medicine have revolutionized drug delivery, enabling tailored treatments based on genetic and molecular profiles. Non-viral vectors, such as polyurethane (PU)-based systems, offer promising alternatives for gene therapy. This study develops mathematical models to analyze PU degradation, DNA/RNA release kinetics, and cellular interactions, optimizing their application in personalized therapy. Methods: This theoretical study utilized mathematical modeling and numerical simulations to analyze PU-based gene delivery, focusing on diffusion, degradation, and cellular uptake. Implemented in Python 3.9, it employed differential equation solvers and adsorption/internalization models to predict vector behavior and optimize delivery efficiency. Results: This study demonstrated that PU degrades in biological environments following first-order kinetics, ensuring a controlled and predictable release of genetic material. The Higuchi diffusion model confirmed a gradual, sustained DNA/RNA release, essential for efficient gene delivery. Simulations of PU adsorption onto cellular membranes using the Langmuir model showed saturation-dependent binding, while the endocytosis model revealed a balance between uptake and degradation. These findings highlight PU's potential as a versatile gene delivery vector, offering controlled biodegradability, optimized release profiles, and effective cellular interaction. Conclusions: Our results confirm that PU-based vectors enable controlled biodegradability, sustained DNA/RNA release, and efficient cellular uptake. Mathematical modeling provides a framework for improving PU's properties, enhancing transport efficiency and therapeutic potential in personalized medicine and gene therapy applications.
Collapse
Affiliation(s)
- Roxana Maria Jeleriu
- Ph.D. School, Faculty of Medicine, Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2 E. Murgu, Sq., 300041 Timisoara, Romania; (R.M.J.); (R.-K.H.); (M.P.)
| | - Roxana-Karin Hajaj
- Ph.D. School, Faculty of Medicine, Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2 E. Murgu, Sq., 300041 Timisoara, Romania; (R.M.J.); (R.-K.H.); (M.P.)
- Department of Pediatric Surgery and Orthopedics, “Louis Țurcanu” Emergency Clinical Hospital for Children, 300011 Timisoara, Romania
| | - Iuliana-Anamaria Trăilă
- Department of Pathology, ‘Pius Brinzeu’ Emergency County Clinical Hospital, 300723 Timisoara, Romania
| | - Mihaela Zaharie
- Department XII Obstetrics-Gynecology, ‘Victor Babeş’ University of Medicine and Pharmacy of Timișoara, 300041 Timisoara, Romania;
- Neonatology-Premature Unit, “Louis Țurcanu” Emergency Clinical Hospital for Children, 300011 Timisoara, Romania
| | - Maria Puiu
- Ph.D. School, Faculty of Medicine, Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2 E. Murgu, Sq., 300041 Timisoara, Romania; (R.M.J.); (R.-K.H.); (M.P.)
- Institute for Research and Development in Genomics, 020021 București, Romania
| |
Collapse
|
23
|
Garcia PS, Antunes BSL, Komatsu D, de Alencar Hausen M, Dicko C, de Rezende Duek EA. Mechanical and rheological properties of Pluronic F127 based-hydrogels loaded with chitosan grafted with hyaluronic acid and propolis, focused to atopic dermatitis treatment. Int J Biol Macromol 2025; 307:141942. [PMID: 40081723 DOI: 10.1016/j.ijbiomac.2025.141942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 02/10/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Hydrogels for biomedical applications have been widely studied once they are able to enhance the wound-healing process, as well as facilitate the controlled release and loading of drugs. In this context, Pluronic F127 (PF127) has a major role as it was shown to have exceptional versatility, once it holds unique gelation properties, as it is thermoreversible and is liquid in lower temperatures, and changes to gel in higher temperatures. Moreover, the gelation behavior of PF127 is influenced by its concentration and can be further modulated by incorporating different compounds, including drugs. In this study, the mechanical and rheological properties of Pluronic F127-based hydrogels were evaluated to be further used as a treatment for atopic dermatitis (AD). To this purpose, it was conducted Fourier transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM), rheology, adhesivity, cohesion, and compression assays, allowed us to understand the effects of different concentrations of PF127 and how it behaves after incorporating compounds (chitosan grafted with hyaluronic acid (Ch/HA), and propolis), aiming to identify optimal combinations for the hydrogel formulation. To provide a reliable formulation, the study first selected the most suitable concentration of PF127, in the sequence it incorporated Ch/HA and lastly, propolis was added. The findings have provided valuable insights regarding the selection of the formulations, correlating the mechanical, rheological, and morphological data. It is expected that the formulation achieved is able to further be applied to the wounded skin, providing a low-cost and effective alternative to the treatment of AD.
Collapse
Affiliation(s)
- Pâmela Soto Garcia
- Biomaterials Laboratory, Medical and Health Sciences Faculty, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, SP 18060-030, Brazil; Pure and Applied Biochemistry, Department of Chemistry, Lund University, Lund SE-221 00, Sweden.
| | - Bianca Sabino Leocádio Antunes
- Biomaterials Laboratory, Medical and Health Sciences Faculty, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, SP 18060-030, Brazil; Post-graduation Program in Materials Sciences (PPGCM), Federal University of São Carlos (UFSCar), Sorocaba 18052-780, SP, Brazil
| | - Daniel Komatsu
- Biomaterials Laboratory, Medical and Health Sciences Faculty, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, SP 18060-030, Brazil
| | - Moema de Alencar Hausen
- Biomaterials Laboratory, Medical and Health Sciences Faculty, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, SP 18060-030, Brazil; Post-graduation Program of Biomaterials and Regenerative Medicine (PPGBMR), Surgery Department, PUC-SP, Sorocaba, SP 18060-030, Brazil
| | - Cedric Dicko
- Pure and Applied Biochemistry, Department of Chemistry, Lund University, Lund SE-221 00, Sweden
| | - Eliana Aparecida de Rezende Duek
- Biomaterials Laboratory, Medical and Health Sciences Faculty, Pontifical Catholic University of São Paulo (PUC-SP), Sorocaba, SP 18060-030, Brazil; Post-graduation Program in Materials Sciences (PPGCM), Federal University of São Carlos (UFSCar), Sorocaba 18052-780, SP, Brazil; Post-graduation Program of Biomaterials and Regenerative Medicine (PPGBMR), Surgery Department, PUC-SP, Sorocaba, SP 18060-030, Brazil
| |
Collapse
|
24
|
Filho D, Guerrero M, Castro R, Rafael D, Andrade F, Marican A, Valdes O, Vargas E, Valenzuela E, Mora C, Durán-Lara EF. Influence of agarose in semi-IPN hydrogels for sustained Polymyxin B release. Colloids Surf B Biointerfaces 2025; 247:114431. [PMID: 39673897 DOI: 10.1016/j.colsurfb.2024.114431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/06/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Hydrogels (HGs) are 3-D polymeric networks with high water content, making them appropriate for biomedical applications such as drug delivery systems. This study examines the impact of agarose in semi-interpenetrating polymer networks (Semi-IPNs) based on poly(acrylic acid) (p(AA)), N, N' Methylenebis(acrylamide) (MBA) and agarose (AGA) on the sustained release of Polymyxin B (PolB). Agarose incorporation improved the mechanical strength, swelling behavior and drug retention capacity of the HG. We synthesized the Semi-IPN HGs via free radical polymerization and characterized their structural and thermal properties using Fourier-transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM), thermogravimetric analysis (TGA), and differential scanning calorimetry (DSC). The features of swelling under physiological conditions were carried out. Additionally, we conducted release kinetics using the three prepared HGs, each of which had a distinct amount of AGA. The findings demonstrated that the Semi-IPN HGs with greater AGA concentrations had drug release profiles that were slower and more sustained, making them perfect for long-term therapeutic uses. We also tested the PolB-loaded HGs' antimicrobial efficacy against Pseudomonas aeruginosa, and they showed sustained antibacterial activity. Using NIH-3T3 fibroblast cells, we verified the HGs' biocompatibility, demonstrating their appropriateness for use in biomedicine. According to these findings, agarose modified Semi-IPN HGs may find application in long-term medication delivery systems that aid in the treatment of infections and promote wound healing.
Collapse
Affiliation(s)
- David Filho
- Laboratory of Bio & Nano Materials, Drug Delivery and Controlled Release, Department of Microbiology, Faculty of Health Sciences, University of Talca, Talca, Chile; PhD Program in Science, R&D Bioactive Products Department, Chemistry Institute of Natural Resources, University of Talca, Talca, Chile
| | - Marcelo Guerrero
- Laboratory of Bio & Nano Materials, Drug Delivery and Controlled Release, Department of Microbiology, Faculty of Health Sciences, University of Talca, Talca, Chile; PhD Program in Science, R&D Bioactive Products Department, Chemistry Institute of Natural Resources, University of Talca, Talca, Chile
| | - Ricardo Castro
- Multidisciplinary Agroindustry Research Laboratory, Instituto de Ciencias Aplicadas, Facultad de Construcción y Medio Ambiente, Universidad Autónoma de Chile, Talca, Chile
| | - Diana Rafael
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, Barcelona 08035, Spain; Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, Madrid, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain
| | - Fernanda Andrade
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, Barcelona 08035, Spain; Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, Madrid, Spain; Department of Pharmacy and Pharmaceutical Technology and Physicochemistry, Faculty of Pharmacy and Food Sciences, School of Pharmacy, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, Barcelona 08028, Spain
| | - Adolfo Marican
- Institute of Chemistry of Natural Research, University of Talca, Talca 3460000, Chile
| | - Oscar Valdes
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile
| | - Esteban Vargas
- Center for the Development of Nanoscience and Nanotechnology. Santiago. Chile
| | - Elisa Valenzuela
- School of Biochemistry, Institute of Biological Sciences, University of Talca, Talca, Chile
| | - Claudia Mora
- Laboratory of Bio & Nano Materials, Drug Delivery and Controlled Release, Department of Microbiology, Faculty of Health Sciences, University of Talca, Talca, Chile
| | - Esteban F Durán-Lara
- Laboratory of Bio & Nano Materials, Drug Delivery and Controlled Release, Department of Microbiology, Faculty of Health Sciences, University of Talca, Talca, Chile.
| |
Collapse
|
25
|
Shaikh S, Chary PS, Mehra NK. Nano-interventions for dengue: a comprehensive review of control, detection and treatment strategies. Inflammopharmacology 2025; 33:979-1011. [PMID: 39976669 DOI: 10.1007/s10787-025-01655-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/12/2025] [Indexed: 03/19/2025]
Abstract
Dengue, a formidable life-threatening malady, currently exerts a profound impact upon the Western Pacific and Southeast-Asian developing and underdeveloped nations. The intricacies inherent in addressing dengue are manifold, requiring a concerted effort not only towards vector control but also the implementation of efficacious host treatments to forestall the progression of the disease into severe manifestations, such as hemorrhage and shock. The only vaccine available for dengue in the market is DENGVAXIA, with several other vaccine candidates which are currently in the clinical developmental stages. However, DENGVAXIA, owing to incidences of adverse events in among children, was withdrawn in Philippines. This warrants the development of new safer vaccine candidates. The existent control strategies, regrettably, demonstrate inadequacy in effectively mitigating the rampant dissemination of this ailment. Moreover, the diagnostic and therapeutic modalities exhibit potential for refinement, specifically through precision diagnostics and tailored therapeutic interventions, to enhance the precision and efficacy of dengue management. This comprehensive review endeavors to provide an in-depth elucidation of the utilization of nanotechnology-based approaches synergistically integrated with conventional methodologies in the overarching domains of dengue control, diagnosis, and treatment.
Collapse
Affiliation(s)
- Samia Shaikh
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Ministry of Chemical and Family Welfare, Hyderabad, Telangana, 500 037, India
| | - Padakanti Sandeep Chary
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Ministry of Chemical and Family Welfare, Hyderabad, Telangana, 500 037, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Ministry of Chemical and Family Welfare, Hyderabad, Telangana, 500 037, India.
| |
Collapse
|
26
|
Akdoğan D, Peksel A. Immobilization and characterization of β-galactosidase from Aspergillus oryzae in PVA-CMC hydrogel. Int J Biol Macromol 2025; 297:139816. [PMID: 39809391 DOI: 10.1016/j.ijbiomac.2025.139816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Creating new formulations of immobilized enzymes has been a major focus of modern biotechnology. In this study, the industrially significant β-galactosidase was immobilized by being trapped in a polyvinyl alcohol and carboxymethyl cellulose (PVA-CMC) gel. The immobilized enzyme was optimized and characterized, and the results were compared with those obtained using free enzymes. The data show that 40 °C to 50 °C is the ideal temperature range for the enzyme after immobilization. The activity rose, the Vmax value increased from 1.94 U/mg to 6.01 U/mg, and the Km value fell from 4.86 mM to 3.35 mM at pH 5, the optimal pH. β-galactosidases immobilized on PVA-CMC gels exhibited 70 % activity at the end of the fifth week and 50 % activity at the end of the eighth week, depending on the storage stability of the immobilized enzyme. After three reuses, the initial activity of the enzymes decreased, yet the thermal stability of the immobilized enzyme remained superior to that of the free form, retaining 82 % of its initial activity. Thus, it might be claimed that immobilization amplifies the enzyme's catalytic impact. Consequently, it has been discovered that immobilized β-galactosidase exhibits stronger enzymatic characteristics than free β-galactosidase, making it potentially more useful in industrial operations.
Collapse
Affiliation(s)
- Doruk Akdoğan
- Department of Chemistry, Faculty of Arts and Science, Yildiz Technical University, Turkey; Department of Pharmacy Services, Health Services Vocational School, Istanbul Nisantasi University, Turkey.
| | - Ayşegül Peksel
- Department of Chemistry, Faculty of Arts and Science, Yildiz Technical University, Turkey.
| |
Collapse
|
27
|
Enoch K, Somasundaram AA. Unravelling the rheological and multifunctionality of Justicia adhatoda-impregnated carboxymethyl cellulose hydrogels for drug delivery systems. Int J Biol Macromol 2025; 306:141419. [PMID: 40024406 DOI: 10.1016/j.ijbiomac.2025.141419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/06/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
Herbal medicine harnesses the therapeutic properties of natural compounds, providing a sustainable approach to healthcare. Due to its limited aqueous solubility, therapeutic potential remains largely unexplored. The enhanced therapeutic potential of the herbal drug entails meticulous formulation and an effective drug delivery matrix. In this study, we formulated a hydrogel system based on Carboxymethylcellulose to deliver the herbal drug, J. adhatoda. The phytochemicals in the J. adhatoda extract involved in the crosslinking of the hydrogel via hydrogen bonding resulted from the interaction with the hydroxyl and carboxyl group of the polymeric chains, confirmed by the FT-IR studies. Rheological studies demonstrated improved mechanical properties with increasing extract concentration (20 μg to 100 μg), with yield stress increasing from 58.83 Pa to 121.5 Pa. The hydrogels exhibited significant antimicrobial activity, reducing biofilm formation against S. aureus, E. faecalis, E. coli, and K. pneumoniae by 71 %, 68 %, 61 %, and 57 %. Antioxidant activity improved with extract concentration, increasing from 29 % to 74 %. The optimal anticancer activity of the hydrogels showed IC50 values of 37.4 μL for MCF-7 and 65 μL for A431 cell lines. These findings demonstrate that J. adhatoda-impregnated CMC hydrogels offer enhanced bioactivity and mechanical strength, positioning them as promising candidates for biomedical applications.
Collapse
Affiliation(s)
- Karolinekersin Enoch
- Soft Matter Laboratory, Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Anbumozhi Angayarkanni Somasundaram
- Soft Matter Laboratory, Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
28
|
Yeo G, Kim J. Antibacterial Chitosan-Based Double-Network Hydrogel Patch Loaded with Antioxidant Ceria Nanoparticles and Betamethasone to Treat Psoriasis. Biomacromolecules 2025. [PMID: 40014754 DOI: 10.1021/acs.biomac.4c01442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Psoriasis is a chronic inflammatory skin disorder characterized by keratinocyte hyperproliferation, oxidative stress, and immune dysregulation. In this study, we developed a multifunctional, double-network hydrogel, composed of chitosan and poly(acrylic acid), embedded with cerium oxide nanoparticles (CeNPs) and betamethasone. The hydrogel harnesses the redox-catalytic properties of CeNPs to scavenge reactive oxygen species (ROS) while ensuring sustained betamethasone release for antibacterial and anti-inflammatory effects. Its mechanical stability and high water retention make it suitable for long-term skin application. In vitro, the hydrogel enhanced keratinocyte viability under oxidative stress and showed significant antibacterial activity against Escherichia coli. In a psoriasis-induced mouse model, the hydrogel significantly reduced epidermal hyperplasia, suppressed keratinocyte proliferation, and lowered inflammatory cytokine levels. The combination of antioxidant, antibacterial, and anti-inflammatory properties suggests that this hydrogel offers a promising therapeutic strategy for psoriasis, addressing both oxidative stress and inflammation for effective treatment.
Collapse
Affiliation(s)
- Gaeun Yeo
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of MetaBioHealth, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| |
Collapse
|
29
|
Kwiatek J, Paczkowska-Walendowska M, Rył A, Karpiński TM, Miklaszewski A, Swora-Cwynar E, Leśna M, Cielecka-Piontek J. Azithromycin-Loaded Nanoparticles Incorporated in Chitosan-Based Soft Hydrogels: A Novel Approach for Dental Drug Delivery. Pharmaceutics 2025; 17:304. [PMID: 40142968 PMCID: PMC11945840 DOI: 10.3390/pharmaceutics17030304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Azithromycin (AZC), a BCS class II/IV antibiotic with broad-spectrum antimicrobial activity, has poor water solubility, limiting its formulation potential. This study aimed to develop and optimize AZC-based soft hydrogels for the first time for improved solubility, local controlled drug release, and local dental applications. Methods: AZC nanoparticles (based on polyvinylpyrrolidone) were synthesized via electrospinning enhanced solubility 40-fold. These were incorporated into chitosan (CS) hydrogels with varying concentrations and degrees of deacetylation (DDA), optimized using a factorial design. Hydrogels were characterized for drug release, mucoadhesion, antioxidant, anti-inflammatory, and antimicrobial properties, with Principal Component Analysis (PCA) assessing correlations. Results: Soft hydrogels with 3% CS and 80% DDA achieved sustained drug release (62.9-94.7% over 48 h), strong mucoadhesion, and enhanced biological activity. Higher CS and DDA improved antioxidant and anti-inflammatory effects due to increased free amino groups. Antimicrobial tests showed efficacy against Streptococcus mutans and Staphylococcus aureus. PCA revealed an inverse correlation between AZC release and mucoadhesion and positive correlations between release and anti-inflammatory activity. Conclusions: AZC-based soft hydrogels significantly improved solubility, controlled release, and biological activity, showing strong potential for dental drug delivery. Further clinical validation and optimization are recommended.
Collapse
Affiliation(s)
- Jakub Kwiatek
- Kwiatek Dental Clinic Sp. z o.o., Kordeckiego 22, 60-144 Poznan, Poland; (J.K.); (M.L.)
| | - Magdalena Paczkowska-Walendowska
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
- Science-Bridge Sp. z o.o., Chociszewskiego 24/8, 60-258 Poznan, Poland
| | - Anna Rył
- Department of Chemical and Molecular Engineering, Lodz University of Technology, Wolczanska 213, 93-005 Lodz, Poland;
| | - Tomasz M. Karpiński
- Department of Medical Microbiology, Medical Faculty, Poznan University of Medical Sciences, Rokietnicka 10, 60-806 Poznan, Poland;
| | - Andrzej Miklaszewski
- Faculty of Materials Engineering and Technical Physics, Institute of Materials Science and Engineering, Poznan University of Technology, 60-965 Poznan, Poland;
| | - Ewelina Swora-Cwynar
- Department of Pharmacology and Phytochemistry, Institute of Natural Fibres and Medicinal Plants—National Research Institute, Wojska Polskiego 71b, 60-630 Poznan, Poland;
| | - Marta Leśna
- Kwiatek Dental Clinic Sp. z o.o., Kordeckiego 22, 60-144 Poznan, Poland; (J.K.); (M.L.)
| | - Judyta Cielecka-Piontek
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
- Science-Bridge Sp. z o.o., Chociszewskiego 24/8, 60-258 Poznan, Poland
| |
Collapse
|
30
|
Waugaman SD, Dementyev M, Abbasi GharehTapeh E, Lopez CG, Mathers RT, Hickey RJ. Nanoparticle Loading in Swollen Polymer Gels: An Unexpected Thermodynamic Twist. NANO LETTERS 2025; 25:3323-3329. [PMID: 39960322 DOI: 10.1021/acs.nanolett.4c06501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Tailoring polymer gel functionality by loading small molecules and nanoparticles is critical for drug delivery and tissue regeneration. Typically, solute loading in gels correlates with the degree of solvent swelling, which is controlled by the cross-link density and polymer/solvent interactions. However, the general assumption that the degree of swelling is the primary factor for nanoparticle loading is incorrect. Here, we demonstrate that the pairwise interactions between the polymer, solvent, and solute dictate the solute loading in gels. We performed gel loading studies of ligand-stabilized gold nanoparticles using different solvents, polymer network hydrophobicity, and cross-link densities, and found that nanoparticle distribution between polymer and solvent correlate with calculated thermodynamic partition coefficients. Despite previous assumptions that the maximum nanoparticle loading occurs at the highest degree of gel swelling, we reveal that nanoparticles preferentially load into gels with lower solvent swelling if ligand/polymer interactions are more favorable than ligand/solvent interactions.
Collapse
Affiliation(s)
- Seth D Waugaman
- Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Mykyta Dementyev
- Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Elmira Abbasi GharehTapeh
- Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Carlos G Lopez
- Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Robert T Mathers
- Chemistry, The Pennsylvania State University, New Kensington, Pennsylvania 15068, United States
| | - Robert J Hickey
- Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Materials Research Institute, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
31
|
Moni SS, Moshi JM, Matou-Nasri S, Alotaibi S, Hawsawi YM, Elmobark ME, Hakami AMS, Jeraiby MA, Sulayli AA, Moafa HN. Advances in Materials Science for Precision Melanoma Therapy: Nanotechnology-Enhanced Drug Delivery Systems. Pharmaceutics 2025; 17:296. [PMID: 40142960 PMCID: PMC11945159 DOI: 10.3390/pharmaceutics17030296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Melanoma, a highly aggressive form of skin cancer, poses a major therapeutic challenge due to its metastatic potential, resistance to conventional therapies, and the complexity of the tumor microenvironment (TME). Materials science and nanotechnology advances have led to using nanocarriers such as liposomes, dendrimers, polymeric nanoparticles, and metallic nanoparticles as transformative solutions for precision melanoma therapy. This review summarizes findings from Web of Science, PubMed, EMBASE, Scopus, and Google Scholar and highlights the role of nanotechnology in overcoming melanoma treatment barriers. Nanoparticles facilitate passive and active targeting through mechanisms such as the enhanced permeability and retention (EPR) effect and functionalization with tumor-specific ligands, thereby improving the accuracy of drug delivery and reducing systemic toxicity. Stimuli-responsive systems and multi-stage targeting further improve therapeutic precision and overcome challenges such as poor tumor penetration and drug resistance. Emerging therapeutic platforms combine diagnostic imaging with therapeutic delivery, paving the way for personalized medicine. However, there are still issues with scalability, biocompatibility, and regulatory compliance. This comprehensive review highlights the potential of integrating nanotechnology with advances in genetics and proteomics, scalable, and patient-specific therapies. These interdisciplinary innovations promise to redefine the treatment of melanoma and provide safer, more effective, and more accessible treatments. Continued research is essential to bridge the gap between evidence-based scientific advances and clinical applications.
Collapse
Affiliation(s)
- Sivakumar S. Moni
- College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
- Health Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| | - Jobran M. Moshi
- Department of Medical Laboratory Technology, College of Nursing and Health Science, Jazan University, Jazan 45142, Saudi Arabia
- Health Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| | - Sabine Matou-Nasri
- Blood and Cancer Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs, Riyadh 11481, Saudi Arabia;
- Biosciences Department, Faculty of the School for Systems Biology, George Mason University, Manassas, VA 22030, USA
| | - Shmoukh Alotaibi
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah 23433, Saudi Arabia; (S.A.); (Y.M.H.)
| | - Yousef M. Hawsawi
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah 23433, Saudi Arabia; (S.A.); (Y.M.H.)
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | | | | | - Mohammed A. Jeraiby
- Department of Basic Medical Science, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia;
| | - Ahmed A. Sulayli
- Laboratory Department, Prince Mohammed bin Nasser Hospital, Jazan Health Cluster, Jazan 82734, Saudi Arabia;
| | - Hassan N. Moafa
- Department of Public Health, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Quality and Patients Safety, Jazan University Hospital, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
32
|
Shajari G, Erfan-Niya H, Fathi M, Amiryaghoubi N. Thiolated gellan gum/polyethylene glycol diacrylate hydrogels containing timolol maleate-loaded chitosan nanoparticles for ophthalmic delivery. Biomed Mater 2025; 20:025029. [PMID: 39938216 DOI: 10.1088/1748-605x/adb555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/12/2025] [Indexed: 02/14/2025]
Abstract
The combination of hydrogels with nanoformulations can significantly enhance the delivery and effectiveness of drugs in ophthalmic drug delivery systems. In the current study, the polyethylene glycol diacrylate (PEGDA)/thiolated gellan gum (GGSH) hydrogels based on GGSH and PEGDA were prepared via thiol-ene reaction using Irgacure 2959 as a photoinitiator. To this end, the modification of GG was achieved by esterification of the hydroxyl groups of GG with the carboxyl group of mercaptopropionic acid with a free thiol amount of 95.5 μmol g-1. To provide sustained release, chitosan nanoparticles (CSNPs) containing timolol maleate (TM) with 56.4% entrapment efficiency were synthesized by the desolvation method and encapsulated in the developed hydrogel. The values of zeta potential and particle size of CSNPs were +26.0 mV and 182.4 nm, respectively. The physico/chemical properties of the hydrogels were investigated via texture analyzer, FT-IR, XRD, and SEM. Thein vitrodegradation, swelling behavior, rheological assessments, cell viability testing, and porosity determination were evaluated. With the increase in PEGDA concentration, the mechanical properties were increased. While the rate of swelling, degradation, and drug release were decreased. Thein vitrobiocompatibility of hydrogels was confirmed using the MTT test. According to anex vivostudy, ocular drug delivery using the obtained transparent hydrogels is promising due to improved drug permeation and sustained release of TM via CSNPs.
Collapse
Affiliation(s)
- Golnaz Shajari
- Faculty of Chemical and Petroleum Engineering, University of Tabriz, 51666-16471 Tabriz, Iran
| | - Hamid Erfan-Niya
- Faculty of Chemical and Petroleum Engineering, University of Tabriz, 51666-16471 Tabriz, Iran
| | - Marziyeh Fathi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazanin Amiryaghoubi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
33
|
Halim N, Nallusamy N, Lakshminarayanan R, Ramakrishna S, Vigneswari S. Electrospinning in Drug Delivery: Progress and Future Outlook. Macromol Rapid Commun 2025:e2400903. [PMID: 39973618 DOI: 10.1002/marc.202400903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/30/2025] [Indexed: 02/21/2025]
Abstract
There is intense research during the past few decades to design and fabricate drug delivery systems using the electrospinning system. Electrospinning is an efficient technique to produce nanofiber materials with different dimensions and morphologies by adjusting the processing parameters. Electrospinning is becoming an innovative technology that promotes the pursuit and maintenance of human health. Herein, the review discusses the contribution of electrospinning technology in drug delivery systems, summarising the modification of the various electrospinning system configurations and the effects of the process parameters on fibers, their application in drug delivery including carrier materials, loaded drugs and their release mechanisms and illustrates their various medical applications. Finally, this review discusses the challenges, bottlenecks, and development prospects of electrospinning technology in the field of drug delivery in terms of scaling up for clinical use and exploring potential solutions to pave the way to establish electrospinning for future drug delivery systems.
Collapse
Affiliation(s)
- Nurfitrah Halim
- Institute of Climate Adaptation and Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, 21030, Malaysia
| | - Nithiskanna Nallusamy
- Institute of Climate Adaptation and Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, 21030, Malaysia
| | - Rajamani Lakshminarayanan
- Ocular Infections and Antimicrobials Research Group, Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore, 169856, Singapore
| | - Seeram Ramakrishna
- Center for Nanotechnology and Sustainability, National University of Singapore, Singapore, 119260, Singapore
| | - Sevakumaran Vigneswari
- Institute of Climate Adaptation and Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, 21030, Malaysia
- Ocular Infections and Antimicrobials Research Group, Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore, 169856, Singapore
| |
Collapse
|
34
|
Xu Z, Liu J, Hu H, Ma J, Yang H, Chen J, Xu H, Hu H, Luo H, Chen G. Recent applications of stimulus-responsive smart hydrogels for osteoarthritis therapy. Front Bioeng Biotechnol 2025; 13:1539566. [PMID: 40035023 PMCID: PMC11872905 DOI: 10.3389/fbioe.2025.1539566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Osteoarthritis is one of the most common degenerative joint diseases, which seriously affects the life of middle-aged and elderly people. Traditional treatments such as surgical treatment and systemic medication, often do not achieve the expected or optimal results, which leads to severe trauma and a variety of side effects. Therefore, there is an urgent need to develop novel therapeutic options to overcome these problems. Hydrogels are widely used in biomedical tissue repairing as a platform for loading drugs, proteins and stem cells. In recent years, smart-responsive hydrogels have achieved excellent results as novel drug delivery systems in the treatment of osteoarthritis. This review focuses on the recent advances of endogenous stimuli (including enzymes, pH, reactive oxygen species and temperature, etc.) responsive hydrogels and exogenous stimuli (including light, shear, ultrasound and magnetism, etc.) responsive hydrogels in osteoarthritis treatment. Finally, the current limitations of application and future prospects of smart responsive hydrogels are summarized.
Collapse
Affiliation(s)
- Zhuoming Xu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jintao Liu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Hanyin Hu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jun Ma
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Haiyang Yang
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jiayi Chen
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Hongwei Xu
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Haodong Hu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Huanhuan Luo
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Gang Chen
- Department of Orthopaedics, Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
35
|
Das D, Chingakham N, Sarma M, Basu S, Bhaladhare S. Cellulose-based biodegradable superabsorbent hydrogel: A sustainable approach for water conservation and plant growth in agriculture. Int J Biol Macromol 2025; 305:141176. [PMID: 39971065 DOI: 10.1016/j.ijbiomac.2025.141176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 02/08/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025]
Abstract
Innovative deficit irrigation technologies are imperative to overcome challenges posed to crop growth/yield and agriculture sustainability due to water scarcity in arid and semiarid regions. In the current study, superabsorbent biodegradable hydrogel based on carboxymethylcellulose sodium salt (NaCMC) and hydroxy ethyl cellulose (HEC) has been developed using citric acid (CA) as a crosslinker. The hydrogel has demonstrated excellent water absorption, retention, and release properties. Moreover, hydrogel (2 %) modified soil (HMS) has depicted increased porosity (57 %) and reduced soil density (1.06 g/cm3), compared to unmodified soil (UMS) with porosity of 53 % and density of 1.16 g/cm3, as well as, the water use efficiency of the plants (25.25 %-45.52 % over UMS) grown in HMS, which is vital for comprehending soil properties and their impact on water retention and aeration. The plant growth study in HMS concerning critical growth parameters such as germination rate, Seedling Vigour Index (SVI), Root Shoot Ratio (RSR), crop growth ratio (CGR), and chlorophyll content of three plants, i.e., one summer-grown plant-cucumber and two winter-grown plants- tomato and mung bean, has manifested promising results. Decisive parameters such as seedling viability (4.51 %), plant growth rate (3.77 %), and photosynthetic ability (16.74 %) were increased for chosen plants grown in HMS as compared to UMS. Improved growth parameters and photosynthetic ability of plants in HMS have suggested ameliorated nutrient and water absorption rates, increased resource utilization, and improved response to extrinsic resource allotment caused by hydrogel modification. Statistical analyses supported the trends in plant growth. Thus, hydrogel modification of the soil can effectively mitigate water use by retaining moisture efficiently and positively facilitating growth.
Collapse
Affiliation(s)
- Dipankar Das
- Department of Chemical and Polymer Engineering, Tripura University, Suryamaninagar 799022, India
| | - Nancy Chingakham
- Department of Botany, Tripura University, Suryamaninagar 799022, India
| | - Mousumi Sarma
- Department of Botany, Tripura University, Suryamaninagar 799022, India
| | - Surochita Basu
- Department of Botany, Tripura University, Suryamaninagar 799022, India.
| | - Sachin Bhaladhare
- Department of Chemical and Polymer Engineering, Tripura University, Suryamaninagar 799022, India.
| |
Collapse
|
36
|
Sahiner M, Suner SS, Demirci S, Ayyala RS, Sahiner N. Toxicity Evaluation of Sulfobetainized Branched Polyethyleneimine via Antibacterial and Biocompatibility Assays. TOXICS 2025; 13:136. [PMID: 39997951 PMCID: PMC11860753 DOI: 10.3390/toxics13020136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Branched polyethyleneimine (PEI), possessing different types of amines-e.g., primary, secondary, and tertiary-in the polymer chains are well known for their antibacterial properties and DNA condensing ability, affording substantial advantages in many biomedical uses, including gene therapy. However, because of PEI's toxicity, depending on the molecular weight, its widespread biomedical use is hindered. Therefore, in this study, PEIs with different molecular weights-i.e., 600, 1200, and 1800 g/mol-were modified with 1,3-propane sultone, undergoing a sulfobetainization reaction in a single step to attain a zwitterionic structure: sulfobetainized PEI (b-PEI). The sulfobetainization reaction was carried out twice to increase the zwitterionic repeating unit on PEI chains. The increasing number of SO3- groups on the PEI chains was confirmed by the increased peak intensities around 1160 and 1035 cm-1 on the FT-IR spectrum, which are assigned to symmetric and asymmetric S=O peaks. The elemental analysis results for first- and second- betainization PEIs, abbreviated as b1-PEI and b2-PEI, respectively, were revealedthe increased wt% of S confirming the successful multiple-sulfobetainization of the PEI chains. Thermal stability analyses of PEIs and their corresponding multiple-sulfobetainized forms showed that multiple-sulfobetainization reactions increased the thermal stability of bare PEI chains. PEIs with lower molecular weights exhibited more antimicrobial properties. As PEI is sulfobetainated, its antimicrobial properties can be further adjusted via sulfobetainization (once or twice), or by adjusting the corresponding solution pH, or by protonating them with different acids with different counter anions. The cell toxicity of PEI on L929 fibroblast cells was slightly increased by increasing the molecular weight of the PEI, but all forms of sulfobetainized PEIs were found to be safe (no toxicity), even at 1000 µg/mL concentrations.
Collapse
Affiliation(s)
- Mehtap Sahiner
- Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, Terzioglu Campus, 17100 Canakkale, Turkey;
- Department of Chemical, Biological, and Materials Engineering, University of South Florida, Tampa, FL 33620, USA
| | - Selin S. Suner
- Department of Chemistry, Faculty of Sciences, Canakkale Onsekiz Mart University, Terzioglu Campus, 17100 Canakkale, Turkey;
| | - Sahin Demirci
- Department of Food Engineering, Faculty of Engineering, Istanbul Aydin University, Florya Halit Aydin Campus, 34153 Istanbul, Turkey;
| | - Ramesh S. Ayyala
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd, MDC21, Tampa, FL 33612, USA
| | - Nurettin Sahiner
- Department of Chemistry, Faculty of Sciences, Canakkale Onsekiz Mart University, Terzioglu Campus, 17100 Canakkale, Turkey;
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd, MDC21, Tampa, FL 33612, USA
- Department of Bioengineering, U.A. Whitaker College of Engineering, Florida Gulf Coast University, 10501 FGCU Boulevard South, Fort Myers, FL 33965, USA
| |
Collapse
|
37
|
Lee HM, Ryu H. Bioresorbable Materials for Wound Management. Biomimetics (Basel) 2025; 10:108. [PMID: 39997131 PMCID: PMC11852938 DOI: 10.3390/biomimetics10020108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Chronic wounds pose a significant healthcare challenge due to their risk of severe complications, necessitating effective management strategies. Bioresorbable materials have emerged as an innovative solution, offering advantages such as eliminating the need for secondary surgical removal, reducing infection risks, and enabling time-delayed drug delivery. This review examines recent advancements in bioresorbable wound healing materials, focusing on a systematic review of bioresorbable materials, systems incorporating electrical stimulation, and drug delivery technologies to accelerate tissue repair. The discussion encompasses the fundamental principles of bioresorbable materials, including their resorption mechanisms and key properties, alongside preclinical applications that demonstrate their practical potential. Critical challenges impeding widespread adoption are addressed, and prospects for integrating these cutting-edge systems into clinical practice are outlined. Together, these insights underscore the promise of bioresorbable materials in revolutionizing chronic wound care.
Collapse
Affiliation(s)
- Hye-Min Lee
- Department of Advanced Materials Engineering, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea
| | - Hanjun Ryu
- Department of Advanced Materials Engineering, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea
- Department of Intelligence Energy and Industry, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
38
|
Jeong JO, Kim M, Kim S, Lee KK, Choi H. Advanced Hydrogel Systems for Local Anesthetic Delivery: Toward Prolonged and Targeted Pain Relief. Gels 2025; 11:131. [PMID: 39996674 PMCID: PMC11854925 DOI: 10.3390/gels11020131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Local anesthetics (LAs) have been indispensable in clinical pain management, yet their limitations, such as short duration of action and systemic toxicity, necessitate improved delivery strategies. Hydrogels, with their biocompatibility, tunable properties, and ability to modulate drug release, have been extensively explored as platforms for enhancing LA efficacy and safety. This narrative review explores the historical development of LAs, their physicochemical properties, and clinical applications, providing a foundation for understanding the integration of hydrogels in anesthetic delivery. Advances in thermoresponsive, stimuli-responsive, and multifunctional hydrogels have demonstrated significant potential in prolonging analgesia and reducing systemic exposure in preclinical studies, while early clinical findings highlight the feasibility of thermoresponsive hydrogel formulations. Despite these advancements, challenges such as burst release, mechanical instability, and regulatory considerations remain critical barriers to clinical translation. Emerging innovations, including nanocomposite hydrogels, biofunctionalized matrices, and smart materials, offer potential solutions to these limitations. Future research should focus on optimizing hydrogel formulations, expanding clinical validation, and integrating advanced fabrication technologies such as 3D printing and artificial intelligence-driven design to enhance personalized pain management. By bridging materials science and anesthetic pharmacology, this review provides a comprehensive perspective on current trends and future directions in hydrogel-based LA delivery systems.
Collapse
Affiliation(s)
- Jin-Oh Jeong
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (J.-O.J.); (K.K.L.)
| | - Minjoo Kim
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Seonwook Kim
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA;
| | - Kyung Kwan Lee
- Wake Forest Institute for Regenerative Medicine (WFIRM), Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; (J.-O.J.); (K.K.L.)
| | - Hoon Choi
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| |
Collapse
|
39
|
Delgado-Pujol EJ, Martínez G, Casado-Jurado D, Vázquez J, León-Barberena J, Rodríguez-Lucena D, Torres Y, Alcudia A, Begines B. Hydrogels and Nanogels: Pioneering the Future of Advanced Drug Delivery Systems. Pharmaceutics 2025; 17:215. [PMID: 40006582 PMCID: PMC11859140 DOI: 10.3390/pharmaceutics17020215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/22/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Conventional drug delivery approaches, including tablets and capsules, often suffer from reduced therapeutic effectiveness, largely attributed to inadequate bioavailability and difficulties in ensuring patient adherence. These challenges have driven the development of advanced drug delivery systems (DDS), with hydrogels and especially nanogels emerging as promising materials to overcome these limitations. Hydrogels, with their biocompatibility, high water content, and stimuli-responsive properties, provide controlled and targeted drug release. This review explores the evolution, properties, and classifications of hydrogels versus nanogels and their applications in drug delivery, detailing synthesis methods, including chemical crosslinking, physical self-assembly, and advanced techniques such as microfluidics and 3D printing. It also examines drug-loading mechanisms (e.g., physical encapsulation and electrostatic interactions) and release strategies (e.g., diffusion, stimuli-responsive, and enzyme-triggered). These gels demonstrate significant advantages in addressing the limitations of traditional DDS, offering improved drug stability, sustained release, and high specificity. Their adaptability extends to various routes of administration, including topical, oral, and injectable forms, while emerging nanogels further enhance therapeutic targeting through nanoscale precision and stimuli responsiveness. Although hydrogels and nanogels have transformative potential in personalized medicine, challenges remain in scalable manufacturing, regulatory approval, and targeted delivery. Future strategies include integrating biosensors for real-time monitoring, developing dual-stimuli-responsive systems, and optimizing surface functionalization for specificity. These advancements aim to establish hydrogels and nanogels as cornerstones of next-generation therapeutic solutions, revolutionizing drug delivery, and paving the way for innovative, patient-centered treatments.
Collapse
Affiliation(s)
- Ernesto J. Delgado-Pujol
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (E.J.D.-P.); (G.M.); (D.C.-J.); (D.R.-L.); (A.A.)
- Departamento de Ingeniería y Ciencia de los Materiales y del Transporte, Escuela Politécnica Superior, Universidad de Sevilla, 41011 Sevilla, Spain;
| | - Guillermo Martínez
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (E.J.D.-P.); (G.M.); (D.C.-J.); (D.R.-L.); (A.A.)
| | - David Casado-Jurado
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (E.J.D.-P.); (G.M.); (D.C.-J.); (D.R.-L.); (A.A.)
- Departamento de Citología e Histología Normal y Patológica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Juan Vázquez
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, 41012 Sevilla, Spain;
| | - Jesús León-Barberena
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, 41012 Sevilla, Spain;
| | - David Rodríguez-Lucena
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (E.J.D.-P.); (G.M.); (D.C.-J.); (D.R.-L.); (A.A.)
| | - Yadir Torres
- Departamento de Ingeniería y Ciencia de los Materiales y del Transporte, Escuela Politécnica Superior, Universidad de Sevilla, 41011 Sevilla, Spain;
| | - Ana Alcudia
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (E.J.D.-P.); (G.M.); (D.C.-J.); (D.R.-L.); (A.A.)
| | - Belén Begines
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (E.J.D.-P.); (G.M.); (D.C.-J.); (D.R.-L.); (A.A.)
| |
Collapse
|
40
|
Alfinaikh RS, Alamry KA, Hussein MA. Sustainable and biocompatible hybrid materials-based sulfated polysaccharides for biomedical applications: a review. RSC Adv 2025; 15:4708-4767. [PMID: 39958265 PMCID: PMC11826415 DOI: 10.1039/d4ra07277d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/26/2025] [Indexed: 02/18/2025] Open
Abstract
Sustainable biomaterials that are both efficient and environmentally friendly are the subject of research and development efforts among scientists and academics from a variety of contemporary scientific disciplines. Due to their significant involvement in several physiological and pathological processes, sulfated polysaccharides (SPs) have garnered growing interest across various application domains, including biomedicine. Nevertheless, mechanical and thermal stability are issues for unmodified polysaccharide materials. Interactions between polymers, such as the mixing of biopolymers with synthetic or biopolymers through chemical interaction or grafting into the main chain structure of raw materials to enhance their therapeutic effects, are essential to meet the high standards of biomedical features. Another way to improve the mechanical and thermal properties is to graft appropriate fillers onto the polysaccharide backbone. The characteristics of polysaccharide bio-nanocomposites in comparison to more traditional polymers have attracted a lot of interest. With an emphasis on anti-inflammatory, anticancer, antiviral, immunoregulatory, and anticoagulant properties, this review delves into the most recent biological uses of sulfated polysaccharides. As well as thoroughly outlining the factors that impact the biological properties, such as the extraction process, molecular weight (Mw), the degree of sulfation, distribution/position, modification procedures, and the filler size, etc., this review aims to: (1) provide a systematic and critical overview of the cutting-edge research on SPs and hybrid sulfated polysaccharide bio-nanocomposites; (2) identify the key factors, mechanisms, methods, and challenges impacting SPs bio-nanocomposites; (3) elucidate the current and potential biomedical applications, advantages, manufacturing challenges, and opportunities associated with SPs bio-nanocomposites; (4) offer insights into future research directions by suggesting improvements for bio-nanocomposites, including novel materials, and advanced processing techniques.
Collapse
Affiliation(s)
- Reem S Alfinaikh
- Chemistry Department, Faculty of Science, King Abdulaziz University P.O. Box 80203 Jeddah 21589 Saudi Arabia
| | - Khalid A Alamry
- Chemistry Department, Faculty of Science, King Abdulaziz University P.O. Box 80203 Jeddah 21589 Saudi Arabia
| | - Mahmoud A Hussein
- Chemistry Department, Faculty of Science, King Abdulaziz University P.O. Box 80203 Jeddah 21589 Saudi Arabia
- Chemistry Department, Faculty of Science, Assiut University Assiut 71516 Egypt
| |
Collapse
|
41
|
Zherdeva VV, Likhov AR, Saidvaliev UA, Fixler D, Demin D, Volodina VN, Apukhtina UA, Pawar S, Atuar B, Tuchin VV. Enhanced Fluorescence Imaging of Implants Based on Polyester Copolymers in Combination With MRI. JOURNAL OF BIOPHOTONICS 2025:e202400147. [PMID: 39899887 DOI: 10.1002/jbio.202400147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 01/09/2025] [Accepted: 01/12/2025] [Indexed: 02/05/2025]
Abstract
Nowadays, many biodegradable materials are offered for biomedical applications, but there are only a few in vivo methods for their detection and monitoring. In this work, implants based on biodegradable polyester copolymers were labeled with indocyanine green (ICG) for fluorescence imaging in combination with tissue optical clearing (TOC) and magnetic resonance imaging (MRI). The results include in vitro degradation modeling followed by in vivo imaging of copolymer samples that were subcutaneously implanted in BALB/c mice. TOC with 70% glycerol has been demonstrated to significantly improve sample visualization. The TOC efficiency parameter Q demonstrated the variability of effects correlating with the timing of follow-up in the postimplantation period. It has been shown that nonhealing wounds, peri-implantation inflammation, or fibrosis, confirmed by MRI, affect the effectiveness of TOC in the range from Q = -30% to 70%.
Collapse
Affiliation(s)
- Victoria V Zherdeva
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Astemir R Likhov
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Ulugbek A Saidvaliev
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, Israel
| | - Dror Fixler
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, Israel
| | - Dmitry Demin
- MIREA - Russian Technological University, Moscow, Russia
| | - Veronika N Volodina
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Uliana A Apukhtina
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Shweta Pawar
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, Israel
| | - Bar Atuar
- Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, Israel
| | - Valery V Tuchin
- Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation
- Institute of Physics and Science Medical Center, Saratov State University, Saratov, Russian Federation
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, Tomsk, Russian Federation
| |
Collapse
|
42
|
Abid Mustafa M, Rashid Hussain H, Akbar Khan J, Ahmad N, Bashir S, Asad M, Saeed Shah H, Ali Khan A, Malik A, Fatima S, Mehmood Yousaf A, Nazir I. Development and In Vitro Characterization of Azadirachta Indica Gum Grafted Polyacrylamide Based pH-Sensitive Hydrogels to Improve the Bioavailability of Lansoprazole. Chem Biodivers 2025; 22:e202401434. [PMID: 39404191 DOI: 10.1002/cbdv.202401434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/14/2024] [Indexed: 11/14/2024]
Abstract
The present study intended to develop a pH-responsive hydrogel based on Neem gum (Ng) to improve Lansoprazole (LSP) oral bioavailability. Azadirachta Indica seed extract was used to obtain Ng. pH-responsive hydrogel formulations (F1-F9) were prepared using different Ng ratios, Acrylamide (AAm), and methylene-bis-acrylamide (MBA). The formulated hydrogels were characterized through FTIR, thermal analysis, swelling ratio, SEM, sol-gel ratios, In-Vitro drug release, and cytotoxicity analysis. Azadirachta Indica was extracted to produce a powder containing 21.5 % Ng. Prepared hydrogels showed maximum swelling at pH 7.4, whereas the swelling at an acidic pH was insignificant. LSP-loaded hydrogel demonstrated a regulated release of LSP for up to 24 h and indicated a Super Case II transport release mechanism. During the cytotoxic evaluation, the delivery system showed minimal cytotoxicity towards normal cells, while percent cytotoxicity was carried out for a longer duration (up to 96 h). The present study revealed Azadirachta indica gum-based pH-responsive hydrogel as a promising technique for precisely delivering LSP.
Collapse
Affiliation(s)
- Muhammad Abid Mustafa
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Lahore University of Biological & Applied Sciences, Lahore, 54000, Pakistan
| | | | - Jawad Akbar Khan
- Center of Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Wahringerstrasse 13a, A-1090, Vienna, Austria
| | - Nadeem Ahmad
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan
| | - Sajid Bashir
- Lords College of Pharmacy, Lahore, 54000, Pakistan
| | | | - Hamid Saeed Shah
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, 54000, Pakistan
| | - Azmat Ali Khan
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Abdul Malik
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Sabiha Fatima
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh, 12371, Saudi Arabia
| | - Abid Mehmood Yousaf
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan
| | - Imran Nazir
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan
| |
Collapse
|
43
|
Heremans J, Ballet S, Martin C. The versatility of peptide hydrogels: From self-assembly to drug delivery applications. J Pept Sci 2025; 31:e3662. [PMID: 39561971 DOI: 10.1002/psc.3662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024]
Abstract
Pharmaceuticals often suffer from limitations such as low solubility, low stability, and short half-life. To address these challenges and reduce the need for frequent drug administrations, a more efficient delivery is required. In this context, the development of controlled drug delivery systems, acting as a protective depot for the drug, has expanded significantly over the last decades. Among these, injectable hydrogels have emerged as a promising platform, especially in view of the rise of biologicals as therapeutics. Hydrogels are functional, solid-like biomaterials, composed of cross-linked hydrophilic polymers and high water content. Their physical properties, which closely mimic the extracellular matrix, make them suitable for various biomedical applications. This review discusses the different types of hydrogel systems and their self-assembly process, with an emphasis on peptide-based hydrogels. Due to their structural and functional diversity, biocompatibility, synthetic accessibility, and tunability, peptides are regarded as promising and versatile building blocks. A comprehensive overview of the variety of peptide hydrogels is outlined, with β-sheet forming sequences being highlighted. Key factors to consider when using peptide hydrogels as a controlled drug delivery system are reviewed, along with a discussion of the main drug release mechanisms and the emerging trend towards affinity-based systems to further refine drug release profiles.
Collapse
Affiliation(s)
- Julie Heremans
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Brussels, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Brussels, Belgium
| | - Charlotte Martin
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
44
|
Okafor NI, Omoteso OA, Choonara YE. The modification of conventional liposomes for targeted antimicrobial delivery to treat infectious diseases. DISCOVER NANO 2025; 20:19. [PMID: 39883380 PMCID: PMC11782757 DOI: 10.1186/s11671-024-04170-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/09/2024] [Indexed: 01/31/2025]
Abstract
Some of the most crucial turning points in the treatment strategies for some major infectious diseases including AIDS, malaria, and TB, have been reached with the introduction of antimicrobials and vaccines. Drug resistance and poor effectiveness are key limitations that need to be overcome. Conventional liposomes have been explored as a delivery system for infectious diseases bioactives to treat infectious diseases to provide an efficient approach to maximize the therapeutic outcomes, drug stability, targetability, to reduce the side-effects of antimicrobials, and enhance vaccine performance where necessary. However, as the pathological understanding of infectious diseases become more known, the need for more advanced liposomal technologies was born to continue having a profound effect on targeted chemotherapy for infectious diseases. This review therefore provides a concise incursion into the most recent and vogue liposomal formulations used to treat infectious diseases. An appraisal of immunological, stimuli-responsive, biomimetic and functionalized liposomes and other novel modifications to conventional liposomes is assimilated in sync with mutations of resistant pathogens.
Collapse
Affiliation(s)
- Nnamdi Ikemefuna Okafor
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | | | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
45
|
Chavda VP, Balar PC. Oral delivery of protein and peptide therapeutics. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 212:355-387. [PMID: 40122651 DOI: 10.1016/bs.pmbts.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Oral administration of proteins and peptides has gained significant attention recently due to its potential to transform therapeutic strategies, providing a non-invasive and patient-friendly method for delivering biopharmaceuticals. The primary hurdle in oral delivery stems from the harsh conditions of the gastrointestinal (GI) tract, characterized by acidic pH, enzymatic degradation, and limited permeability across the intestinal epithelium. Various innovative approaches have emerged to overcome these challenges, including nanoparticle-based delivery systems, mucoadhesive formulations, and chemical modifications of peptides aimed at improving stability and absorption rates. Nanoparticle-based delivery systems, such as liposomes, polymeric nanoparticles, and solid lipid nanoparticles, hold promise in protecting proteins and peptides from enzymatic degradation while enhancing their bioavailability. These nanoparticles can be tailored to target specific areas within the GI tract, extending drug release and enhancing therapeutic effectiveness. Mucoadhesive formulations utilize polymers like chitosan, alginate, and polyethylene glycol (PEG) derivatives to adhere to GI mucosal surfaces, prolonging residence time and facilitating drug absorption. Chemical modifications, such as PEGylation, glycosylation, and lipidation have been employed to enhance the stability and permeability of proteins and peptides in the GI tract. PEGylation, in particular, has been widely used to extend the circulation half-life and reduce the immunogenicity of therapeutic proteins. Advancements in nanotechnology, especially the development of smart nanocarriers capable of responsive drug release triggered by pH or enzymatic stimuli, show promise in further improving oral delivery of proteins and peptides. The integration of bioinformatics and computational modeling techniques has facilitated the design of novel drug delivery systems with optimized pharmacokinetic profiles. This chapter focuses on the advancements and challenges in the oral delivery of protein and peptide-based drugs, highlighting the innovative strategies being explored to enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India.
| | | |
Collapse
|
46
|
Suriyaamporn P, Pornpitchanarong C, Charoenying T, Dechsri K, Ngawhirunpat T, Opanasopit P, Pamornpathomkul B. Artificial intelligence-driven hydrogel microneedle patches integrating 5-fluorouracil inclusion complex-loaded flexible pegylated liposomes for enhanced non-melanoma skin cancer treatment. Int J Pharm 2025; 669:125072. [PMID: 39675535 DOI: 10.1016/j.ijpharm.2024.125072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/24/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
The current study focused on the development of crosslinked hydrogel microneedle patches (cHMNs) incorporating 5-FU-hydroxypropyl beta-cyclodextrin inclusion complex-loaded flexible PEGylated liposomes (5-FU-HPβCD-loaded FP-LPs) to enhance treatment efficacy and reduce drug toxicity. The research utilized artificial intelligence (AI) algorithms to design, optimize, and evaluate the cHMNs. Various AI models were assessed for accuracy, with metrics such as root mean square error and coefficient of determination guiding the selection of the most effective formulation. The physicochemical and mechanical properties, swelling behavior, in vitro skin permeation, and safety of the chosen cHMNs were tested. The results demonstrated that the 5-FU-HPβCD-loaded FP-LPs, stabilized with limonene, had an optimal particle size of 36.23 ± 2.42 nm, narrow size distribution, and zeta potential of -10.24 ± 0.37 mV, with high encapsulation efficiency. The cHMNs exhibited a conical needle shape with sufficient mechanical strength to penetrate the stratum corneum up to approximately 467.87 ± 65.12 μm. The system provided a high skin permeation rate of 41.78 ± 4.26 % and significant drug accumulation in the skin. Additionally, the formulation was proven safe in cell culture while effectively inhibiting cancer growth and promoting apoptosis. This study highlights the potential of AI-enhanced cHMNs for delivering 5-FU-HPβCD-loaded FP-LPs transdermally, offering a promising new treatment avenue for non-melanoma skin cancers.
Collapse
Affiliation(s)
- Phuvamin Suriyaamporn
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand; Research and Innovation Center for Advanced Therapy Medicinal Products, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Chaiyakarn Pornpitchanarong
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand; Research and Innovation Center for Advanced Therapy Medicinal Products, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Thapakorn Charoenying
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand; Research and Innovation Center for Advanced Therapy Medicinal Products, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Koranat Dechsri
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Tanasait Ngawhirunpat
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Praneet Opanasopit
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand; Research and Innovation Center for Advanced Therapy Medicinal Products, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Boonnada Pamornpathomkul
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand; Research and Innovation Center for Advanced Therapy Medicinal Products, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
| |
Collapse
|
47
|
Silva CG, Luz VF, Nunes VL, Verzoto ABM, Cotrim ACDM, dos Santos WB, França EL, Honorio-França AC. Colostrum-Derived Melatonin Plus PEG Microspheres Modulate the Oxidative Metabolism of Human Colostrum Phagocytes. Metabolites 2025; 15:57. [PMID: 39852399 PMCID: PMC11767932 DOI: 10.3390/metabo15010057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/02/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Exogenous melatonin adsorbed onto PEG microspheres can modulate the functional activity of phagocytes in colostrum, but no data are available on the activity of melatonin found in colostrum. Therefore, the objective of this study was to extract melatonin from human colostrum, develop and characterize PEG microspheres with the extracted melatonin adsorbed onto them, and evaluate the effects of this system on the oxidative metabolism of colostrum phagocytes. METHODS Thirty colostrum samples were collected; ten were used for melatonin extraction, while twenty were used to obtain phagocytes. Melatonin was extracted from the colostrum supernatant through affinity chromatography and quantified by ELISA. The polyethylene glycol microspheres produced were analyzed using fluorescence microscopy and flow cytometry. Oxidative metabolism was assessed by measuring the release of the superoxide anion and superoxide enzymes. A control was conducted using commercial melatonin. RESULTS The fluorescence microscopy and flow cytometry analyses demonstrated that PEG microspheres can adsorb melatonin. There was an increase in superoxide release in phagocytes incubated with colostrum-derived or synthetic melatonin. When exposed to bacteria, colostrum phagocytes treated with colostrum melatonin adsorbed to PEG microspheres exhibited increased superoxide, accompanied by a decrease in the release of superoxide dismutase (SOD) and a lower SOD-to-superoxide ratio. In contrast, synthetic melatonin reduced the release of superoxide and increased the release of the enzyme and the SOD-to-superoxide ratio. CONCLUSIONS These data highlight the importance of melatonin on cellular metabolism and suggest that colostrum-derived melatonin may be a more effective option for controlling oxidative metabolism, particularly during infectious processes.
Collapse
Affiliation(s)
- Caroline G. Silva
- Programa de Pós-Graduação em Ciência de Materiais, Campus Universitário do Araguaia, Universidade Federal de Mato Grosso, Barra do Garças 78605-091, Brazil; (C.G.S.); (V.F.L.); (A.C.d.M.C.); (W.B.d.S.)
| | - Viviane F. Luz
- Programa de Pós-Graduação em Ciência de Materiais, Campus Universitário do Araguaia, Universidade Federal de Mato Grosso, Barra do Garças 78605-091, Brazil; (C.G.S.); (V.F.L.); (A.C.d.M.C.); (W.B.d.S.)
| | - Victor L. Nunes
- Instituto de Ciências Biológicas e da Saúde, Campus Universitário do Araguaia, Universidade Federal de Mato Grosso, Barra do Garças 78605-091, Brazil; (V.L.N.); (A.B.M.V.)
| | - Ana B. M. Verzoto
- Instituto de Ciências Biológicas e da Saúde, Campus Universitário do Araguaia, Universidade Federal de Mato Grosso, Barra do Garças 78605-091, Brazil; (V.L.N.); (A.B.M.V.)
| | - Aron C. de M. Cotrim
- Programa de Pós-Graduação em Ciência de Materiais, Campus Universitário do Araguaia, Universidade Federal de Mato Grosso, Barra do Garças 78605-091, Brazil; (C.G.S.); (V.F.L.); (A.C.d.M.C.); (W.B.d.S.)
| | - Wagner B. dos Santos
- Programa de Pós-Graduação em Ciência de Materiais, Campus Universitário do Araguaia, Universidade Federal de Mato Grosso, Barra do Garças 78605-091, Brazil; (C.G.S.); (V.F.L.); (A.C.d.M.C.); (W.B.d.S.)
| | - Eduardo L. França
- Programa de Pós-Graduação em Ciência de Materiais, Campus Universitário do Araguaia, Universidade Federal de Mato Grosso, Barra do Garças 78605-091, Brazil; (C.G.S.); (V.F.L.); (A.C.d.M.C.); (W.B.d.S.)
- Instituto de Ciências Biológicas e da Saúde, Campus Universitário do Araguaia, Universidade Federal de Mato Grosso, Barra do Garças 78605-091, Brazil; (V.L.N.); (A.B.M.V.)
| | - Adenilda C. Honorio-França
- Programa de Pós-Graduação em Ciência de Materiais, Campus Universitário do Araguaia, Universidade Federal de Mato Grosso, Barra do Garças 78605-091, Brazil; (C.G.S.); (V.F.L.); (A.C.d.M.C.); (W.B.d.S.)
- Instituto de Ciências Biológicas e da Saúde, Campus Universitário do Araguaia, Universidade Federal de Mato Grosso, Barra do Garças 78605-091, Brazil; (V.L.N.); (A.B.M.V.)
| |
Collapse
|
48
|
Damiri F, Fatimi A, Liu Y, Musuc AM, Fajardo AR, Gowda BHJ, Vora LK, Shavandi A, Okoro OV. Recent advances in 3D bioprinted polysaccharide hydrogels for biomedical applications: A comprehensive review. Carbohydr Polym 2025; 348:122845. [PMID: 39567171 DOI: 10.1016/j.carbpol.2024.122845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 11/22/2024]
Abstract
Polysaccharide hydrogels, which can mimic the natural extracellular matrix and possess appealing physicochemical and biological characteristics, have emerged as significant bioinks for 3D bioprinting. They are highly promising for applications in tissue engineering and regenerative medicine because of their ability to enhance cell adhesion, proliferation, and differentiation in a manner akin to the natural cellular environment. This review comprehensively examines the fabrication methods, characteristics, and applications of polysaccharide hydrogel-driven 3D bioprinting, underscoring its potential in tissue engineering, drug delivery, and regenerative medicine. To contribute pertinent knowledge for future research in this field, this review critically examines key aspects, including the chemistry of carbohydrates, manufacturing techniques, formulation of bioinks, and characterization of polysaccharide-based hydrogels. Furthermore, this review explores the primary advancements and applications of 3D-printed polysaccharide hydrogels, encompassing drug delivery systems with controlled release kinetics and targeted therapy, along with tissue-engineered constructs for bone, cartilage, skin, and vascular regeneration. The use of these 3D bioprinted hydrogels in innovative research fields, including disease modeling and drug screening, is also addressed. Despite notable progress, challenges, including modulating the chemistry and properties of polysaccharides, enhancing bioink printability and mechanical properties, and achieving long-term in vivo stability, have been highlighted.
Collapse
Affiliation(s)
- Fouad Damiri
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium; Chemical Science and Engineering Research Team (ERSIC), Department of Chemistry, Polydisciplinary Faculty of Beni Mellal (FPBM), Sultan Moulay Slimane University (USMS), Beni Mellal 23000, Morocco.
| | - Ahmed Fatimi
- Chemical Science and Engineering Research Team (ERSIC), Department of Chemistry, Polydisciplinary Faculty of Beni Mellal (FPBM), Sultan Moulay Slimane University (USMS), Beni Mellal 23000, Morocco
| | - Yang Liu
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Adina Magdalena Musuc
- "Ilie Murgulescu" Institute of Physical Chemistry, 202 Spl. Independentei, 060021 Bucharest, Romania
| | - André R Fajardo
- Laboratório de Tecnologia e Desenvolvimento de Compósitos e Materiais Poliméricos (LaCoPol), Federal University of Pelotas (UFPel), 96010-900 Pelotas, RS, Brazil
| | - B H Jaswanth Gowda
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, United Kingdom
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, United Kingdom.
| | - Armin Shavandi
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium
| | - Oseweuba V Okoro
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050 Brussels, Belgium
| |
Collapse
|
49
|
Dinescu VC, Martin L, Bica M, Vasile RC, Gresita A, Bunescu M, Ruscu MA, Aldea M, Rotaru-Zavaleanu AD. Hydrogel-Based Innovations in Carpal Tunnel Syndrome: Bridging Pathophysiological Complexities and Translational Therapeutic Gaps. Gels 2025; 11:52. [PMID: 39852023 PMCID: PMC11764971 DOI: 10.3390/gels11010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/24/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Carpal Tunnel Syndrome (CTS) is a prevalent neuropathic disorder caused by chronic compression of the median nerve, leading to sensory and motor impairments. Conventional treatments, such as corticosteroid injections, wrist splinting, and surgical decompression, often fail to provide adequate outcomes for chronic or recurrent cases, emphasizing the need for innovative therapies. Hydrogels, highly biocompatible three-dimensional biomaterials with customizable properties, hold significant potential for CTS management. Their ability to mimic the extracellular matrix facilitates localized drug delivery, anti-adhesion barrier formation, and tissue regeneration. Advances in hydrogel engineering have introduced stimuli-responsive systems tailored to the biomechanical environment of the carpal tunnel, enabling sustained therapeutic release and improved tissue integration. Despite these promising developments, hydrogel applications for CTS remain underexplored. Key challenges include the absence of CTS-specific preclinical models and the need for rigorous clinical validation. Addressing these gaps could unlock the full potential of hydrogel-based interventions, which offer minimally invasive, customizable solutions that could improve long-term outcomes and reduce recurrence rates. This review highlights hydrogels as a transformative approach to CTS therapy, advocating for continued research to address translational barriers. These innovations have the potential to redefine the treatment landscape, significantly enhancing patient care and quality of life.
Collapse
Affiliation(s)
- Venera Cristina Dinescu
- Department of Health Promotion and Occupational Medicine, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Str., 200349 Craiova, Romania;
| | - Liviu Martin
- Faculty of Medical Care, Titu Maiorescu University, Văcărești Road, no 187, 040051 Bucharest, Romania;
| | - Marius Bica
- Department of Surgery, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Str., 200349 Craiova, Romania;
| | - Ramona Constantina Vasile
- Department of Epidemiology, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Str., 200349 Craiova, Romania; (M.A.R.); (A.D.R.-Z.)
| | - Andrei Gresita
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Str., 200349 Craiova, Romania
| | - Marius Bunescu
- Department of Occupational Medicine, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Str., 200349 Craiova, Romania;
| | - Mihai Andrei Ruscu
- Department of Epidemiology, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Str., 200349 Craiova, Romania; (M.A.R.); (A.D.R.-Z.)
| | - Madalina Aldea
- Department of Psychiatry, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Str., 200349 Craiova, Romania;
| | - Alexandra Daniela Rotaru-Zavaleanu
- Department of Epidemiology, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Str., 200349 Craiova, Romania; (M.A.R.); (A.D.R.-Z.)
| |
Collapse
|
50
|
Dias MF, Cruz-Cazarim ELC, Pittella F, Baião A, Pacheco AC, Sarmento B, Fialho SL. Co-delivery of antioxidants and siRNA-VEGF: promising treatment for age-related macular degeneration. Drug Deliv Transl Res 2025:10.1007/s13346-024-01772-x. [PMID: 39751765 DOI: 10.1007/s13346-024-01772-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
Current treatments for retinal disorders are anti-angiogenic agents, laser photocoagulation, and photodynamic therapies. These conventional treatments focus on reducing abnormal blood vessel formation in the retina, which, in a low-oxygen environment, can lead to harmful proliferation of endothelial cells. This results in dysfunctional, leaky blood vessels that cause retinal edema, hemorrhage, and vision loss. Age-related Macular Degeneration is a primary cause of vision loss and blindness in the elderly, impacting around 20% of those over 50 years old. This complex disease is also closely related to oxidative stress in retina. In this review, we explore the challenge of treating retinal diseases, alternatives and possibilities of enhancing the effectiveness of therapies using co-delivery systems containing both antiangiogenic and antioxidant therapeutic agents. Despite recent proposals potential, the lack of extensive clinical studies on the long-term outcomes and optimal combinations of therapies means that the full risk profile and effectiveness of combined therapy are not yet completely understood. These factors must be carefully considered and managed by healthcare providers to optimize treatment outcomes and ensure patient safety.
Collapse
Affiliation(s)
- Marina F Dias
- Pharmaceutical Research and Development, Ezequiel Dias Foundation, Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, CEP 30510-010, Minas Gerais, Brazil
| | - Estael L C Cruz-Cazarim
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Juiz de Fora, Juiz de Fora, CEP 36036-900, Minas Gerais, Brazil
| | - Frederico Pittella
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Juiz de Fora, Juiz de Fora, CEP 36036-900, Minas Gerais, Brazil
| | - Ana Baião
- i3S - Instituto Nacional de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Instituto de Engenharia Biomédica, INEB, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, ICBAS, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Ana Catarina Pacheco
- i3S - Instituto Nacional de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Instituto de Engenharia Biomédica, INEB, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- CESPU-IUCS, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal
| | - Bruno Sarmento
- i3S - Instituto Nacional de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Instituto de Engenharia Biomédica, INEB, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- CESPU-IUCS, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal
| | - Silvia L Fialho
- Pharmaceutical Research and Development, Ezequiel Dias Foundation, Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, CEP 30510-010, Minas Gerais, Brazil.
| |
Collapse
|