1
|
Liu H, Zhu Y, Chen W, Sheng R, Liu C, Sun Y, Liu J, Wang M, Lu J, Chen J, Zhang W. Fullerol Initiates Stem Cell-Nanomaterials Interactions for Enhanced Tissue Regeneration via Clathrin-Mediated Endocytosis and Nuclear Factor Erythroid 2-Related Factor 2 Signaling. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40241445 DOI: 10.1021/acsami.5c01731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
The advancement of nanomedicine requires a thorough understanding of the intrinsic bioactivity and molecular interactions of nanomaterials for safe and effective clinical applications, which remains lacking for most currently developed nanomaterials. Here, we uncover the unique intrinsic bioactivity and regulatory mechanisms of carbon-based fullerol nanomaterials through high-throughput molecular analysis and explore their therapeutic potential for tissue regeneration using tissue engineering approaches. Fullerol exhibits intrinsic pro-differentiation and antioxidant properties that enhance the osteogenesis and chondrogenesis of MSCs. Mechanistically, proteomic analysis combined with small-molecule inhibition studies reveals that fullerol is internalized by MSCs via clathrin-mediated endocytosis and activates NRF2 signaling, thereby exerting antioxidant effects that restore impaired MSC viability and differentiation under oxidative stress. Leveraging these unique bioactivities, we develop a fullerol-functionalized hydrogel with feasible physicochemical properties and triple biological functions in antioxidant, pro-osteogenic, and pro-chondrogenic effects and confirm its great regenerative capacity for both cartilage and subchondral bone by promoting structural restoration and improving functional recovery in a rat osteochondral defect model. Our findings offer new insights into the intricate interactions between stem cells and nanomaterials at the cellular and molecular levels and broaden the potential biomedical applications of fullerol for future cartilage and bone regeneration therapies.
Collapse
Affiliation(s)
- Haoyang Liu
- School of Medicine, and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210009 Nanjing, China
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, Southeast University, 210009 Nanjing, China
| | - Yue Zhu
- School of Medicine, and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210009 Nanjing, China
| | - Weixu Chen
- School of Medicine, and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210009 Nanjing, China
- Xuzhou Central Hospital Affiliated to Medical School of Southeast University, 221000 Xuzhou, China
| | - Renwang Sheng
- School of Medicine, and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210009 Nanjing, China
| | - Chuanquan Liu
- School of Medicine, and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210009 Nanjing, China
| | - Yuzhi Sun
- School of Medicine, and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210009 Nanjing, China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, 210008 Nanjing, China
| | - Jia Liu
- School of Medicine, and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210009 Nanjing, China
| | - Mingyue Wang
- School of Medicine, and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210009 Nanjing, China
| | - Jun Lu
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, Southeast University, 210009 Nanjing, China
| | - Jialin Chen
- School of Medicine, and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210009 Nanjing, China
- Department of Ophthalmology, Zhongda Hospital, Southeast University, 210009 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| | - Wei Zhang
- School of Medicine, and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210009 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| |
Collapse
|
2
|
Zhou J, Wang G, Zhou Y, Lin X, Zhao Z, Xue Y, An Y, Shao H, Wang Y, Hou S, Wang L, Fan Y. Bioinspired Lipid Nanoparticles with Prolonged Cartilage Retention Boost Regeneration in Early Osteoarthritis and Large Cartilage Defects. ACS NANO 2025; 19:13654-13672. [PMID: 40184476 DOI: 10.1021/acsnano.4c13828] [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: 04/06/2025]
Abstract
Osteoarthritis (OA) leads to the progressive degeneration of articular cartilage, yet there is currently no effective treatment available for both the early and late stages of osteoarthritis. Cartilage regeneration requires the action and prolonged retention of multiple drugs at injured sites to recruit endogenous cells and facilitate cartilage formation. Here, we propose a cartilage-binding-peptide-modified lipid nanoparticle as a drug carrier to achieve sustained release of protein (TGF-β3) and small molecular drugs (KGN) for one month. Through systematic screening of multiple peptides targeting collagen II or chondrocytes, we identify a decorin-derived-peptide-modified lipid nanoparticle with precise targeting and prolonged retention capability in cartilage. Improved nanoparticle stability, high drug loading, and sustainable dual-drug release are achieved through interbilayer cross-linking of adjacent lipid bilayers within multilamellar vesicles. In a surgical model of rat OA, the nanoparticle loading with TGF-β3 and KGN protects injured cartilage from degeneration progression. For severe cartilage injury (full-thickness defects) in a rabbit model, the nanoparticle facilitates the regeneration of high-quality hyaline-like cartilage, which is a rare achievement in full-thickness cartilage regeneration through nanoparticle-based drug delivery. This work presents a strategy for the rational design of bioinspired cartilage-binding peptide-modified lipid-based drug carriers to promote hyaline-like cartilage regeneration.
Collapse
Affiliation(s)
- Jin Zhou
- Medical Engineering & Engineering Medicine Innovation Center, Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Guanhuier Wang
- Department of Plastic and Reconstructive Surgery, Peking University Third Hospital, NO.49 of North Huayuan Road, Haidian District, Beijing 100191, China
| | - Yue Zhou
- Medical Engineering & Engineering Medicine Innovation Center, Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xubo Lin
- Medical Engineering & Engineering Medicine Innovation Center, Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China
| | - Zhenmin Zhao
- Department of Plastic and Reconstructive Surgery, Peking University Third Hospital, NO.49 of North Huayuan Road, Haidian District, Beijing 100191, China
| | - Yumeng Xue
- Medical Engineering & Engineering Medicine Innovation Center, Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yang An
- Department of Plastic and Reconstructive Surgery, Peking University Third Hospital, NO.49 of North Huayuan Road, Haidian District, Beijing 100191, China
| | - Hui Shao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Ying Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Sen Hou
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Lizhen Wang
- Medical Engineering & Engineering Medicine Innovation Center, Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yubo Fan
- Medical Engineering & Engineering Medicine Innovation Center, Hangzhou International Innovation Institute, Beihang University, Hangzhou 311115, China
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
3
|
Voitovich I, Ty N, Couderc M, Moreau E, Peyrode C, Weber V. Design, synthesis and in vitro evaluation of non-steroidal anti-inflammatory prodrugs for osteoarthritis. Bioorg Chem 2025; 160:108410. [PMID: 40220712 DOI: 10.1016/j.bioorg.2025.108410] [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: 01/10/2025] [Revised: 03/03/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025]
Abstract
Osteoarthritis (OA), a degenerative joint disease characterized by chronic pain and stiffness, is the most common cause of disability in older adults. To date, OA lacks curative treatment and medical care is limited to symptom relief particularly through the use of oral nonsteroidal anti-inflammatory drugs (NSAIDs). However, gastrointestinal and cardiovascular adverse effects and only limited benefits in long-term relief of pain are still associated. Moreover, efficiency is in part impeded by rapid clearance of the drugs and by the special physiological environment of the joint impeding deep penetration of drugs. Hence, to overcome those limitations and improve patient outcome, developing new therapeutic approaches based on anti-inflammatory prodrugs with prolonged drug residence time within the joints appears as a promising strategy in OA. We report herein the development of NSAID prodrugs, derived from diclofenac and naproxen, bearing a positively charged quaternary ammonium (QA) to target the negative-fixed charge density in cartilage by the mean of electrostatic interactions. Our charge-based targeted approach aims to extend the residence time of NSAID within the cartilaginous tissues, leading to a potential decrease of side effects and improved efficiency of locally released drugs. Syntheses of various amide and esters prodrugs of diclofenac and naproxen bearing a QA function were performed, including some hypoxia-activated prodrugs. Since most diclofenac derivatives suffered from high instability preventing any further development, we focused on the naproxen derivatives that were relatively stable in PBS buffer over a 24-h period even if three different degradation patterns were observed in murine plasma. A preliminary screening of their in vitro anti-inflammatory efficacy highlighted a correlation between the PGE-2 inhibition and these cleavage patterns. These results support further in vitro and in vivo evaluations of four of these derivatives in OA models.
Collapse
Affiliation(s)
- Iuliia Voitovich
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Nancy Ty
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Marion Couderc
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France; Rheumatology Department, Gabriel Montpied University Hospital, Clermont-Ferrand, France
| | - Emmanuel Moreau
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Caroline Peyrode
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Valérie Weber
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
4
|
Xie S, Liao P, Mi S, Song L, Chen X. Emerging patterns in nanoparticle-based therapeutic approaches for rheumatoid arthritis: A comprehensive bibliometric and visual analysis spanning two decades. Open Life Sci 2025; 20:20251071. [PMID: 40129468 PMCID: PMC11931663 DOI: 10.1515/biol-2025-1071] [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: 12/01/2024] [Revised: 02/04/2025] [Accepted: 02/04/2025] [Indexed: 03/26/2025] Open
Abstract
The aim of this study is to analyze scientific literature to investigate the current research status, focus areas, and developmental trends in nanoparticle systems for rheumatoid arthritis (RA) therapy. To do that, Research articles published from 2003 to 2023 were retrieved from the Web of Science database, and analysis included quantitative output, distribution by country/region, collaborative publishing data, influential authors, high-yield institutions, keywords, hotspots, and development trends. Visual knowledge maps were generated using VOSviewer and Citespace. Findings reveal a steady increase in publications related to nanoparticle systems for RA therapy, indicating growing global interest. China leads with 487 papers (37.433%), followed by the United States (233, 17.909%), India (179, 13.759%), South Korea (89, 6.841%), and Egypt (50, 3.843%). Active collaboration is observed, particularly between the United States and countries such as China, Germany, Saudi Arabia, India, England, and Pakistan. The Chinese Academy of Sciences ranks first in total articles published (55), with Liu Y from China being the most prolific author. The Journal of Controlled Release emerges as a primary outlet in this field. Primary keyword clusters include "Drug delivery systems," "Gold nanoparticles," "Transdermal delivery," "Angiogenesis," "Collagen-induced arthritis," "Rheumatoid arthritis," "Oxidant stress," "Dendritic cells," and "pH sensitive." Research hotspots with great development potential include "Immunopathological Mechanisms," "Novel drugs," and "Smart delivery system." In conclusion, research on nanoparticle systems for RA therapy has significantly expanded over the past two decades, with a focus on elucidating pathogenetic mechanisms and advancing novel drug delivery strategies anticipated to be prominent in the foreseeable future.
Collapse
Affiliation(s)
- Shenwei Xie
- Department of Rheumatology and Immunology, Hunan University of Medicine General Hospital, HuaiHua, 418000, China
| | - Pan Liao
- Department of Rheumatology and Immunology, Hunan University of Medicine General Hospital, HuaiHua, 418000, China
| | - Shuang Mi
- Department of Respiratory and Critical Care Medicine, Shenzhen Yantian District People’s Hospital, Shenzhen, 518000, China
| | - Liang Song
- Department of Rheumatology and Immunology, Hunan University of Medicine General Hospital, HuaiHua, 418000, China
| | - Xiaoyuan Chen
- Department of Respiratory and Critical Care Medicine, Shenzhen Yantian District People’s Hospital, Shenzhen, 518000, China
| |
Collapse
|
5
|
Sriram A, Ithape H, Singh PK. Deep-insights: Nanoengineered gel-based localized drug delivery for arthritis management. Asian J Pharm Sci 2025; 20:101012. [PMID: 39995751 PMCID: PMC11848107 DOI: 10.1016/j.ajps.2024.101012] [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: 07/17/2023] [Revised: 05/15/2024] [Accepted: 07/03/2024] [Indexed: 02/26/2025] Open
Abstract
Arthritis is an inflammatory joint disorder that progressively impairs function and diminishes quality of life. Conventional therapies often prove ineffective, as oral administration lacks specificity, resulting in off-target side effects like hepatotoxicity and GIT-related issues. Intravenous administration causes systemic side effects. The characteristic joint-localized symptoms such as pain, stiffness, and inflammation make the localized drug delivery suitable for managing arthritis. Topical/transdermal/intra-articular routes have become viable options for drug delivery in treating arthritis. However, challenges with those localized drug delivery routes include skin barrier and cartilage impermeability. Additionally, conventional intra-articular drug delivery also leads to rapid clearance of drugs from the synovial joint tissue. To circumvent these limitations, researchers have developed nanocarriers that enhance drug permeability through skin and cartilage, influencing localized action. Gel-based nanoengineered therapy employs a gel matrix to incorporate the drug-encapsulated nanocarriers. This approach combines the benefits of gels and nanocarriers to enhance therapeutic effects and improve patient compliance. This review emphasizes deep insights into drug delivery using diverse gel-based novel nanocarriers, exploring their various applications embedded in hyaluronic acid (biopolymer)-based gels, carbopol-based gels, and others. Furthermore, this review discusses the influence of nanocarrier pharmacokinetics on the localization and therapeutic manipulation of macrophages mediated by nanocarriers. The ELVIS (extravasation through leaky vasculature and inflammatory cell-mediated sequestration) effect associated with arthritis is advantageous in drug delivery. Simply put, the ELVIS effect refers to the extravasation of nanocarriers through leaky vasculatures, which finally results in the accumulation of nanocarriers in the joint cavity.
Collapse
Affiliation(s)
| | | | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Telangana 500037, India
| |
Collapse
|
6
|
Morici L, Allémann E, Rodríguez-Nogales C, Jordan O. Cartilage-targeted drug nanocarriers for osteoarthritis therapy. Int J Pharm 2024; 666:124843. [PMID: 39424088 DOI: 10.1016/j.ijpharm.2024.124843] [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: 07/30/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Osteoarthritis (OA) is a joint disease common worldwide. Currently, no disease-modifying osteoarthritis drugs (DMOADs) have successfully passed clinical trials, often due to a lack of cartilage penetration. Thus, targeting the extracellular matrix (ECM) is a major priority. The design of cartilage-targeting drug delivery systems (DDSs) for intra-articular administration requires consideration of the physicochemical properties of articular cartilage, such as its porosity and negative fixed charge. Various positively charged biomaterials such as polyaminoacids, proteins, polymers, and lipids can be used as DDSs to enhance cartilage penetration. Cationic nanocarriers interact electrostatically with anionic glycosaminoglycans of the ECM, ensuring passive cartilage-targeting penetration and prolonged retention. Active targeting strategies involve DDSs surface decoration using antibodies or peptides with a strong affinity for collagen II and chondrocytes in the cartilage. This review presents all the relevant bio-physicochemical properties of healthy and OA cartilages, as well as state-of-the-art intra-articular cartilage-targeted DDSs, intending to better understand the recent advances in the application of cartilage-targeting delivery systems for OA therapy.
Collapse
Affiliation(s)
- Luca Morici
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1206 Geneva, Switzerland
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1206 Geneva, Switzerland
| | - Carlos Rodríguez-Nogales
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1206 Geneva, Switzerland.
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1206 Geneva, Switzerland.
| |
Collapse
|
7
|
Sun M, Ma B, Pan Z, Zhao Y, Tian L, Fan Y, Kong W, Wang J, Xu B, Ao Y, Guo Q, Wang X, Peng X, Li X, Cheng J, Miao L, Wang K, Hu X. Targeted Therapy of Osteoarthritis via Intra-Articular Delivery of Lipid-Nanoparticle-Encapsulated Recombinant Human FGF18 mRNA. Adv Healthc Mater 2024; 13:e2400804. [PMID: 39363784 PMCID: PMC11582510 DOI: 10.1002/adhm.202400804] [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: 03/01/2024] [Revised: 07/08/2024] [Indexed: 10/05/2024]
Abstract
Fibroblast growth factor 18 (FGF18) emerges as a promising therapeutic target for osteoarthritis (OA). In this study, a novel articular cavity-localized lipid nanoparticle (LNP) named WG-PL14 is developed. This optimized formulation has a nearly 30-fold increase in mRNA expression as well as better articular cavity enrichment compared to commercial lipids MC3 when performing intra-articular injection. Then, a mRNA sequence encoding recombinant human FGF18 (rhFGF18) for potential mRNA therapy in OA is optimized. In vitro assays confirm the translation of rhFGF18 mRNA into functional proteins within rat and human chondrocytes, promoting cell proliferation and extracellular matrix (ECM) synthesis. Subsequently, the therapeutic efficacy of the LNP-rhFGF18 mRNA complex is systematically assessed in a mouse OA model. The administration exhibits several positive outcomes, including an improved pain response, upregulation of ECM-related genes (e.g., AGRN and HAS2), and remodeling of subchondral bone homeostasis compared to a control group. Taken together, these findings underscore the potential of localized LNP-rhFGF18 mRNA therapy in promoting the regeneration of cartilage tissue and mitigating the progression of OA.
Collapse
Affiliation(s)
- Mengze Sun
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Bin Ma
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- Beijing Key Laboratory of Molecular PharmaceuticsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Zihang Pan
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Yun Zhao
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Liangliang Tian
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Yifei Fan
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Weijing Kong
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Junyan Wang
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Boyang Xu
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Yingfang Ao
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Quanyi Guo
- Institute of OrthopedicsThe Fourth Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLANo. 28 Fuxing Road, Haidian DistrictBeijing100853China
| | - Xi Wang
- State Key Laboratory of Female Fertility PromotionClinical Stem Cell Research CenterPeking University Third HospitalBeijing100191China
| | - Xiaohong Peng
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Xiaoxia Li
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Jin Cheng
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Lei Miao
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- Beijing Key Laboratory of Molecular PharmaceuticsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Kai Wang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Xiaoqing Hu
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| |
Collapse
|
8
|
Chen K, Wang J, Cao J, Liu F, Fang J, Zheng W, Liu S, Zhao Y, Shuai X, Huang J, Chen B. Enzyme-responsive microgel with controlled drug release, lubrication and adhesion capability for osteoarthritis attenuation. Acta Biomater 2024:S1742-7061(24)00618-4. [PMID: 39427765 DOI: 10.1016/j.actbio.2024.10.026] [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: 04/16/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
The treatment of osteoarthritis (OA) remains challenging due to the narrow therapeutic window and rapid clearance of therapeutic agents, even with intra-articular administration, resulting in a low treatment index. Recent advancements in local drug delivery systems have yet to overcome the issues of uncontrolled burst release and short retention time, leading to suboptimal OA treatment outcome. Herein, we developed a methacrylate-crosslinking hyaluronic acid (HA) microgel (abbreviated as CXB-HA-CBP) that covalently conjugates the anti-inflammatory drug celecoxib (CXB) via a metalloproteinase-2 (MMP-2)-responsive peptide linker (GGPLGLAGGC) and a collagen II binding peptide (WYRGRLC). The GGPLGLAGGC linker is specifically cleaved by the overexpressed MMP-2 enzyme within the OA joint, enabling the sustained and on-demand release of CXB entity. The synergistic action of CXB and HA effectively inhibited macrophage activation and reduced the production of pro-inflammatory cytokines, protecting chondrocytes from damage. Furthermore, the collagen II peptide introduced on the microgel surface enabled a cartilage-binding function to form an artificial lubrication microgel layer on the cartilage surface to reduce cartilage wear. The CXB-HA-CBP microgel showed an extended retention time of up to 18 days in the affected joint, leading to an effective OA treatment in rats. This sophistically designed microgel, characterized by the prolonged retention time, sustained drug delivery, and enhanced lubrication, presents a promising biomedicine for OA treatment. STATEMENT OF SIGNIFICANCE: A new methacrylate-crosslinking hyaluronic acid (HA) microgel, covalently conjugated with the celecoxib (CXB)-GGPLGLAGGC and the collagen II binding peptide (CBP, peptide sequence: WYRGRLC), was developed. The overexpressed MMP-2 in OA joint cleaved the GGPLGLAGGC linker to trigger the CXB moiety release. Besides, the CBP on the surface of microgels enabled a cartilage-attaching ability, resulting in a prolonged retention time and an improved lubrication property in joint. This advanced drug-loading microgel remarkably reduced macrophage activation and pro-inflammation cytokine production, while protecting the chondrocytes via a dual action of CXB and HA. This study demonstrated that the enzyme-responsive drug-loading microgel could serve as an platform to efficiently attenuate osteoarthritis.
Collapse
Affiliation(s)
- Keyu Chen
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiachen Wang
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jue Cao
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Liu
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jintao Fang
- Department of Microsurgery, Trauma and Hand Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Weixin Zheng
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shubo Liu
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuexin Zhao
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China; PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Jinsheng Huang
- Department of Urology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China.
| | - Bin Chen
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Fei Y, Li X, Lv Z, Liu Z, Xie Y, Chen J, Li W, Liu X, Guo H, Liu H, Zhang Z, Wang X, Fan J, Hu C, Jin X, Jiang R, Xu N, Xia J, Li Y, Shi D. Promoting chondrogenesis by targeted delivery to the degenerating cartilage in early treatment of osteoarthritis. Bioact Mater 2024; 40:624-633. [PMID: 39247402 PMCID: PMC11377143 DOI: 10.1016/j.bioactmat.2024.08.004] [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: 04/08/2024] [Revised: 07/21/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Osteoarthritis (OA) is a highly incident total joint degenerative disease with cartilage degeneration as the primary pathogenesis. The cartilage matrix is mainly composed of collagen, a matrix protein with a hallmark triple-helix structure, which unfolds with collagen degradation on the cartilage surface. A collagen hybridizing peptide (CHP) is a synthetic peptide that binds the denatured collagen triple helix, conferring a potential disease-targeting possibility for early-stage OA. Here, we constructed an albumin nanoparticle (An) conjugated with CHP, loaded with a chondrogenesis-promoting small molecule drug, kartogenin (KGN). The CHP-KGN-An particle exhibited sustained release of KGN in vitro and prolonged in vivo retention selectively within the degenerated cartilage in the knee joints of model mice with early-stage OA. Compared to treatment with KGN alone, CHP-KGN-An robustly attenuated cartilage degradation, synovitis, osteophyte formation, and subchondral bone sclerosis in OA model mice and exhibited a more prominent effect on physical activity improvement and pain alleviation. Our study showcases that targeting the degenerated cartilage by collagen hybridization can remarkably promote the efficacy of small molecule drugs and may provide a novel delivery strategy for early-stage OA therapeutics.
Collapse
Affiliation(s)
- Yuxiang Fei
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xiaojing Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, PR China
| | - Zhongyang Lv
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China
| | - Zizheng Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Ya Xie
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Jiaqi Chen
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Weitong Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xiyu Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Hu Guo
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Huan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Zhaofeng Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xunhao Wang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Jingjing Fan
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Chunqing Hu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xiaoyu Jin
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Ruiyang Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Nuo Xu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR China
| | - Yang Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, PR China
| | - Dongquan Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| |
Collapse
|
10
|
Yu T, Wang J, Zhou Y, Ma C, Bai R, Huang C, Wang S, Liu K, Han B. Harnessing Engineered Extracellular Vesicles from Mesenchymal Stem Cells as Therapeutic Scaffolds for Bone‐Related Diseases. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202402861] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Indexed: 10/05/2024]
Abstract
AbstractMesenchymal stem cells (MSCs) play a crucial role in maintaining bone homeostasis and are extensively explored for cell therapy in various bone‐related diseases. In addition to direct cell therapy, the secretion of extracellular vesicles (EVs) by MSCs has emerged as a promising alternative approach. MSC‐derived EVs (MSC‐EVs) offer equivalent therapeutic efficacy to MSCs while mitigating potential risks. These EVs possess unique properties that enable them to traverse biological barriers and deliver bioactive cargos to target cells. Furthermore, by employing modification and engineering strategies, the therapeutic effects and tissue targeting specificity of MSC‐EVs can be further enhanced to meet specific therapeutic needs. In this review, the mechanisms and advantages of MSC‐EV therapy in diseased bone tissues are highlighted. Through simple isolation and modification techniques, MSC‐EV‐based biomaterials have demonstrated great promise for bone regeneration. Finally, future perspectives on MSC‐EV therapy are presented, envisioning the development of next‐generation regenerative materials and bioactive agents for clinical translation in the field of bone regeneration.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Jingwei Wang
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Yusai Zhou
- School of Materials Science and Engineering Beihang University Beijing 100191 P. R. China
| | - Chao Ma
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education) Department of Chemistry Tsinghua University Beijing 100084 P. R. China
| | - Rushui Bai
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Cancan Huang
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Shidong Wang
- Musculoskeletal Tumor Center Peking University People's Hospital No.11 Xizhimen South St. Beijing 100044 P. R. China
| | - Kai Liu
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education) Department of Chemistry Tsinghua University Beijing 100084 P. R. China
| | - Bing Han
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| |
Collapse
|
11
|
Makled S, Abbas H, Ali ME, Zewail M. Melatonin hyalurosomes in collagen thermosensitive gel as a potential repurposing approach for rheumatoid arthritis management via the intra-articular route. Int J Pharm 2024; 661:124449. [PMID: 38992734 DOI: 10.1016/j.ijpharm.2024.124449] [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: 02/23/2024] [Revised: 06/30/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
Despite the fact that several rheumatoid arthritis treatments have been utilized, none of them achieved complete joint healing and has been accompanied by several side effects that compromise patient compliance. This study aims to provide an effective safe RA treatment with minimum side effects through the encapsulation of melatonin (MEL) in hyalurosomes and loading these hyalurosomes in collagen thermos-sensitive poloxamer 407 (PCO) hydrogels, followed by their intra-articular administration in AIA model rats. In vitro characterization of MEL-hyalurosomes and PCO hydrogel along with in vivo evaluation of the selected formulation were conducted. Particle size, PDI and EE % of the selected formulation were 71.5 nm, 0.09 and 90 %. TEM micrographs demonstrated that the particles had spherical shape with no aggregation signs. Loading PCO hydrogels with MEL-hyalurosomes did not cause significant changes in pH although it increased its viscosity and injection time. FTIR analysis showed that no interactions were noted among the delivery system components. In vivo results revealed the superior effect of MEL-hyalurosomes PCO hydrogel over MEL-PCO hydrogel and blank PCO hydrogels in improving joint healing, cartilage repair, pannus formation and cell infiltrations. Also, MEL-hyalurosomes PCO hydrogel group showed comparable levels of TNF-α, IL1, MDA, NRF2 and HO-1 with the negative control group. These findings highlight the MEL encapsulation role in augmenting its pharmacological effects along with the synergistic effect of hyaluronic acid in hyalurosomes and collagen in PCO hydrogel in promoting joint healing.
Collapse
Affiliation(s)
- Shaimaa Makled
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, 21521, Egypt
| | - Haidy Abbas
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Egypt P.O. Box 22511, Damanhour, Egypt.
| | - Merhan E Ali
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Mariam Zewail
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Egypt P.O. Box 22511, Damanhour, Egypt
| |
Collapse
|
12
|
Ji Z, Ren X, Jin J, Ye X, Yu H, Fang W, Li H, Zhao Y, Tao S, Kong X, Cheng J, Shan Z, Chen J, Yao Q, Zhao F, Liu J. Injectable hydrogel encapsulating siMMP13 with anti-ROS and anti-apoptotic functions for osteoarthritis treatment. J Nanobiotechnology 2024; 22:466. [PMID: 39095867 PMCID: PMC11297633 DOI: 10.1186/s12951-024-02740-w] [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: 04/12/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative joint disease characterized by the progressive degeneration of articular cartilage, leading to pain, stiffness, and loss of joint function. The pathogenesis of OA involves multiple factors, including increased intracellular reactive oxygen species (ROS), enhanced chondrocyte apoptosis, and disturbances in cartilage matrix metabolism. These processes contribute to the breakdown of the extracellular matrix (ECM) and the loss of cartilage integrity, ultimately resulting in joint damage and dysfunction. RNA interference (RNAi) therapy has emerged as a promising approach for the treatment of various diseases, including hATTR and acute hepatic porphyria. By harnessing the natural cellular machinery for gene silencing, RNAi allows for the specific inhibition of target genes involved in disease pathogenesis. In the context of OA, targeting key molecules such as matrix metalloproteinase-13 (MMP13), which plays a critical role in cartilage degradation, holds great therapeutic potential. RESULTS In this study, we developed an innovative therapeutic approach for OA using a combination of liposome-encapsulated siMMP13 and NG-Monomethyl-L-arginine Acetate (L-NMMA) to form an injectable hydrogel. The hydrogel served as a delivery vehicle for the siMMP13, allowing for sustained release and targeted delivery to the affected joint. Experiments conducted on destabilization of the medial meniscus (DMM) model mice demonstrated the therapeutic efficacy of this composite hydrogel. Treatment with the hydrogel significantly inhibited the degradation of cartilage matrix, as evidenced by histological analysis showing preserved cartilage structure and reduced loss of proteoglycans. Moreover, the hydrogel effectively suppressed intracellular ROS accumulation in chondrocytes, indicating its anti-oxidative properties. Furthermore, it attenuated chondrocyte apoptosis, as demonstrated by decreased levels of apoptotic markers. CONCLUSION In summary, the injectable hydrogel containing siMMP13, endowed with anti-ROS and anti-apoptotic properties, may represent an effective therapeutic strategy for osteoarthritis in the future.
Collapse
Affiliation(s)
- Zhongyin Ji
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Department of Orthopedics Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
| | - Xiaobin Ren
- School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, P.R. China
| | - Jiayan Jin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Xin Ye
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Hao Yu
- School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, P.R. China
| | - Wenhan Fang
- College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, P.R. China
| | - Hui Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Yihao Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Siyue Tao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Xiangxi Kong
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Jiao Cheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Zhi Shan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Jian Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Qingqing Yao
- School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, P.R. China.
| | - Fengdong Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China.
| | - Junhui Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China.
| |
Collapse
|
13
|
Weber P, Asadikorayem M, Surman F, Zenobi-Wong M. Zwitterionic polymer-dexamethasone conjugates penetrate and protect cartilage from inflammation. Mater Today Bio 2024; 26:101049. [PMID: 38654933 PMCID: PMC11035115 DOI: 10.1016/j.mtbio.2024.101049] [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: 01/26/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/26/2024] Open
Abstract
Improving the pharmacokinetics of intra-articularly injected therapeutics is a major challenge in treating joint disease. Small molecules and biologics are often cleared from the joint within hours, which greatly reduces their therapeutic efficacy. Furthermore, they are often injected at high doses, which can lead to local cytotoxicity and systemic side effects. In this study, we present modular polymer-drug conjugates of zwitterionic poly(carboxybetaine acrylamide) (pCBAA) and the anti-inflammatory glucocorticoid dexamethasone (DEX) to create cartilage-targeted carriers with slow-release kinetics. pCBAA polymers showed excellent cartilage penetration (full thickness in 1 h) and retention (>50 % after 2 weeks of washing). DEX was loaded onto the pCBAA polymer by employing two different DEX-bearing comonomers to produce pCBAA-co-DEX conjugates with different release kinetics. The slow-releasing conjugate showed zero-order release kinetics in PBS over 70 days. The conjugates elicited no oxidative stress on chondrocytes compared to dose-matched free DEX and protected bovine cartilage explants from the inflammatory response after treatment with IL-1β. By combining cartilage targeting and sustained drug release properties, the pCBAA-co-DEX conjugates solve many issues of today's intra-articular therapeutics, which could ultimately enable better long-term clinical outcomes with fewer side effects.
Collapse
Affiliation(s)
- Patrick Weber
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Maryam Asadikorayem
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - František Surman
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| |
Collapse
|
14
|
Gonzales G, Hoque J, Gilpin A, Maity B, Zauscher S, Varghese S. Branched poly-l-lysine for cartilage penetrating carriers. Bioeng Transl Med 2024; 9:e10612. [PMID: 38818117 PMCID: PMC11135149 DOI: 10.1002/btm2.10612] [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: 06/27/2023] [Revised: 09/22/2023] [Accepted: 10/01/2023] [Indexed: 06/01/2024] Open
Abstract
Joint diseases, such as osteoarthritis, often require delivery of drugs to chondrocytes residing within the cartilage. However, intra-articular delivery of drugs to cartilage remains a challenge due to their rapid clearance within the joint. This problem is further exacerbated by the dense and negatively charged cartilage extracellular matrix (ECM). Cationic nanocarriers that form reversible electrostatic interactions with the anionic ECM can be an effective approach to overcome the electrostatic barrier presented by cartilage tissue. For an effective therapeutic outcome, the nanocarriers need to penetrate, accumulate, and be retained within the cartilage tissue. Nanocarriers that adhere quickly to cartilage tissue after intra-articular administration, transport through cartilage, and remain within its full thickness are crucial to the therapeutic outcome. To this end, we used ring-opening polymerization to synthesize branched poly(l-lysine) (BPL) cationic nanocarriers with varying numbers of poly(lysine) branches, surface charge, and functional groups, while maintaining similar hydrodynamic diameters. Our results show that the multivalent BPL molecules, including those that are highly branched (i.e., generation two), can readily adhere and transport through the full thickness of cartilage, healthy and degenerated, with prolonged intra-cartilage retention. Intra-articular injection of the BPL molecules in mouse knee joint explants and rat knee joints showed their localization and retention. In summary, this study describes an approach to design nanocarriers with varying charge and abundant functional groups while maintaining similar hydrodynamic diameters to aid the delivery of macromolecules to negatively charged tissues.
Collapse
Affiliation(s)
- Gavin Gonzales
- Department of Biomedical EngineeringDuke UniversityDurhamNorth CarolinaUSA
| | - Jiaul Hoque
- Department of Orthopedic SurgeryDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Anna Gilpin
- Department of Biomedical EngineeringDuke UniversityDurhamNorth CarolinaUSA
| | - Biswanath Maity
- Department of Orthopedic SurgeryDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Stefan Zauscher
- Department of Mechanical Engineering and Materials ScienceDuke UniversityDurhamNorth CarolinaUSA
| | - Shyni Varghese
- Department of Biomedical EngineeringDuke UniversityDurhamNorth CarolinaUSA
- Department of Orthopedic SurgeryDuke University School of MedicineDurhamNorth CarolinaUSA
- Department of Mechanical Engineering and Materials ScienceDuke UniversityDurhamNorth CarolinaUSA
| |
Collapse
|
15
|
Xiong W, Han Z, Ding S, Wang H, Du Y, Cui W, Zhang M. In Situ Remodeling of Efferocytosis via Lesion-Localized Microspheres to Reverse Cartilage Senescence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400345. [PMID: 38477444 PMCID: PMC11109622 DOI: 10.1002/advs.202400345] [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: 01/30/2024] [Revised: 02/19/2024] [Indexed: 03/14/2024]
Abstract
Efferocytosis, an intrinsic regulatory mechanism to eliminate apoptotic cells, will be suppressed due to the delayed apoptosis process in aging-related diseases, such as osteoarthritis (OA). In this study, cartilage lesion-localized hydrogel microspheres are developed to remodel the in situ efferocytosis to reverse cartilage senescence and recruit endogenous stem cells to accelerate cartilage repair. Specifically, aldehyde- and methacrylic anhydride (MA)-modified hyaluronic acid hydrogel microspheres (AHM), loaded with pro-apoptotic liposomes (liposomes encapsulating ABT263, A-Lipo) and PDGF-BB, namely A-Lipo/PAHM, are prepared by microfluidic and photo-cross-linking techniques. By a degraded porcine cartilage explant OA model, the in situ cartilage lesion location experiment illustrated that aldehyde-functionalized microspheres promote affinity for degraded cartilage. In vitro data showed that A-Lipo induced apoptosis of senescent chondrocytes (Sn-chondrocytes), which can then be phagocytosed by the efferocytosis of macrophages, and remodeling efferocytosis facilitated the protection of normal chondrocytes and maintained the chondrogenic differentiation capacity of MSCs. In vivo experiments confirmed that hydrogel microspheres localized to cartilage lesion reversed cartilage senescence and promoted cartilage repair in OA. It is believed this in situ efferocytosis remodeling strategy can be of great significance for tissue regeneration in aging-related diseases.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730P. R. China
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Zeyu Han
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730P. R. China
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Sheng‐Long Ding
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730P. R. China
| | - Haoran Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Yawei Du
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Ming‐Zhu Zhang
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730P. R. China
| |
Collapse
|
16
|
Mohseni M, Shokrollahi P, Barzin J. Gelatin/O-carboxymethyl chitosan injectable self-healing hydrogels for ibuprofen and naproxen dual release. Int J Biol Macromol 2024; 263:130266. [PMID: 38368982 DOI: 10.1016/j.ijbiomac.2024.130266] [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: 08/16/2023] [Revised: 02/11/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Recently, a significantly greater clinical benefit has been reported with a combination of glucosamine sulfate and nonsteroidal anti-inflammatory drugs (NSAIDs) compared to either treatment alone for the growing osteoarthritis (OA) disease. So, this study introduces hydrogels using O-carboxymethyl chitosan (O-CMC, structurally akin glucosamine glycan), and Gelatin type A (GA) in a 1:2 ratio with β-glycerophosphate (βGPh) at varying percentages (5 %, 12.5 %, and 15 %). We show that hydrogel properties, adaptable for drug delivery or tissue engineering, can be fine-tuned based on OCMC:βGPh ratio. CMC/GA/βGPh-12.5 exhibited a swelling rate of 189 %, compressive stress of 164 kPa, and compressive modulus of 3.4 kPa. The self-healing hydrogel also exhibited excellent injectability through a 21-gauge needle, requiring only 5 N of force. Ibuprofen and Naproxen release from CMC/GA/βGPh-12.5 and CMC/GA/βGPh-15 of designed dimensions (bi-layer structures of different diameter and height) were measured, and drug release kinetics were estimated using mathematical equations (MATLAB and polyfit program). CMC/GA/βGPh-12.5 demonstrated significant antibacterial effects against E. coli and S. aureus, a high cell survival rate of 89 % against L929 fibroblasts, and strong cell adhesion, all indicating biocompatibility. These findings underscore potential of these hydrogels as promising candidates for treating inflammatory diseases such as osteoarthritis.
Collapse
Affiliation(s)
- Mahshad Mohseni
- Department of Biomaterials, Faculty of Science, Iran Polymer and Petrochemical Institute (IPPI), Tehran 14975-112, Iran
| | - Parvin Shokrollahi
- Department of Biomaterials, Faculty of Science, Iran Polymer and Petrochemical Institute (IPPI), Tehran 14975-112, Iran.
| | - Jalal Barzin
- Department of Biomaterials, Faculty of Science, Iran Polymer and Petrochemical Institute (IPPI), Tehran 14975-112, Iran
| |
Collapse
|
17
|
Lu G, Yang C, Chu K, Zhu Y, Huang S, Zheng J, Jia H, Li X, Ban J. Implantable celecoxib nanofibers made by electrospinning: fabrication and characterization. Nanomedicine (Lond) 2024; 19:657-669. [PMID: 38305028 DOI: 10.2217/nnm-2023-0314] [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] [Indexed: 02/03/2024] Open
Abstract
Background: Osteoarthritis causes tremendous damage to the joints, reducing the quality of life and imposing significant financial burden. An implantable drug-delivery system can improve the symptomatic manifestations with low doses and frequencies. However, the free drug has short retention in the joint cavity. Materials & methods: This study used electrostatic spinning technology to create an implantable drug-delivery system loaded with celecoxib (celecoxib nanofibers [Cel-NFs]) to improve retention and bioavailability. Results: Cel-NFs exhibited good formability, hydrophilicity and tensile properties. Cel-NFs were able to continuously release drugs for 2 weeks and increase the uptake capacity of Raw 264.7 cells, ultimately ameliorating symptoms in osteoarthritis rats. Conclusion: These results suggest that Cel-NFs can effectively ameliorate cartilage damage, reduce joint pain and alleviate osteoarthritis progression.
Collapse
Affiliation(s)
- Geng Lu
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Chuangzan Yang
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Kedi Chu
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yi Zhu
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Sa Huang
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Juying Zheng
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Huanhuan Jia
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery Sysytems, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xiaofang Li
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, 510663, China
| | - Junfeng Ban
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, 510663, China
| |
Collapse
|
18
|
Hsu YY, Hwang SW, Chen SJ, Alsberg E, Liu AP. Development of mechanosensitive synthetic cells for biomedical applications. SLAS Technol 2024; 29:100095. [PMID: 37385542 DOI: 10.1016/j.slast.2023.06.004] [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: 04/21/2023] [Revised: 06/08/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023]
Abstract
The ability of cells to sense and respond to their physical environment plays a fundamental role in a broad spectrum of biological processes. As one of the most essential molecular force sensors and transducers found in cell membranes, mechanosensitive (MS) ion channels can convert mechanical inputs into biochemical or electrical signals to mediate a variety of sensations. The bottom-up construction of cell-sized compartments displaying cell-like organization, behaviors, and complexity, also known as synthetic cells, has gained popularity as an experimental platform to characterize biological functions in isolation. By reconstituting MS channels in the synthetic lipid bilayers, we envision using mechanosensitive synthetic cells for several medical applications. Here, we describe three different concepts for using ultrasound, shear stress, and compressive stress as mechanical stimuli to activate drug release from mechanosensitive synthetic cells for disease treatments.
Collapse
Affiliation(s)
- Yen-Yu Hsu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sung-Won Hwang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Samuel J Chen
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eben Alsberg
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60612, USA; Departments of Mechanical & Industrial Engineering, Orthopaedic Surgery, and Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Allen P Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
19
|
Wang M, Jin Z, Huang H, Cheng X, Zhang Q, Tang Y, Zhu X, Zong Z, Li H, Ning Z. Neutrophil hitchhiking: Riding the drug delivery wave to treat diseases. Drug Dev Res 2024; 85:e22169. [PMID: 38477422 DOI: 10.1002/ddr.22169] [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: 10/08/2023] [Revised: 02/06/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024]
Abstract
Neutrophils are a crucial component of the innate immune system and play a pivotal role in various physiological processes. From a physical perspective, hitchhiking is considered a phenomenon of efficient transportation. The combination of neutrophils and hitchhikers has given rise to effective delivery systems both in vivo and in vitro, thus neutrophils hitchhiking become a novel approach to disease treatment. This article provides an overview of the innovative and feasible application of neutrophils as drug carriers. It explores the mechanisms underlying neutrophil function, elucidates the mechanism of drug delivery mediated by neutrophil-hitchhiking, and discusses the potential applications of this strategy in the treatment of cancer, immune diseases, inflammatory diseases, and other medical conditions.
Collapse
Affiliation(s)
- Menghui Wang
- Department of Day Ward, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang University, Nanchang, Jiangxi Province, China
- HuanKui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhenhua Jin
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Haoyu Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Xifu Cheng
- Department of Day Ward, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Qin Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Ying Tang
- Department of Day Ward, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiaoping Zhu
- Department of Day Ward, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Hui Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhikun Ning
- Department of Day Ward, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
20
|
Svensson E, von Mentzer U, Stubelius A. Achieving Precision Healthcare through Nanomedicine and Enhanced Model Systems. ACS MATERIALS AU 2024; 4:162-173. [PMID: 38496040 PMCID: PMC10941278 DOI: 10.1021/acsmaterialsau.3c00073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/17/2023] [Accepted: 11/28/2023] [Indexed: 03/19/2024]
Abstract
The ability to customize medical choices according to an individual's genetic makeup and biomarker patterns marks a significant advancement toward overall improved healthcare for both individuals and society at large. By transitioning from the conventional one-size-fits-all approach to tailored treatments that can account for predispositions of different patient populations, nanomedicines can be customized to target the specific molecular underpinnings of a patient's disease, thus mitigating the risk of collateral damage. However, for these systems to reach their full potential, our understanding of how nano-based therapeutics behave within the intricate human body is necessary. Effective drug administration to the targeted organ or pathological niche is dictated by properties such as nanocarrier (NC) size, shape, and targeting abilities, where understanding how NCs change their properties when they encounter biomolecules and phenomena such as shear stress in flow remains a major challenge. This Review specifically focuses on vessel-on-a-chip technology that can provide increased understanding of NC behavior in blood and summarizes the specialized environment of the joint to showcase advanced tissue models as approaches to address translational challenges. Compared to conventional cell studies or animal models, these advanced models can integrate patient material for full customization. Combining such models with nanomedicine can contribute to making personalized medicine achievable.
Collapse
Affiliation(s)
| | | | - Alexandra Stubelius
- Division of Chemical Biology,
Department of Life Sciences, Chalmers University
of Technology, Gothenburg 412 96, Sweden
| |
Collapse
|
21
|
Nhan J, Strebel N, Virah Sawmy K, Yin J, St-Pierre JP. Characterization of Calcium- and Strontium-Polyphosphate Particles Toward Drug Delivery into Articular Cartilage. Macromol Biosci 2024; 24:e2300345. [PMID: 37777870 DOI: 10.1002/mabi.202300345] [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: 07/27/2023] [Revised: 09/21/2023] [Indexed: 10/02/2023]
Abstract
Drug delivery into articular cartilage poses many challenges due in part to its lack of vasculature. While intra-articular injections are effective for the local administration of drugs, small molecules are rapidly cleared from the synovial fluid. As such, there is a need to develop effective drug delivery strategies to improve the residence times of bioactive molecules in the joint and elicit a sustained therapeutic effect. In this study, calcium- and strontium-polyphosphate particles are synthesized and characterized as potential drug carriers into articular cartilage. Physicochemical characterization reveals that the particles exhibit a spherical morphology, have a negative zeta potential, and are nanoscale in size. Biological characterization in chondrocytes confirms cellular uptake of the particles and demonstrates both size and concentration-dependent cytotoxicity at high concentrations. Furthermore, treatment of chondrocytes with these particles results in a reduction in cell proliferation and metabolic activity, confirming biological effects. Finally, incubation with cartilage tissue explants suggests successful uptake, despite the particles exhibiting a negative surface charge. Therefore, from the results of this study, these polyphosphate-based particles have potential as a drug carrier into articular cartilage and warrant further development.
Collapse
Affiliation(s)
- Jordan Nhan
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Nicolas Strebel
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Khushnouma Virah Sawmy
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Jordan Yin
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Jean-Philippe St-Pierre
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| |
Collapse
|
22
|
Deng R, Zhao R, Zhang Z, Chen Y, Yang M, Lin Y, Ye J, Li N, Qin H, Yan X, Shi J, Yuan F, Song S, Xu Z, Song Y, Fu J, Xu B, Nie G, Yu JK. Chondrocyte membrane-coated nanoparticles promote drug retention and halt cartilage damage in rat and canine osteoarthritis. Sci Transl Med 2024; 16:eadh9751. [PMID: 38381849 DOI: 10.1126/scitranslmed.adh9751] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024]
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by progressive degeneration of articular cartilage. A challenge in the development of disease-modifying drugs is effective delivery to chondrocytes. The unique structure of the joint promotes rapid clearance of drugs through synovial fluid, and the dense and avascular cartilage extracellular matrix (ECM) limits drug penetration. Here, we show that poly(lactide-co-glycolic acid) nanoparticles coated in chondrocyte membranes (CM-NPs) were preferentially taken up by rat chondrocytes ex vivo compared with uncoated nanoparticles. Internalization of the CM-NPs was mediated primarily by E-cadherin, clathrin-mediated endocytosis, and micropinocytosis. These CM-NPs adhered to the cartilage ECM in rat knee joints in vivo and penetrated deeply into the cartilage matrix with a residence time of more than 34 days. Simulated synovial fluid clearance studies showed that CM-NPs loaded with a Wnt pathway inhibitor, adavivint (CM-NPs-Ada), delayed the catabolic metabolism of rat and human chondrocytes and cartilage explants under inflammatory conditions. In a surgical model of rat OA, drug-loaded CM-NPs effectively restored gait, attenuated periarticular bone remodeling, and provided chondroprotection against cartilage degeneration. OA progression was also mitigated by CM-NPs-Ada in a canine model of anterior cruciate ligament transection. These results demonstrate the feasibility of using chondrocyte membrane-coated nanoparticles to improve the pharmacokinetics and efficacy of anti-OA drugs.
Collapse
Affiliation(s)
- Ronghui Deng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zining Zhang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yang Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Meng Yang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yixuan Lin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jing Ye
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Nan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hao Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xin Yan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Fuzhen Yuan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Shitang Song
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Zijie Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yifan Song
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Jiangnan Fu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Bingbing Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jia-Kuo Yu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
- Orthopedic Sports Medicine Center, Beijing Tsinghua Changgung Hospital, Affiliated Hospital of Tsinghua University, Beijing 102218, P. R. China
| |
Collapse
|
23
|
Liu Y, Yao J, Deng G, Zhong G, Zhao J, Lan Q, Meng J, Yu Y, Chen F. Microgel Encapsulated Nanoparticles for Intra-articular Disulfiram Delivery to Treat Osteoarthritis. Mol Pharm 2024; 21:87-101. [PMID: 38100656 DOI: 10.1021/acs.molpharmaceut.3c00462] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Osteoarthritis (OA) affects numerous patients worldwide, and there are no approved disease-modifying drugs. Repurposing FDA-approved small molecular drugs could be a promising alternative strategy to treat OA. Disulfiram (DSF), a clinically approved drug for treatment of alcoholism, inhibits inflammasome activation and exhibits a protective role in interleukin-1β-induced cardiac injury. However, its efficacy in treating OA remains to be explored due to its poor water solubility and stability, which limit its use in OA treatment. Here, the anti-inflammatory effect of DSF is evaluated in vitro, and a double-layer encapsulation approach is developed for intra-articular delivery of DSF for OA treatment in vivo. DSF is loaded into poly(lactic-co-glycolic acid)-based nanoparticles and encapsulated in gelatin methacrylate microgels through a microfluidic device. Results show that DSF effectively inhibits the expression of key inflammatory cytokines in OA chondrocytes, and the double-layer encapsulation approach reduces the burst release of DSF and prolongs its retention time in the in vitro study. Sustained release of DSF from microgels mitigates cartilage inflammation and subchondral bone erosion in a monoiodoacetate-induced rat OA model. This work demonstrates the potential of repurposing FDA-approved drugs for OA treatment and provides a promising platform for intra-articular delivery of small molecules for superior therapeutic effect.
Collapse
Affiliation(s)
- Yisi Liu
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Bone and Joint Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jun Yao
- Department of Bone and Joint Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Guotao Deng
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Gang Zhong
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jianping Zhao
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qiumei Lan
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jinzhi Meng
- Department of Bone and Joint Surgery & Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yin Yu
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fei Chen
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
24
|
Scanu A, Luisetto R, Pavan M, Guarise C, Beninatto R, Giraudo C, Galuppini F, Lazzarin V, Guzzardo V, Pennelli G, Galesso D, Masiero S. Effect of intra-articular injection of a hyaluronic acid-alendronate conjugate on post-traumatic osteoarthritis induced by destabilization of the medial meniscus in rats. Sci Rep 2023; 13:20692. [PMID: 38001135 PMCID: PMC10673944 DOI: 10.1038/s41598-023-46965-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by pain and cartilage damage. Intra-articular (i.a) viscosupplementation with hyaluronic acid (HA) is frequently used for the management of OA. Preclinical studies have reported that bisphosphonates (BPs) may have a therapeutic potential to slow down or reverse the progression of OA. Among these, alendronate (ALN) has demonstrated chondroprotective effects in both in vitro and vivo experiments. This study evaluated the effects of a novel alendronate-hyaluronic acid (ALN-HA) conjugate on an OA in vivo model induced by medial meniscus destabilization (DMM). DMM surgery was performed on the knees of Sprague Dawley rats that received, after four weeks, one intra-articular (i.a.) injection of: (1) ALN-HA; (2) HA; (3) sodium chloride (NaCl). Sham-operated rats were used as control. Allodynia was assessed by Von Frey test. Joint degeneration was evaluated eight weeks after treatment by micro-computed tomography (micro-CT), histology, and immunohistochemistry. Collagen cross-linked C-telopeptides (CTX-I and CTX-II) serum levels were determined by ELISA. Paw withdrawal threshold increased in ALN-HA group when compared to rats treated with NaCl or HA. Micro-CT did not show differences between ALN-HA, HA and NaCl groups. ALN-HA injection produced significant improvements in articular cartilage degeneration showing an OARSI score lower than those of HA and NaCl, and reduced matrix metalloproteinase (MMP)-13, MMP-3, interleukin-6, vascular endothelial growth factor and Caspase-3 expression. CTX-I was reduced after ALN-HA treatment when compared to NaCl. Our results indicate that i.a. use of ALN after conjugation with HA limits OA development and progression in the rat DMM model, and may lead to the development of novel therapeutic strategies in OA management.
Collapse
Affiliation(s)
- Anna Scanu
- Rehabilitation Unit, Department of Neuroscience-DNS, University of Padova, 35128, Padua, Italy
| | - Roberto Luisetto
- Department of Surgery, Oncology and Gastroenterology-DISCOG, University of Padova, 35128, Padua, Italy
| | - Mauro Pavan
- R&D-Discovery, Fidia Farmaceutici SpA, Via Ponte della Fabbrica, 3/a, 35031, Abano Terme, Italy.
| | - Cristian Guarise
- R&D-Discovery, Fidia Farmaceutici SpA, Via Ponte della Fabbrica, 3/a, 35031, Abano Terme, Italy
| | - Riccardo Beninatto
- R&D-Discovery, Fidia Farmaceutici SpA, Via Ponte della Fabbrica, 3/a, 35031, Abano Terme, Italy
| | - Chiara Giraudo
- Nuclear Medicine Unit, Department of Medicine-DIMED, Padova University Hospital, 35128, Padua, Italy
| | - Francesca Galuppini
- Surgical Pathology Unit, Department of Medicine-DIMED, University of Padova, 35128, Padua, Italy
| | - Vanni Lazzarin
- Surgical Pathology Unit, Department of Medicine-DIMED, University of Padova, 35128, Padua, Italy
| | - Vincenza Guzzardo
- Surgical Pathology Unit, Department of Medicine-DIMED, University of Padova, 35128, Padua, Italy
| | - Gianmaria Pennelli
- Surgical Pathology Unit, Department of Medicine-DIMED, University of Padova, 35128, Padua, Italy
| | - Devis Galesso
- R&D-Discovery, Fidia Farmaceutici SpA, Via Ponte della Fabbrica, 3/a, 35031, Abano Terme, Italy
| | - Stefano Masiero
- Rehabilitation Unit, Department of Neuroscience-DNS, University of Padova, 35128, Padua, Italy
| |
Collapse
|
25
|
Santos MM, Santos AM, Nascimento Júnior JAC, Andrade TDA, Rajkumar G, Frank LA, Serafini MR. The management of osteoarthritis symptomatology through nanotechnology: a patent review. J Microencapsul 2023; 40:475-490. [PMID: 37698545 DOI: 10.1080/02652048.2023.2258955] [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: 03/17/2023] [Accepted: 09/11/2023] [Indexed: 09/13/2023]
Abstract
Osteoarthritis is considered a degenerative joint disease that is characterised by inflammation, chronic pain, and functional limitation. The increasing development of nanotechnology in drug delivery systems has provided new ideas and methods for osteoarthritis therapy. This review aimed to evaluate patents that have developed innovations, therapeutic strategies, and alternatives using nanotechnology in osteoarthritis treatment. The results show patents deposited from 2015 to November 2021 in the online databases European Patent Office and World Intellectual Property Organisation. A total of 651 patents were identified for preliminary assessment and 16 were selected for full reading and discussion. The evaluated patents are focused on the intraarticular route, oral route, and topical route for osteoarthritis treatment. The intraarticular route presented a higher patent number, followed by the oral and topical routes, respectively. The development of new technologies allows us to envision a promising and positive future in osteoarthritis treatment.
Collapse
Affiliation(s)
| | | | | | | | - Gomathi Rajkumar
- Department of Botany, Sri Sarada College for Women (Autonomous), Affiliated to Periyar University, Salem, India
| | - Luiza Abrahão Frank
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Mairim Russo Serafini
- Postgraduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Brazil
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Brazil
| |
Collapse
|
26
|
Kotla NG, Langlois JB, Fisch A, Kramer I, Halleux C. Surface modified cationic PLGA microparticles as long-acting injectable carriers for intra-articular small molecule drug delivery. Eur J Pharm Biopharm 2023; 193:S0939-6411(23)00286-2. [PMID: 39491137 DOI: 10.1016/j.ejpb.2023.10.020] [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: 09/18/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024]
Abstract
Controlled local delivery of therapeutics (small molecule drug crystals or biologics) for knee-associated diseases such as osteoarthritis necessitates patient compliance, ensuring that the injected depot does not trigger local tissue inflammation and immune responses. A local drug delivery strategy that releases drug at a controlled rate while ensuring minimal tolerability issues at the injection site would be an appealing paradigm in intra-articular (IA) therapies. Herein, we report the formulation development and characterization of surface modified PLGA microparticles (MPs) through the surface integration of a cationic lipid, DOTAP (1,2-Dioleoyl-3-trimethylammonium propane). Following IA administration, these particles are able to interact with anionic synovial fluid glycosaminoglycans (GAGs) to form an in-situ surface coating in the knee joint, thereby reducing the depot-associated local inflammatory response. The formulated microparticles were about 10-40 µm in size range, with a +19 to +33 mV overall surface charge after DOTAP lipid surface integration. These particles showed preferential surface adhesion with endogenous anionic GAGs (e.g., hyaluronic acid) due to electrostatic interactions in vitro, and approximately 65% of the model drug triamcinolone acetonide (TCA) was released after 10 weeks in simulated synovial fluid. The uncoated and DOTAP-coated PLGA microparticles had no effect on mouse osteoblast MC3T3 cell viability and human macrophage inflammatory response. Further, DOTAP-coated particles showed a marginal decrease in pro-inflammatory cytokines in naïve rats following knee injection. Together, the results suggest that surface-modified PLGA particles may have promising potential as delivery carriers for long-acting injectables.
Collapse
Affiliation(s)
- Niranjan G Kotla
- Diseases of Aging and Regenerative Medicine, Novartis Institute for BioMedical Research (NIBR), Novartis Pharma AG, Basel 4056, Switzerland.
| | - Jean-Baptiste Langlois
- Diseases of Aging and Regenerative Medicine, Novartis Institute for BioMedical Research (NIBR), Novartis Pharma AG, Basel 4056, Switzerland
| | - Andreas Fisch
- Technical Research and Development (TRD), Novartis Pharma AG, Basel 4056, Switzerland
| | - Ina Kramer
- Diseases of Aging and Regenerative Medicine, Novartis Institute for BioMedical Research (NIBR), Novartis Pharma AG, Basel 4056, Switzerland
| | - Christine Halleux
- Diseases of Aging and Regenerative Medicine, Novartis Institute for BioMedical Research (NIBR), Novartis Pharma AG, Basel 4056, Switzerland
| |
Collapse
|
27
|
Zhao T, Li X, Li H, Deng H, Li J, Yang Z, He S, Jiang S, Sui X, Guo Q, Liu S. Advancing drug delivery to articular cartilage: From single to multiple strategies. Acta Pharm Sin B 2023; 13:4127-4148. [PMID: 37799383 PMCID: PMC10547919 DOI: 10.1016/j.apsb.2022.11.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/09/2022] [Accepted: 10/28/2022] [Indexed: 11/27/2022] Open
Abstract
Articular cartilage (AC) injuries often lead to cartilage degeneration and may ultimately result in osteoarthritis (OA) due to the limited self-repair ability. To date, numerous intra-articular delivery systems carrying various therapeutic agents have been developed to improve therapeutic localization and retention, optimize controlled drug release profiles and target different pathological processes. Due to the complex and multifactorial characteristics of cartilage injury pathology and heterogeneity of the cartilage structure deposited within a dense matrix, delivery systems loaded with a single therapeutic agent are hindered from reaching multiple targets in a spatiotemporal matched manner and thus fail to mimic the natural processes of biosynthesis, compromising the goal of full cartilage regeneration. Emerging evidence highlights the importance of sequential delivery strategies targeting multiple pathological processes. In this review, we first summarize the current status and progress achieved in single-drug delivery strategies for the treatment of AC diseases. Subsequently, we focus mainly on advances in multiple drug delivery applications, including sequential release formulations targeting various pathological processes, synergistic targeting of the same pathological process, the spatial distribution in multiple tissues, and heterogeneous regeneration. We hope that this review will inspire the rational design of intra-articular drug delivery systems (DDSs) in the future.
Collapse
Affiliation(s)
- Tianyuan Zhao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, 999077, Hong Kong, China
| | - Hao Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Haoyuan Deng
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Jianwei Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Zhen Yang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing 100044, China
| | - Songlin He
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuangpeng Jiang
- Department of Joint Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
| | - Quanyi Guo
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
28
|
Gong J, Nhan J, St-Pierre JP, Gillies ER. Designing polymers for cartilage uptake: effects of architecture and molar mass. J Mater Chem B 2023; 11:8804-8816. [PMID: 37668597 DOI: 10.1039/d3tb01417g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Osteoarthritis (OA) is a progressive disease, involving the progressive breakdown of cartilage, as well as changes to the synovium and bone. There are currently no disease-modifying treatments available clinically. An increasing understanding of the disease pathophysiology is leading to new potential therapeutics, but improved approaches are needed to deliver these drugs, particularly to cartilage tissue, which is avascular and contains a dense matrix of collagens and negatively charged aggrecan proteoglycans. Cationic delivery vehicles have been shown to effectively penetrate cartilage, but these studies have thus far largely focused on proteins or nanoparticles, and the effects of macromolecular architectures have not yet been explored. Described here is the synthesis of a small library of polycations composed of N-(2-hydroxypropyl)methacrylamide (HPMA) and N-(3-aminopropyl)methacrylamide (APMA) with linear, 4-arm, or 8-arm structures and varying degrees of polymerization (DP) by reversible addition fragmentation chain-transfer (RAFT) polymerization. Uptake and retention of the polycations in bovine articular cartilage was assessed. While all polycations penetrated cartilage, uptake and retention generally increased with DP before decreasing for the highest DP. In addition, uptake and retention were higher for the linear polycations compared to the 4-arm and 8-arm polycations. In general, the polycations were well tolerated by bovine chondrocytes, but the highest DP polycations imparted greater cytotoxicity. Overall, this study reveals that linear polymer architectures may be more favorable for binding to the cartilage matrix and that the DP can be tuned to maximize uptake while minimizing cytotoxicity.
Collapse
Affiliation(s)
- Jue Gong
- Department of Chemistry, The University of Western Ontario, 1151 Richmond St., London, Ontario, N6A 5B7, Canada.
| | - Jordan Nhan
- Department of Chemical and Biological Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada.
| | - Jean-Philippe St-Pierre
- Department of Chemical and Biological Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada.
| | - Elizabeth R Gillies
- Department of Chemistry, The University of Western Ontario, 1151 Richmond St., London, Ontario, N6A 5B7, Canada.
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, 1151 Richmond St., London, Ontario, N6A 5B9, Canada
| |
Collapse
|
29
|
Porcello A, Gonzalez-Fernandez P, Jeannerat A, Peneveyre C, Abdel-Sayed P, Scaletta C, Raffoul W, Hirt-Burri N, Applegate LA, Allémann E, Laurent A, Jordan O. Thermo-Responsive Hyaluronan-Based Hydrogels Combined with Allogeneic Cytotherapeutics for the Treatment of Osteoarthritis. Pharmaceutics 2023; 15:pharmaceutics15051528. [PMID: 37242774 DOI: 10.3390/pharmaceutics15051528] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/14/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Thermo-responsive hyaluronan-based hydrogels and FE002 human primary chondroprogenitor cell sources have both been previously proposed as modern therapeutic options for the management of osteoarthritis (OA). For the translational development of a potential orthopedic combination product based on both technologies, respective technical aspects required further optimization phases (e.g., hydrogel synthesis upscaling and sterilization, FE002 cytotherapeutic material stabilization). The first aim of the present study was to perform multi-step in vitro characterization of several combination product formulas throughout the established and the optimized manufacturing workflows, with a strong focus set on critical functional parameters. The second aim of the present study was to assess the applicability and the efficacy of the considered combination product prototypes in a rodent model of knee OA. Specific characterization results (i.e., spectral analysis, rheology, tribology, injectability, degradation assays, in vitro biocompatibility) of hyaluronan-based hydrogels modified with sulfo-dibenzocyclooctyne-PEG4-amine linkers and poly(N-isopropylacrylamide) (HA-L-PNIPAM) containing lyophilized FE002 human chondroprogenitors confirmed the suitability of the considered combination product components. Specifically, significantly enhanced resistance toward oxidative and enzymatic degradation was shown in vitro for the studied injectable combination product prototypes. Furthermore, extensive multi-parametric (i.e., tomography, histology, scoring) in vivo investigation of the effects of FE002 cell-laden HA-L-PNIPAM hydrogels in a rodent model revealed no general or local iatrogenic adverse effects, whereas it did reveal some beneficial trends against the development of knee OA. Overall, the present study addressed key aspects of the preclinical development process for novel biologically-based orthopedic combination products and shall serve as a robust methodological basis for further translational investigation and clinical work.
Collapse
Affiliation(s)
- Alexandre Porcello
- School of Pharmaceutical Sciences, University of Geneva, CH-1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, Switzerland
| | - Paula Gonzalez-Fernandez
- School of Pharmaceutical Sciences, University of Geneva, CH-1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, Switzerland
| | - Annick Jeannerat
- Preclinical Research Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland
| | - Cédric Peneveyre
- Preclinical Research Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland
- STI School of Engineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Wassim Raffoul
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
- Plastic, Reconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, CH-1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, Switzerland
| | - Alexis Laurent
- Preclinical Research Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, CH-1206 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CH-1206 Geneva, Switzerland
| |
Collapse
|
30
|
Li X, Li X, Yang J, Du Y, Chen L, Zhao G, Ye T, Zhu Y, Xu X, Deng L, Cui W. In Situ Sustained Macrophage-Targeted Nanomicelle-Hydrogel Microspheres for Inhibiting Osteoarthritis. RESEARCH (WASHINGTON, D.C.) 2023; 6:0131. [PMID: 37223475 PMCID: PMC10202383 DOI: 10.34133/research.0131] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 04/10/2023] [Indexed: 05/25/2023]
Abstract
There are still challenges in applying drug nanocarriers for in situ sustained macrophage targeting and regulation, due to the rapid clearance of nanocarriers and burst drug release in vivo. Herein, a nanomicelle-hydrogel microsphere, characterized by its macrophage-targeted nanosized secondary structure that allows it to accurately bind to M1 macrophages through active endocytosis, is employed for in situ sustained macrophage targeting and regulation, and addresses the insufficient osteoarthritis therapeutic efficacy caused by rapid clearance of drug nanocarriers. The 3-dimensional structure of a microsphere can prevent the rapid escape and clearance of a nanomicelle, thus keeping it in joints, while the ligand-guided secondary structure can carry drugs to accurately target and enter M1 macrophages, and release drugs via the transition from hydrophobicity to hydrophilicity of nanomicelles under inflammatory stimulation inside the macrophages. The experiments show that the nanomicelle-hydrogel microsphere can in situ sustainably target and regulate M1 macrophages for more than 14 days in joints, and attenuate local "cytokine storm" by continuous M1 macrophage apoptosis promotion and polarization inhibition. This micro/nano-hydrogel system shows excellent ability to sustainably target and regulate macrophage, realizes the improvement of drug utilization and efficacy inside the macrophage, and thereby can be a potential platform for treating macrophage-related diseases.
Collapse
Affiliation(s)
| | | | | | - Yawei Du
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Liang Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Gang Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Tingjun Ye
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Yuan Zhu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Xiangyang Xu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| |
Collapse
|
31
|
Wen J, Li H, Dai H, Hua S, Long X, Li H, Ivanovski S, Xu C. Intra-articular nanoparticles based therapies for osteoarthritis and rheumatoid arthritis management. Mater Today Bio 2023; 19:100597. [PMID: 36910270 PMCID: PMC9999238 DOI: 10.1016/j.mtbio.2023.100597] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/19/2023] [Accepted: 02/24/2023] [Indexed: 02/27/2023] Open
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are chronic and progressive inflammatory joint diseases that affect a large population worldwide. Intra-articular administration of various therapeutics is applied to alleviate pain, prevent further progression, and promote cartilage regeneration and bone remodeling in both OA and RA. However, the effectiveness of intra-articular injection with traditional drugs is uncertain and controversial due to issues such as rapid drug clearance and the barrier afforded by the dense structure of cartilage. Nanoparticles can improve the efficacy of intra-articular injection by facilitating controlled drug release, prolonged retention time, and enhanced penetration into joint tissue. This review systematically summarizes nanoparticle-based therapies for OA and RA management. Firstly, we explore the interaction between nanoparticles and joints, including articular fluids and cells. This is followed by a comprehensive analysis of current nanoparticles designed for OA/RA, divided into two categories based on therapeutic mechanisms: direct therapeutic nanoparticles and nanoparticles-based drug delivery systems. We highlight nanoparticle design for tissue/cell targeting and controlled drug release before discussing challenges of nanoparticle-based therapies for efficient OA and RA treatment and their future clinical translation. We anticipate that rationally designed local injection of nanoparticles will be more effective, convenient, and safer than the current therapeutic approach.
Collapse
Affiliation(s)
- Juan Wen
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
| | - Huimin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Huan Dai
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
| | - Shu Hua
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
| | - Xing Long
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Huang Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210009, China
| | - Sašo Ivanovski
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Corresponding author. School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia.
| | - Chun Xu
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
- Corresponding author. School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia.
| |
Collapse
|
32
|
Liu S, Zhang C, Zhou Y, Zhang F, Duan X, Liu Y, Zhao X, Liu J, Shuai X, Wang J, Cao Z. MRI-visible mesoporous polydopamine nanoparticles with enhanced antioxidant capacity for osteoarthritis therapy. Biomaterials 2023; 295:122030. [PMID: 36758340 DOI: 10.1016/j.biomaterials.2023.122030] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 01/21/2023] [Accepted: 01/26/2023] [Indexed: 02/01/2023]
Abstract
Since the progression of osteoarthritis (OA) is closely associated with synovitis and cartilage destruction, the inhibition of inflammatory responses in synovial macrophages and reactive oxygen species (ROS) induced apoptosis in chondrocytes is crucial for OA amelioration. However, most of the current anti-inflammatory and antioxidant drugs are small molecules apt to be eliminated in vivo. Herein, mesoporous polydopamine nanoparticles (DAMM NPs) doped with arginine and manganese (Mn) ions were prepared to load dexamethasone (DEX) for OA intervention. A series of in vitro studies showed that the sustained release of DEX from DAMM NPs suppressed synovial inflammation and simultaneously inhibited toll-like receptor 3 (TLR-3) production in chondrocytes, contributing to prevention of chondrocyte apoptosis through the inflammatory factor-dependent TLR-3/NF-κB signaling pathway via modulation of macrophage-chondrocyte crosstalk. In addition, DAMM NPs exerted a predominant role in removal of ROS generated in chondrocytes. Therefore, the DEX-loaded DAMM NPs significantly attenuated OA development in mice model. Importantly, the T1-T2 magnetic contrast capabilities of DAMM NPs allowed an MRI-trackable delivery, manifesting a distinct feature widely regarded to boost the potential of nanomedicines for clinical applications. Together, our developed antioxidant-enhanced DAMM NPs with MRI-visible signals may serve as a novel multifunctional nanocarriers for prevention of OA progression.
Collapse
Affiliation(s)
- Sitong Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Chen Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yuanyuan Zhou
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Fang Zhang
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Xiaohui Duan
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Yang Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Xibang Zhao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jiali Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| | - Zhong Cao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| |
Collapse
|
33
|
Zhang S, Wang L, Kang Y, Wu J, Zhang Z. Nanomaterial-based Reactive Oxygen Species Scavengers for Osteoarthritis Therapy. Acta Biomater 2023; 162:1-19. [PMID: 36967052 DOI: 10.1016/j.actbio.2023.03.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/17/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
Reactive oxygen species (ROS) play distinct but important roles in physiological and pathophysiological processes. Recent studies on osteoarthritis (OA) have suggested that ROS plays a crucial role in its development and progression, serving as key mediators in the degradation of the extracellular matrix, mitochondrial dysfunction, chondrocyte apoptosis, and OA progression. With the continuous development of nanomaterial technology, the ROS-scavenging ability and antioxidant effects of nanomaterials are being explored, with promising results already achieved in OA treatment. However, current research on nanomaterials as ROS scavengers for OA is relatively non-uniform and includes both inorganic and functionalized organic nanomaterials. Although the therapeutic efficacy of nanomaterials has been reported to be conclusive, there is still no uniformity in the timing and potential of their use in clinical practice. This paper reviews the nanomaterials currently used as ROS scavengers for OA treatment, along with their mechanisms of action, with the aim of providing a reference and direction for similar studies, and ultimately promoting the early clinical use of nanomaterials for OA treatment. STATEMENT OF SIGNIFICANCE: Reactive oxygen species (ROS) play an important role in the pathogenesis of osteoarthritis (OA). Nanomaterials serving as promising ROS scavengers have gained increasing attention in recent years. This review provides a comprehensive overview of ROS production and regulation, as well as their role in OA pathogenesis. Furthermore, this review highlights the applications of various types of nanomaterials as ROS scavengers in OA treatment and their mechanisms of action. Finally, the challenges and future prospects of nanomaterial-based ROS scavengers in OA therapy are discussed.
Collapse
|
34
|
Chu K, Zhu Y, Lu G, Huang S, Yang C, Zheng J, Chen J, Ban J, Jia H, Lu Z. Formation of Hydrophilic Nanofibers from Nanostructural Design in the Co-Encapsulation of Celecoxib through Electrospinning. Pharmaceutics 2023; 15:pharmaceutics15030730. [PMID: 36986590 PMCID: PMC10051909 DOI: 10.3390/pharmaceutics15030730] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
This study presents a method for a one-step co-encapsulation of PLGA nanoparticles in hydrophilic nanofibers. The aim is to effectively deliver the drug to the lesion site and achieve a longer release time. The celecoxib nanofiber membrane (Cel-NPs-NFs) was prepared by emulsion solvent evaporation and electrospinning with celecoxib as a model drug. By this method, nanodroplets of celecoxib PLGA are entrapped within polymer nanofibers during an electrospinning process. Moreover, Cel-NPs-NFs exhibited good mechanical strength and hydrophilicity, with a cumulative release of 67.74% for seven days, and the cell uptake at 0.5 h was 2.7 times higher than that of pure nanoparticles. Furthermore, pathological sections of the joint exhibited an apparent therapeutic effect on rat OA, and the drug was delivered effectively. According to the results, this solid matrix containing nanodroplets or nanoparticles could use hydrophilic materials as carriers to prolong drug release time.
Collapse
Affiliation(s)
- Kedi Chu
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yi Zhu
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Geng Lu
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Sa Huang
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou 510663, China
| | - Chuangzan Yang
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Juying Zheng
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Junming Chen
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Junfeng Ban
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Engineering Center of Topical Precision Drug Delivery System, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huanhuan Jia
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou 510663, China
- Correspondence: (H.J.); (Z.L.)
| | - Zhufen Lu
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Engineering Center of Topical Precision Drug Delivery System, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Correspondence: (H.J.); (Z.L.)
| |
Collapse
|
35
|
Dravid AA, Dhanabalan KM, Naskar S, Vashistha A, Agarwal S, Padhan B, Dewani M, Agarwal R. Sustained release resolvin D1 liposomes are effective in the treatment of osteoarthritis in obese mice. J Biomed Mater Res A 2023; 111:765-777. [PMID: 36773024 DOI: 10.1002/jbm.a.37512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/12/2023]
Abstract
Osteoarthritis (OA) is the most common joint disorder and currently affects >500 million patients worldwide, with ~60% of them also suffering from obesity. There is no drug approved for human use that changes the course of OA progression. OA is one of the most common comorbidities of obesity, and obesity-related OA (ObOA) is a serious health concern because it shows heightened severity of tissue damage and also predominantly affects the working population. Unresolved inflammation is a major driver of ObOA, thus, resolving disease-associated inflammation is a viable strategy to treat ObOA. Resolvins are highly potent molecules that play a role in the resolution of inflammation and promote tissue healing. However, small molecules (like Resolvin D1; RvD1) have to be administered frequently or prior to injury because they lose their in vivo activity rapidly either by lymphatic clearance, or oxidation-mediated deactivation. In this study, we have encapsulated RvD1 in liposomes and established its efficacy in the mouse model of ObOA at much lower dosages than freely administered RvD1. Liposomal RvD1 (lipo-RvD1) acted as a source of the RvD1 molecules for ~11 days in vitro in synovial fluid derived from patients. When administered prophylactically or therapeutically, lipo-RvD1 suppressed cartilage damage in male C57BL/6 mice compared to untreated and free RvD1 treatments. This efficacy was achieved by increasing the proportion of the proresolution M2 macrophages over proinflammatory M1 macrophages in the synovial membrane. These results show the potential of lipo-RvD1 as an anti-OA agent.
Collapse
|
36
|
He T, Zhang C, Colombani T, Bencherif SA, Porter RM, Bajpayee AG. Intra-articular kinetics of a cartilage targeting cationic PEGylated protein for applications in drug delivery. Osteoarthritis Cartilage 2023; 31:187-198. [PMID: 36241136 PMCID: PMC9892226 DOI: 10.1016/j.joca.2022.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 08/22/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Cartilage targeting cationic glycoprotein Avidin was PEGylated to synthesize a multi-arm Avidin (mAv) nano-construct with high drug loading content. Here we investigate mAv biodistribution and kinetics over a 7-day period following intra-articular (IA) administration in rat knee joints. METHODS Labeled mAv was injected into healthy rat knees, and joint tissues (articular cartilage, menisci, ligaments, tendons, fat pad) were harvested following sacrifice at 6 h, 1, 4 and 7 days. Its IA biodistribution and retention were measured using fluorescence microscopy. Tissue localization was compared in young vs old rats by immunohistochemistry. mAv chondrotoxicity and immune response were evaluated to determine safe carrier dose limits. RESULTS mAv penetrated through the full thickness of rat cartilage and other joint tissues within 6 h, remaining detectable within most joint tissues over 7 days. Intra-tissue uptake correlated strongly with tissue GAG concentration, confirming the dominant role of electrostatic interactions between positively charged mAv and the negatively charged aggrecan proteoglycans. mAv was uptaken by chondrocytes and also penetrated the osteocyte lacuno-canalicular system of peri-articular bone in both young and old rats. mAv did not cause cytotoxicity at concentrations up to 300 μM but elicited a dose dependent immunogenic response. CONCLUSIONS mAv's ability to target a variety of joint tissues, chondrocytes, and peri-articular osteocytes without sequestration in synovial fluid makes it a versatile carrier for delivering a wide range of drugs for treating a broad class of musculoskeletal diseases. Drugs can be conjugated using simple aqueous based avidin-biotin reaction, supporting its clinical prospects.
Collapse
Affiliation(s)
- T He
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - C Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - T Colombani
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA.
| | - S A Bencherif
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA; Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA.
| | - R M Porter
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - A G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA; Department of Mechanical Engineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
37
|
Dhanabalan KM, Dravid AA, Agarwal S, Sharath RK, Padmanabhan AK, Agarwal R. Intra-articular injection of rapamycin microparticles prevent senescence and effectively treat osteoarthritis. Bioeng Transl Med 2023; 8:e10298. [PMID: 36684078 PMCID: PMC9842044 DOI: 10.1002/btm2.10298] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/29/2022] [Accepted: 02/03/2022] [Indexed: 01/25/2023] Open
Abstract
Trauma to the knee joint is associated with significant cartilage degeneration and erosion of subchondral bone, which eventually leads to osteoarthritis (OA), resulting in substantial morbidity and healthcare burden. With no disease-modifying drugs in clinics, the current standard of care focuses on symptomatic relief and viscosupplementation. Modulation of autophagy and targeting senescence pathways are emerging as potential treatment strategies. Rapamycin has shown promise in OA disease amelioration by autophagy upregulation, yet its clinical use is hindered by difficulties in achieving therapeutic concentrations, necessitating multiple weekly injections. Rapamycin-loaded in poly(lactic-co-glycolic acid) microparticles (RMPs) induced autophagy, prevented senescence, and sustained sulphated glycosaminoglycans production in primary human articular chondrocytes from OA patients. RMPs were potent, nontoxic, and exhibited high retention time (up to 35 days) in mice joints. Intra-articular delivery of RMPs effectively mitigated cartilage damage and inflammation in surgery-induced OA when administered as a prophylactic or therapeutic regimen. Together, the study demonstrates the feasibility of using RMPs as a potential clinically translatable therapy to prevent the progression of post-traumatic OA.
Collapse
Affiliation(s)
- Kaamini M. Dhanabalan
- Centre for BioSystems Science and EngineeringIndian Institute of ScienceBengaluruIndia
| | - Ameya A. Dravid
- Centre for BioSystems Science and EngineeringIndian Institute of ScienceBengaluruIndia
| | - Smriti Agarwal
- Centre for BioSystems Science and EngineeringIndian Institute of ScienceBengaluruIndia
| | | | | | - Rachit Agarwal
- Centre for BioSystems Science and EngineeringIndian Institute of ScienceBengaluruIndia
| |
Collapse
|
38
|
Amirsaadat S, Amirazad H, Hashemihesar R, Zarghami N. An update on the effect of intra-articular intervention strategies using nanomaterials in osteoarthritis: Possible clinical application. Front Bioeng Biotechnol 2023; 11:1128856. [PMID: 36873347 PMCID: PMC9978162 DOI: 10.3389/fbioe.2023.1128856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Osteoarthritis (OA) is the most common progressive condition affecting joints. It mainly affects the knees and hips as predominant weight-bearing joints. Knee osteoarthritis (KOA) accounts for a large proportion of osteoarthritis and presents numerous symptoms that impair quality of life, such as stiffness, pain, dysfunction, and even deformity. For more than two decades, intra-articular (IA) treatment options for managing knee osteoarthritis have included analgesics, hyaluronic acid (HA), corticosteroids, and some unproven alternative therapies. Before effective disease-modifying treatments for knee osteoarthritis, treatments are primarily symptomatic, mainly including intra-articular corticosteroids and hyaluronic acid, so these agents represent the most frequently used class of drugs for managing knee osteoarthritis. But research suggests other factors, such as the placebo effect, have an essential role in the effectiveness of these drugs. Several novel intra-articular therapies are currently in the clinical trial processes, such as biological therapies, gene and cell therapies. Besides, it has been shown that the development of novel drug nanocarriers and delivery systems could improve the effectiveness of therapeutic agents in osteoarthritis. This review discusses the various treatment methods and delivery systems for knee osteoarthritis and the new agents that have been introduced or are in development.
Collapse
Affiliation(s)
- Soumayeh Amirsaadat
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Halimeh Amirazad
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Hashemihesar
- Department of Histology and Embryology, Faculty of Medicine, Altinbas University, Istanbul, Türkiye
| | - Nosratollah Zarghami
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Türkiye.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
39
|
Wang Y, Liu L, Le Z, Tay A. Analysis of Nanomedicine Efficacy for Osteoarthritis. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Yuwen Wang
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
| | - Ling Liu
- Institute of Health Innovation and Technology National University of Singapore Singapore 117599 Singapore
| | - Zhicheng Le
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
| | - Andy Tay
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
- Institute of Health Innovation and Technology National University of Singapore Singapore 117599 Singapore
- Tissue Engineering Programme National University of Singapore Singapore 117510 Singapore
| |
Collapse
|
40
|
Thampi P, Samulski RJ, Grieger JC, Phillips JN, McIlwraith CW, Goodrich LR. Gene therapy approaches for equine osteoarthritis. Front Vet Sci 2022; 9:962898. [PMID: 36246316 PMCID: PMC9558289 DOI: 10.3389/fvets.2022.962898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/08/2022] [Indexed: 01/24/2023] Open
Abstract
With an intrinsically low ability for self-repair, articular cartilage injuries often progress to cartilage loss and joint degeneration resulting in osteoarthritis (OA). Osteoarthritis and the associated articular cartilage changes can be debilitating, resulting in lameness and functional disability both in human and equine patients. While articular cartilage damage plays a central role in the pathogenesis of OA, the contribution of other joint tissues to the pathogenesis of OA has increasingly been recognized thus prompting a whole organ approach for therapeutic strategies. Gene therapy methods have generated significant interest in OA therapy in recent years. These utilize viral or non-viral vectors to deliver therapeutic molecules directly into the joint space with the goal of reprogramming the cells' machinery to secrete high levels of the target protein at the site of injection. Several viral vector-based approaches have demonstrated successful gene transfer with persistent therapeutic levels of transgene expression in the equine joint. As an experimental model, horses represent the pathology of human OA more accurately compared to other animal models. The anatomical and biomechanical similarities between equine and human joints also allow for the use of similar imaging and diagnostic methods as used in humans. In addition, horses experience naturally occurring OA and undergo similar therapies as human patients and, therefore, are a clinically relevant patient population. Thus, further studies utilizing this equine model would not only help advance the field of human OA therapy but also benefit the clinical equine patients with naturally occurring joint disease. In this review, we discuss the advancements in gene therapeutic approaches for the treatment of OA with the horse as a relevant patient population as well as an effective and commonly utilized species as a translational model.
Collapse
Affiliation(s)
- Parvathy Thampi
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - R. Jude Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, United States
| | - Joshua C. Grieger
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, United States
| | - Jennifer N. Phillips
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - C. Wayne McIlwraith
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - Laurie R. Goodrich
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States,*Correspondence: Laurie R. Goodrich
| |
Collapse
|
41
|
Jiang Z, Wang H, Zhang Z, Pan J, Yuan H. Cartilage targeting therapy with reactive oxygen species-responsive nanocarrier for osteoarthritis. J Nanobiotechnology 2022; 20:419. [PMID: 36123746 PMCID: PMC9484188 DOI: 10.1186/s12951-022-01629-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/10/2022] [Indexed: 11/12/2022] Open
Abstract
Targeting cartilage is a promising strategy for the treatment of osteoarthritis, and various delivery vehicles were developed to assist the therapeutic agents into cartilage. However, the underlying biomechanisms and potential bioactivities remain oversimplified. Inspired by oxidative stress in the pathogenesis of osteoarthritis, we firstly testified the antioxidant capacity of a synthetic small molecule compound, oltipraz (OL), to the chondrocytes treated by IL-1β. Then a functional reactive oxygen species (ROS) responsive nanocarrier, mesoporous silica nanoparticles (MSN) modified with methoxy polyethylene glycol-thioketal, was constructed. In vitro biomolecular results showed that compared with OL alone, MSN-OL could significantly activate Nrf2/HO-1 signaling pathway, which exhibited better ROS-scavenging proficiency and greater anti-apoptotic ability to protect mitochondrial membrane potential of chondrocytes. Further bioinformatics analysis revealed that MSN-OL suppressed clusters of genes associated with extracellular matrix organization, cell apoptosis and cellular response to oxidative stress. Animal experiments further confirmed the great cartilage-protecting ability of MSN-OL through upregulating the expression of Nrf2/HO-1 signaling pathway without obvious toxicity. In summary, this study provided a delivery system through ROS-responsive regulation of the therapeutic agents into chondrocytes of the cartilage, and confirmed the exact biological mechanisms of this innovative strategy.
Collapse
Affiliation(s)
- Zengxin Jiang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Hao Wang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zeng Zhang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Jianfeng Pan
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| | - Hengfeng Yuan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China. .,Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
42
|
Xiao L, Cui J, Sun Z, Liu Y, Zheng J, Dong Y. Therapeutic potential of nanotechnology-based approaches in osteoarthritis. Front Pharmacol 2022; 13:920824. [PMID: 36003519 PMCID: PMC9394598 DOI: 10.3389/fphar.2022.920824] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022] Open
Abstract
Osteoarthritis (OA) is a multifactorial disease that affects the entire joint, often resulting in severe pain, disability, psychological distress, and a lower quality of life. Patient self-management is emphasized in OA clinical recommendations. Currently, the clinical treatment of OA mainly focuses on pain relief and the improvement of joint function, with few options for regenerating degenerative cartilage or slowing the progression of OA. Therefore, we first reviewed the current treatment of OA, and then summarized the research advances of nanotechnology in OA treatment, including nano drug delivery systems for small molecule drugs, nucleic acids and proteins, nano-scaffolds for cartilage regeneration, and nanoparticle lubricants. Finally, we discussed the opportunities and potential challenges of nanotechnology in OA treatment.
Collapse
Affiliation(s)
- Likang Xiao
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Jiarui Cui
- School of Rehabilitation and Health Preservation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhuang Sun
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Yunke Liu
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Jia Zheng
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
- *Correspondence: Jia Zheng, ; Yonghui Dong,
| | - Yonghui Dong
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
- *Correspondence: Jia Zheng, ; Yonghui Dong,
| |
Collapse
|
43
|
Abdeltawab H, Svirskis D, Hill AG, Sharma M. Increasing the Hydrophobic Component of Poloxamers and the Inclusion of Salt Extend the Release of Bupivacaine from Injectable In Situ Gels, While Common Polymer Additives Have Little Effect. Gels 2022; 8:gels8080484. [PMID: 36005085 PMCID: PMC9407117 DOI: 10.3390/gels8080484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/18/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Various strategies have been applied to reduce the initial burst of drug release and sustain release from poloxamer-based thermoresponsive gels. This work focussed on investigating different formulation approaches to minimise the initial burst of release and sustain the release of the small hydrophilic drug bupivacaine hydrochloride from poloxamer-based thermoresponsive gels. Various in situ gel formulations were prepared by varying the polypropylene oxide (PPO)/polyethylene oxide (PEO) ratio and by adding additives previously described in the literature. It was observed that increasing the PPO/PEO ratio from 0.28 to 0.30 reduced the initial burst release from 17.3% ± 1.8 to 9.1% ± 1.2 during the first six hours and extended the release profile from 10 to 14 days. Notably, the inclusion of sodium chloride (NaCl 0.4% w/w) further reduced the initial burst release to 1.8% ± 1.1 over the first 6 h. Meanwhile, physical blending with additive polymers had a negligible effect on the burst release and overall release profile. The findings suggest that extended release of bupivacaine hydrochloride, with reduced initial burst release, can be achieved simply by increasing the PPO/PEO ratio and the inclusion of NaCl.
Collapse
Affiliation(s)
- Hani Abdeltawab
- School of Pharmacy, Faculty of Medical & Health Sciences, The University of Auckland, Auckland 1023, New Zealand; (H.A.); (D.S.)
| | - Darren Svirskis
- School of Pharmacy, Faculty of Medical & Health Sciences, The University of Auckland, Auckland 1023, New Zealand; (H.A.); (D.S.)
| | - Andrew G. Hill
- Department of Surgery, South Auckland Clinical Campus, The University of Auckland, Middlemore Hospital, Auckland 2025, New Zealand;
| | - Manisha Sharma
- School of Pharmacy, Faculty of Medical & Health Sciences, The University of Auckland, Auckland 1023, New Zealand; (H.A.); (D.S.)
- Correspondence: ; Tel.: +64-9-373-7599 (ext. 81830); Fax: +64-9-367-7192
| |
Collapse
|
44
|
Anchi P, Chilvery S, Tekalkar S, bolla L, Rao Gajula SN, Sonti R, Godugu C. Nimbolide loaded sustained release microparticles as single-dose formulations for effective management of arthritis. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
45
|
宗 路, 吴 乾, 董 仲, 黄 立, 杨 惠. [Research progress of nanomaterials for intra-articular targeted drug delivery in treatment of osteoarthritis]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2022; 36:908-914. [PMID: 35848190 PMCID: PMC9288906 DOI: 10.7507/1002-1892.202203033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/27/2022] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To review the research progress of intra-articular targeted delivery of nanomaterials in the treatment of osteoarthritis (OA). METHODS The domestic and foreign related literature on intra-articular targeted delivery of nanomaterials for the treatment of OA was extensively reviewed, and their targeting strategies were discussed and summarized. RESULTS Rapid drug clearance from the joint remains a critical limitation in drug efficacy. Nanocarriers can not only significantly improve the residence profiles of drugs in the joint, but also achieve targeted delivery of drugs to specific joint tissues through active or passive targeting strategies. CONCLUSION With the continuous development of various emerging tissue- or cell-specific drugs, the targeted delivery of drugs with nanomaterials promise to realize the clinical translation of these drugs in the treatment of OA.
Collapse
Affiliation(s)
- 路杰 宗
- 苏州大学附属第一医院骨科(江苏苏州 215000)Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou Jiangsu, 215000, P. R. China
| | - 乾 吴
- 苏州大学附属第一医院骨科(江苏苏州 215000)Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou Jiangsu, 215000, P. R. China
- 昆山市第一人民医院骨科(江苏昆山 215300)Department of Orthopedics, the First People’s Hospital of Kunshan, Kunshan Jiangsu, 215300, P. R. China
| | - 仲琛 董
- 苏州大学附属第一医院骨科(江苏苏州 215000)Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou Jiangsu, 215000, P. R. China
| | - 立新 黄
- 苏州大学附属第一医院骨科(江苏苏州 215000)Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou Jiangsu, 215000, P. R. China
| | - 惠林 杨
- 苏州大学附属第一医院骨科(江苏苏州 215000)Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou Jiangsu, 215000, P. R. China
| |
Collapse
|
46
|
Wang C, Zhang J, Li B, Zuo J, Li Y, Sun Y, Wang F, Liu K, Li J. High-Efficiency Treatment for Osteoarthritis via Self-Assembled Dual-Functionalized Nanobiologics. ACS Biomater Sci Eng 2022; 8:3320-3328. [PMID: 35801691 DOI: 10.1021/acsbiomaterials.2c00332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Osteoarthritis (OA) is a progressive joint disease that has a complex pathogenesis and lacks effective drugs. OA develops with cartilage degeneration and inflammation, thus synthesizing a drug with both anti-inflammatory properties and cartilage-repair capacity provides a promising treatment strategy. Therefore, in this study, we report self-assembled nanobiologics composed of an engineered recombinant IL-1 receptor antagonist (IL-1Ra) chimeric protein with chondroitin sulfate (CS). The nanobiologics, termed ICN, exhibit extraordinary biocompatibility, low immunogenicity, and good bioefficacy. Furthermore, our study revealed that ICN significantly reduced cartilage degradation, inhibited synovial inflammation, and suppressed osteophyte formation in OA rat models. The excellent therapeutic effects on OA can be attributed to the synergistic anti-inflammatory and cartilage-repair properties of ICN's constituents. Thus, our novel strategy offers insights into the development of drugs for OA treatment and research on nanobiomedicine, which can also be adapted for other diseases with similar pathologies.
Collapse
Affiliation(s)
- Chenyu Wang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, China.,State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jinrui Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Bo Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.,Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Jianlin Zuo
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yuanxin Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yao Sun
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Fan Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Kai Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.,Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Jingjing Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| |
Collapse
|
47
|
Siefen T, Bjerregaard S, Borglin C, Lamprecht A. Assessment of joint pharmacokinetics and consequences for the intraarticular delivery of biologics. J Control Release 2022; 348:745-759. [PMID: 35714731 DOI: 10.1016/j.jconrel.2022.06.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 01/15/2023]
Abstract
Intraarticular (IA) injections provide the opportunity to deliver biologics directly to their site of action for a local and efficient treatment of osteoarthritis. However, the synovial joint is a challenging site of administration since the drug is rapidly eliminated across the synovial membrane and has limited distribution into cartilage, resulting in unsatisfactory therapeutic efficacy. In order to rationally develop appropriate drug delivery systems, it is essential to thoroughly understand the unique biopharmaceutical environments and kinetics in the joint to adequately simulate them in relevant experimental models. This review presents a detailed view on articular kinetics and drug-tissue interplay of IA administered drugs and summarizes how these can be translated into reasonable formulation strategies by identification of key factors through which the joint residence time can be prolonged and specific structures can be targeted. In this way, pros and cons of the delivery approaches for biologics will be evaluated and the extent to which biorelevant models are applicable to gain mechanistic insights and ameliorate formulation design is discussed.
Collapse
Affiliation(s)
- Tobias Siefen
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany
| | | | | | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; PEPITE (EA4267), University of Burgundy/Franche-Comté, Besançon, France.
| |
Collapse
|
48
|
Baumann JR, Stoker AM, Bozynski CC, Sherman SL, Cook JL. An Injectable Containing Morphine, Ropivacaine, Epinephrine, and Ketorolac Is Not Cytotoxic to Articular Cartilage Explants From Degenerative Knees. Arthroscopy 2022; 38:1980-1995. [PMID: 34952188 DOI: 10.1016/j.arthro.2021.12.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 02/02/2023]
Abstract
PURPOSE The purpose of this study was to determine the effects of a multidrug injectate containing morphine, ropivacaine, epinephrine, and ketorolac, commonly referred to as the "Orthococktail," on cartilage tissue viability and metabolic responses using an established in vitro model. METHODS With institutional review board approval and informed patient consent, tissues normally discarded after total knee arthroplasty (TKA) were recovered. Full-thickness cartilage explants (n = 72, Outerbridge grade 1 to 3) were created and bisected. Paired explant halves were treated with either 1 mL Orthococktail or 1 mL of saline and cultured for 8 hours at 37°C, with 0.5 mL of the treatment being removed and replaced with tissue culture media every hour. Explants were cultured for 6 days, and media were changed and collected on days 3 and 6. After day 6, tissues were processed for cell viability, weighed, and processed for histologic grading. Outcome measures were compared for significant differences between treated and untreated samples. RESULTS There were no significant differences in cartilage viability between control and Orthococktail-treated samples across a spectrum of cartilage pathologies. Orthococktail treatment consistently resulted in a significant decrease in the release of PGE2, MCP-1, MMP-7, and MMP-8 on day 3 of culture and PGE2, MMP-3, MMP-7, and MMP-8 on day 6 of culture, compared with saline controls. CONCLUSION The results of the present study indicate that an Orthococktail injection composed of morphine, ropivacaine, epinephrine, and ketorolac is associated with a transient decrease in degradative and inflammatory mediators produced by more severely affected articular cartilage and may mitigate perioperative joint pain such that postoperative narcotic drug use could be reduced. CLINICAL RELEVANCE The Orthococktail solution used in this study may be a safe intraoperative, intra-articular injection option for patients undergoing joint arthroplasty and other joint preservation surgical procedures.
Collapse
Affiliation(s)
- John R Baumann
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri, U.S.A
| | - Aaron M Stoker
- Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri, U.S.A.; Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri, U.S.A..
| | - Chantelle C Bozynski
- Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri, U.S.A.; Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri, U.S.A
| | - Seth L Sherman
- Department of Orthopaedic Surgery, Stanford University, CalifCornia, U.S.A
| | - James L Cook
- Department of Orthopaedic Surgery, University of Missouri, Columbia, Missouri, U.S.A.; Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, Missouri, U.S.A
| |
Collapse
|
49
|
Dunshee LC, McDonough R, Price C, Kiick KL. Retention of peptide-based vesicles in murine knee joints after intra-articular injection. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
50
|
Mertz N, Bock F, Østergaard J, Yaghmur A, Weng Larsen S. Investigation of diclofenac release and dynamic structural behavior of non-lamellar liquid crystal formulations during in situ formation by UV-Vis imaging and SAXS. Int J Pharm 2022; 623:121880. [PMID: 35661744 DOI: 10.1016/j.ijpharm.2022.121880] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 10/18/2022]
Abstract
In situ formation of high viscous inverse lyotropic non-lamellar liquid crystalline phases is a promising approach for sustained drug delivery in the joint. The in situ forming process on exposure of two diclofenac-loaded preformulations to aqueous media was characterized with respect to depot size and shape, initial release and structural transitions using UV-Vis imaging and spatially and time-resolved synchrotron small-angle X-ray scattering (SAXS). The preformulations consisted of 10 % (w/w) ethanol, 10 % (w/w) water and a binary lipid mixture of glycerol monooleate (GMO):1,2-dioleoyl-sn-glycero-3-phospho-rac-(1-glycerol) (DOPG) or GMO:medium chain triglycerides (MCT). Upon injection of preformulations into an employed injection-cell containing excess of bio-relevant medium, rapid generation of liquid crystalline depots was observed. UV-Vis images and constructed 2D SAXS maps of the injection-cell showed depots with different shapes and sizes, and features with high nanostructural heterogeneity. More extensive swelling of the GMO:DOPG-based preformulation was observed compared to the GMO:MCT-based preformulation. The UV image analysis found that a higher amount of diclofenac was released in the image area after 20 h from the GMO:MCT-depot compared to the GMO:DOPG-depot. The injection-cell setup employing UV-Vis imaging and synchrotron SAXS constitutes an attractive approach for evaluating the in situ forming processes of liquid crystalline depots.
Collapse
Affiliation(s)
- Nina Mertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - Frederik Bock
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - Jesper Østergaard
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - Anan Yaghmur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - Susan Weng Larsen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|