1
|
Chen F, Wang W, Zhao H, Zhang Z, Pang N, Tang Y, Wang T, Liu C, Huang Z, Mou F, Yu C, Zhang H. MXene-Based Cartilage-Adhesive Microspheres for Photothermal-Controlled Hydrophobic Drug Release and Mesenchymal Stem Cell Delivery in Osteoarthritis. ACS NANO 2025; 19:20502-20515. [PMID: 40446309 DOI: 10.1021/acsnano.4c16918] [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: 06/11/2025]
Abstract
Intra-articular drug injection is an effective treatment for osteoarthritis (OA). However, the rapid clearance of drugs from the joint cavity results in low drug utilization and suboptimal therapeutic outcomes. This study describes MXene-based cartilage-adhesive microspheres for photothermal-controlled hydrophobic drug release and bone marrow mesenchymal stem cell (BMSC) delivery. Nano cationic amylose (NCA) was obtained by modifying amylose with glycidyltrimethylammonium chloride (GTAC), and hydrophobic drug Kartogenin (KGN) was encapsulated in the hydrophobic helical cavity of NCA through ultrasonic treatment, resulting in nano cationic amylose@KGN complexes (NCA@KGN). HAMA/MXene-NCA@KGN (H/M-NCA@KGN) microspheres were prepared using a microfluidic device. These microspheres exhibited excellent biocompatibility, effectively adhered to the cartilage surface, and carried BMSCs. H/M-NCA@KGN microspheres demonstrated photothermal-controlled release of the hydrophobic drug KGN. Notably, KGN promoted the differentiation of BMSCs into chondrocytes, thereby improving the loss of extracellular matrix in joint cartilage. Additionally, appropriate thermal stimulation induced the expression of heat shock protein 70 (HSP70) in OA chondrocytes, providing a protective effect and delaying the progression of OA. H/M-NCA@KGN microspheres enable controlled hydrophobic drug release and stem cell delivery for potential OA treatment applications.
Collapse
Affiliation(s)
- Fan Chen
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| | - Wenzhe Wang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| | - Hengxin Zhao
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| | - Zian Zhang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| | - Nanyu Pang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| | - Yijie Tang
- Rehabilitation Medicine Department, the First People's Hospital of Yancheng, Yancheng, Jiangsu 224002, PR China
| | - Tian Wang
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textiles and Clothing, State Key Laboratory of Bio-Fibers and Eco-Textiles, Research Center for Intelligent and Wearable Technology, Qingdao University, Qingdao 266071, PR China
| | - Chang Liu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| | - Zhenchao Huang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| | - Feiyu Mou
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| | - Chaoqun Yu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| | - Haining Zhang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| |
Collapse
|
2
|
Kosonen JP, Eskelinen ASA, Orozco GA, Coleman MC, Goetz JE, Anderson DD, Grodzinsky AJ, Tanska P, Korhonen RK. Mechanobiochemical finite element model to analyze impact-loading-induced cell damage, subsequent proteoglycan loss, and anti-oxidative treatment effects in articular cartilage. Biomech Model Mechanobiol 2025:10.1007/s10237-025-01961-8. [PMID: 40348944 DOI: 10.1007/s10237-025-01961-8] [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: 09/26/2024] [Accepted: 04/05/2025] [Indexed: 05/14/2025]
Abstract
Joint trauma often leads to articular cartilage degeneration and post-traumatic osteoarthritis (PTOA). Pivotal determinants include trauma-induced excessive tissue strains that damage cartilage cells. As a downstream effect, these damaged cells can trigger cartilage degeneration via oxidative stress, cell death, and proteolytic tissue degeneration. N-acetylcysteine (NAC) has emerged as an antioxidant capable of inhibiting oxidative stress, cell death, and cartilage degeneration post-impact. However, the temporal effects of NAC are not fully understood and remain difficult to assess solely by physical experiments. Thus, we developed a computational finite element analysis framework to simulate a drop-tower impact of cartilage in Abaqus, and subsequent oxidative stress-related cell damage, and NAC treatment upon cartilage proteoglycan content in Comsol Multiphysics, based on prior ex vivo experiments. Model results provide evidence that immediate NAC treatment can reduce proteoglycan loss by mitigating oxidative stress, cell death (improved proteoglycan biosynthesis), and enzymatic proteoglycan depletion. Our simulations also indicate that delayed NAC treatment may not inhibit cartilage proteoglycan loss despite reduced cell death after impact. These results enhance understanding of the temporal effects of impact-related cell damage and treatment that are critical for the development of effective treatments for PTOA. In the future, our modeling framework could increase understanding of time-dependent mechanisms of oxidative stress and downstream effects in injured cartilage and aid in developing better treatments to mitigate PTOA progression.
Collapse
Affiliation(s)
- Joonas P Kosonen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland.
| | - Atte S A Eskelinen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Gustavo A Orozco
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Mitchell C Coleman
- Departments of Orthopedics and Rehabilitation and Biomedical Engineering, University of Iowa, Iowa, USA
| | - Jessica E Goetz
- Departments of Orthopedics and Rehabilitation and Biomedical Engineering, University of Iowa, Iowa, USA
| | - Donald D Anderson
- Departments of Orthopedics and Rehabilitation and Biomedical Engineering, University of Iowa, Iowa, USA
| | - Alan J Grodzinsky
- Departments of Biological Engineering, Electrical Engineering and Computer Science, and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, USA
| | - Petri Tanska
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| | - Rami K Korhonen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
3
|
Scull G, Thompson J, Osareh M, Rey Y, Aligwekwe A, Finkelstein S, Schnabel LV, Fisher M, Brown A. Noninvasive Fibrin Targeting Colloid-Mediated Intra-Articular Repair. J Biomed Mater Res A 2025; 113:e37901. [PMID: 40145327 PMCID: PMC11974503 DOI: 10.1002/jbm.a.37901] [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: 01/10/2025] [Revised: 02/27/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025]
Abstract
Musculoskeletal knee injuries are common and debilitating, with the most prevalent soft tissue injuries being anterior cruciate ligament (ACL) and meniscal tears. These tears do not heal well naturally, and biological therapies involving scaffolds are often unsuccessful, due in part to the synovial fluid environment of the joint. Viscous synovial fluid contains high concentrations of degradative enzymes, including plasmin, which prevents the stable formation of provisional fibrin scaffolds. Lack of provisional scaffold formation prevents bridging of torn tissue and subsequent remodeling for permanent tissue repair. Coagulation factors such as fibrinogen and thrombin, reinforced with synthetic platelet-like particles (PLPs), can be introduced to synovial fluid to promote fibrin scaffold formation. PLPs bind to and retract fibrin fibers to enhance stiffness, density, and stability of fibrin scaffolds. Therefore, the objective of this work is to investigate the role of PLPs in enhancing fibrin scaffold formation and degradation capabilities within synovial fluid and to characterize the resulting scaffold structure, density, and mechanics. We investigated effects in synovial fluid with high or low viscosity, as viscosity can change with injury and can vary between individuals. Following the addition of clotting factors and PLPs to synovial fluid, we found an increase in fibrin scaffold density, structure, and maximum mechanics for low viscosity, but not high viscosity, synovial fluid groups. Furthermore, by lowering the viscosity of synovial fluid with hyaluronidase, the increase in scaffold density following PLP addition was restored, indicating the strong role of synovial fluid viscosity on stable scaffold formation. This technology contributes to the development of a more robust fibrin-based therapy for intra-articular musculoskeletal injuries.
Collapse
Affiliation(s)
- Grant Scull
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Jacob Thompson
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Melika Osareh
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Ysabel Rey
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Adrian Aligwekwe
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Sofie Finkelstein
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Lauren V. Schnabel
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| | - Matthew Fisher
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
- Department of Orthopaedics, UNC-Chapel Hill, Chapel Hill, NC
| | - Ashley Brown
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| |
Collapse
|
4
|
Stealey S, Dharmesh E, Bhagat M, Tyagi AM, Schab A, Hong M, Osbourn D, Abu-Amer Y, Jelliss PA, Zustiak SP. Super-lubricous polyethylene glycol hydrogel microspheres for use in knee osteoarthritis treatments. NPJ BIOMEDICAL INNOVATIONS 2025; 2:11. [PMID: 40144306 PMCID: PMC11932927 DOI: 10.1038/s44385-025-00011-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/25/2025] [Indexed: 03/28/2025]
Abstract
Knee osteoarthritis (OA) is characterized by cartilage degeneration and significant reduction in lubrication. One strategy to recover the natural lubrication of the synovial fluid is the injection of hydrogel microspheres. Here, we have fabricated polyethylene glycol (PEG)-based hydrogel microspheres via a modified electrospraying setup. To improve throughout, crosslinking of PEG droplets was delayed until after droplet formation was complete. A custom-synthesized super-lubricious copolymer consisting of adhesive dopamine methacrylate (DMA), zwitterionic sulfobetaine methacrylate (SBMA), and fluorescent rhodamine B was used to dip-coat the PEG microspheres. Super-lubricious PEG microspheres coating reduced coefficient of friction by 57% compared to simulated synovial fluid, indicating beneficial lubrication properties. When injected into C57BL6 mice, PEG microspheres exhibited stability for up to 26 d and did not adversely affect mouse behavior. These super-lubricious PEG microspheres offer great promise to reduce the friction that is a hallmark of progressive OA, potentially mitigating the need for total knee arthroplasty.
Collapse
Affiliation(s)
- Samuel Stealey
- Department of Biomedical Engineering, School of Science and Engineering, Saint Louis University, Saint Louis, MO USA
| | - Ether Dharmesh
- Department of Biomedical Engineering, School of Science and Engineering, Saint Louis University, Saint Louis, MO USA
| | - Maitreyi Bhagat
- Department of Chemistry, School of Science and Engineering, Saint Louis University, Saint Louis, MO USA
| | - Abdul Malik Tyagi
- Department of Orthopaedics, Washington University in Saint Louis, Saint Louis, MO USA
| | - Andrew Schab
- Department of Biomedical Engineering, School of Science and Engineering, Saint Louis University, Saint Louis, MO USA
| | - Melissa Hong
- Department of Chemistry, School of Science and Engineering, Saint Louis University, Saint Louis, MO USA
| | - Damon Osbourn
- Department of Chemistry, School of Science and Engineering, Saint Louis University, Saint Louis, MO USA
| | - Yousef Abu-Amer
- Department of Orthopaedics, Washington University in Saint Louis, Saint Louis, MO USA
| | - Paul A. Jelliss
- Department of Chemistry, School of Science and Engineering, Saint Louis University, Saint Louis, MO USA
| | - Silviya Petrova Zustiak
- Department of Biomedical Engineering, School of Science and Engineering, Saint Louis University, Saint Louis, MO USA
| |
Collapse
|
5
|
Li W, Sun J, Feng S, La Rosa A, Zhang P, Wu EY, Loeser R, Li C. Secreted PD-L1 alleviates inflammatory arthritis in mice through local and systemic AAV gene therapy. Front Immunol 2025; 16:1527858. [PMID: 39963137 PMCID: PMC11830590 DOI: 10.3389/fimmu.2025.1527858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction Rheumatoid arthritis (RA) primarily affects the joints but can also affect multiple organs and profoundly impacts patients' ability to carry out daily activities, mental health, and life expectancy. Current treatments for RA are limited in terms of duration, efficacy, and adverse effects. PD-L1 is a checkpoint protein that plays important roles in immune regulation and has been implicated in the initiation and progression of multiple autoimmune diseases. Method In a previous study, we demonstrated that intra-articular injection with adeno-associated virus (AAV) vectors encoding wild type PD-L1 improved local inflammation in the joint in the collagen-induced arthritis (CIA) mouse model of RA. To further improve efficacy, we explored AAV-mediated delivery of the soluble PD-L1 (sPD-L1) to CIA mice. Result After intra-articular injection of AAV6 vectors expressing the optimal isoform of sPD-L1 (shPD-L1), more potency was observed when compared to wild type PD-L1, with a lower dose of AAV6/shPD-L1 needed for arthritis improvement. To study the therapeutic effect of systemic expression of sPD-L1, we administered AAV8/shPD-L1 gene therapy in CIA mice via retro-orbital injection and found significant improvements in joint inflammation and paw swelling, exhibiting similar phenotypes to that in naïve mice. The levels of total immunoglobulin and anti-collagen specific antibodies were lower in AAV8/shPD-L1 treated CIA mice than those in controls. The levels of pro-inflammatory cytokines in blood were also significantly decreased in shPD-L1 treated mice. Additionally, T cell apoptosis rates in the spleen showed a 2-fold increase in treated mice. Finally, we investigated the therapeutic effect of AAV/shPD-L1 via intramuscular injection. After injection of AAV6/shPD-L1, decreased paw swelling, reduced joint inflammation, and lower levels of pro-inflammatory cytokines in blood were achieved. The therapeutic effect of shPD-L1 was dose dependent via intramuscular treatment with AAV vectors. Conclusion In conclusion, the findings in this study suggest that intra-articular injection of AAV vectors encoding sPD-L1 results in greater therapeutic benefit on arthritis, and systemic AAV/sPD-L1 is able to block the development of inflammatory arthritis with inhibition of the systemic immune response, underlining the potential of gene therapy with systemic delivery of shPD-L1 via AAV vectors in RA.
Collapse
Affiliation(s)
- Wenjun Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Oral and Craniofacial Biomedicine, University of North Carolina Adams School of Dentistry, Chapel Hill, NC, United States
| | - Junjiang Sun
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Susi Feng
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ariana La Rosa
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Panli Zhang
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Eveline Y. Wu
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Richard Loeser
- Division of Rheumatology, Allergy, and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
6
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2025; 14:e2402737. [PMID: 39506433 PMCID: PMC11730424 DOI: 10.1002/adhm.202402737] [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: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Wei Liu
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Jagadeesh K. Venkatesan
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Henning Madry
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Magali Cucchiarini
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| |
Collapse
|
7
|
Smith JS, Malla GD, Garcia JD, Gebert JE, Noll CV, Mulon PY, Knych HK. Pharmacokinetics of intraarticular liposomal amphotericin B in goats (Capra aegagrus hircus). J Vet Pharmacol Ther 2024; 47:252-256. [PMID: 38557931 DOI: 10.1111/jvp.13442] [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: 01/05/2024] [Revised: 02/16/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024]
Abstract
Lameness is a significant welfare concern in goats. Amphotericin B is used via intraarticular (IA) administration in models to study experimentally induced lameness in large animals. The main objective of this study was to estimate plasma pharmacokinetic (PK) parameters for amphotericin B in goats after a single IA administration. Liposomal amphotericin B was administered to ten Kiko-cross goats at a dose of 10 mg total (range: 0.34-0.51 mg/kg) via IA administration into the right hind lateral distal interphalangeal joint. Plasma samples were collected over 96 h. Amphotericin B concentrations were measured via liquid chromatography/mass spectrometry (LC-MS/MS). A non-compartmental analysis was used to derive PK parameters. Following single IA administration, maximum plasma concentration was estimated at 54.6 ± 16.5 ng/mL, and time to maximum concentration ranged from 6 to 12 h. Elimination half-life was estimated at 30.9 ± 16.5 h, and mean residence time was 45.1 ± 10.4 h. The volume of distribution after IA administration was 13.3 ± 9.4 L/kg. The area under the curve was 1481 ± 761 h*ng/mL. The achieved maximum concentration was less than the observed concentrations for other species and routes of administration. Further research is needed into the pharmacodynamics of IA liposomal amphotericin B in goats to determine specific research strategies.
Collapse
Affiliation(s)
- Joe S Smith
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Grace D Malla
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Jessica D Garcia
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Jessica E Gebert
- College of Veterinary Medicine, Lincoln Memorial University, Harrogate, Tennessee, USA
| | - Charlene V Noll
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Pierre-Yves Mulon
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Heather K Knych
- K. L. Maddy Equine Analytical Pharmacology Laboratory, School of Veterinary Medicine, University of California-Davis, Davis, California, USA
| |
Collapse
|
8
|
Roy HS, Murugesan P, Kulkarni C, Arora M, Nagar GK, Guha R, Chattopadhyay N, Ghosh D. On-demand release of a selective MMP-13 blocker from an enzyme-responsive injectable hydrogel protects cartilage from degenerative progression in osteoarthritis. J Mater Chem B 2024; 12:5325-5338. [PMID: 38669084 DOI: 10.1039/d3tb02871b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
In osteoarthritis (OA), the degradation of cartilage is primarily driven by matrix metalloprotease-13 (MMP-13). Hence, the inhibition of MMP-13 has emerged as an attractive target for OA treatment. Among the various approaches that are being explored for MMP-13 regulation, blocking of the enzyme with specific binding molecules appears to be a more promising strategy for preventing cartilage degeneration. To enhance effectiveness and ensure patient compliance, it is preferable for the binding molecule to exhibit sustained activity when administered directly into the joint. Herein, we present an enzyme-responsive hydrogel that was designed to exhibit on-demand, the sustained release of BI-4394, a potent and highly selective MMP-13 blocker. The stable and compatible hydrogel was prepared using triglycerol monostearate. The efficacy of the hydrogel to prevent cartilage damage was assessed in a rat model of OA induced by anterior cruciate ligament transection (ACLT). The results revealed that in comparison to the rats administrated weekly with intra-articular BI-4394, the hydrogel implanted rats had reduced levels of inflammation and bone erosion. In comparison to untreated control, the cartilage in animals administered with BI-4394/hydrogel exhibited significant levels of collagen-2 and aggrecan along with reduced MMP-13. Overall, this study confirmed the potential of BI-4394 delivery using an enzyme-responsive hydrogel as a promising treatment option to treat the early stages of OA by preventing further cartilage degradation.
Collapse
Affiliation(s)
- Himadri Shekhar Roy
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Preethi Murugesan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Chirag Kulkarni
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Malika Arora
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Geet Kumar Nagar
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Rajdeep Guha
- Division of Laboratory Animal Facility, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Deepa Ghosh
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| |
Collapse
|
9
|
Xiao P, Han X, Huang Y, Yang J, Chen L, Cai Z, Hu N, Cui W, Huang W. Reprogramming macrophages via immune cell mobilized hydrogel microspheres for osteoarthritis treatments. Bioact Mater 2024; 32:242-259. [PMID: 37869722 PMCID: PMC10589729 DOI: 10.1016/j.bioactmat.2023.09.010] [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: 06/02/2023] [Revised: 08/12/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Regulating macrophage activation precisely is crucial in treating chronic inflammation in osteoarthritis (OA). However, the stable pro-inflammatory state and deep distribution of macrophages in vivo pose a great challenge to treatment. In this study, inspired by the innate immune, immune cell mobilized hydrogel microspheres were constructed by microfluidic methods and load chemokines, macrophage antibodies and engineered cell membrane vesicles (sEVs) via covalent and non-covalent junctions. The immune cell mobilized hydrogel microspheres, based on a mixture of streptavidin grafted hyaluronic acid methacrylate (HAMA-SA) and Chondroitin sulfate methacrylate (ChSMA) microspheres (HCM), can recruit, capture and reprogram proinflammatory macrophages in the joint cavity to improve the joint inflammatory microenvironment. In vitro experiments demonstrated that immune cell mobilized hydrogel microspheres had excellent macrophage recruitment, capture, and reprogramming abilities. Pro-inflammatory macrophages can be transformed into anti-inflammatory macrophages with an efficiency of 88.5 %. Animal experiments also revealed significant reduction in synovial inflammation and cartilage matrix degradation of OA. Therefore, the immune cell mobilized hydrogel microspheres may be an effective treatment of OA inflammation for the future.
Collapse
Affiliation(s)
- Pengcheng Xiao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
| | - Xiaoyu Han
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
- 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, PR China
| | - Yanran Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
| | - Jianye Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
| | - Li Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
| | - Zhengwei Cai
- 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, PR China
| | - Ning Hu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, 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, PR China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
| |
Collapse
|
10
|
Hollander JM, Goraltchouk A, Liu J, Xu E, Luppino F, McAlindon TE, Zeng L, Seregin A. Single Injection AAV2-FGF18 Gene Therapy Reduces Cartilage Loss and Subchondral Bone Damage in a Mechanically Induced Model of Osteoarthritis. Curr Gene Ther 2024; 24:331-345. [PMID: 38783531 DOI: 10.2174/0115665232275532231213063634] [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: 08/04/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 05/25/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a highly debilitating, degenerative pathology of cartilaginous joints affecting over 500 million people worldwide. The global economic burden of OA is estimated at $260-519 billion and growing, driven by aging global population and increasing rates of obesity. To date, only the multi-injection chondroanabolic treatment regimen of Fibroblast Growth Factor 18 (FGF18) has demonstrated clinically meaningful disease-modifying efficacy in placebo-controlled human trials. Our work focuses on the development of a novel single injection disease-modifying gene therapy, based on FGF18's chondroanabolic activity. METHODS OA was induced in Sprague-Dawley rats using destabilization of the medial meniscus (DMM) (3 weeks), followed by intra-articular treatment with 3 dose levels of AAV2-FGF18, rh- FGF18 protein, and PBS. Durability, redosability, and biodistribution were measured by quantifying nLuc reporter bioluminescence. Transcriptomic analysis was performed by RNA-seq on cultured human chondrocytes and rat knee joints. Morphological analysis was performed on knee joints stained with Safranin O/Fast Green and anti-PRG antibody. RESULTS Dose-dependent reductions in cartilage defect size were observed in the AAV2-FGF18- treated joints relative to the vehicle control. Total defect width was reduced by up to 76% and cartilage thickness in the thinnest zone was increased by up to 106%. Morphologically, the vehicle- treated joints exhibited pronounced degeneration, ranging from severe cartilage erosion and bone void formation, to subchondral bone remodeling and near-complete subchondral bone collapse. In contrast, AAV2-FGF18-treated joints appeared more anatomically normal, with only regional glycosaminoglycan loss and marginal cartilage erosion. While effective at reducing cartilage lesions, treatment with rhFGF18 injections resulted in significant joint swelling (19% increase in diameter), as well as a decrease in PRG4 staining uniformity and intensity. In contrast to early-timepoint in vitro RNA-seq analysis, which showed a high degree of concordance between protein- and gene therapy-treated chondrocytes, in vivo transcriptomic analysis, revealed few gene expression changes following protein treatment. On the other hand, the gene therapy treatment exhibited a high degree of durability and localization over the study period, upregulating several chondroanabolic genes while downregulating OA- and fibrocartilage-associated markers. CONCLUSION FGF18 gene therapy treatment of OA joints can provide benefits to both cartilage and subchondral bone, with a high degree of localization and durability.
Collapse
Affiliation(s)
- Judith M Hollander
- Department of Immunology, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, United States of America
- Remedium Bio, Inc. 1116 Great Plain Ave, Suite 203, Needham, MA, United States of America
| | - Alex Goraltchouk
- Remedium Bio, Inc. 1116 Great Plain Ave, Suite 203, Needham, MA, United States of America
| | - Jingshu Liu
- Department of Immunology, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, United States of America
| | - Ellyn Xu
- Department of Immunology, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, United States of America
| | - Francesco Luppino
- Remedium Bio, Inc. 1116 Great Plain Ave, Suite 203, Needham, MA, United States of America
| | - Timothy E McAlindon
- Division of Rheumatology, Immunology, and Allergy, Tufts Medical Center, Boston, MA, United States of America
| | - Li Zeng
- Department of Immunology, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, United States of America
| | - Alexey Seregin
- Remedium Bio, Inc. 1116 Great Plain Ave, Suite 203, Needham, MA, United States of America
| |
Collapse
|
11
|
Lamela-Gómez I, Gonçalves LM, Almeida AJ, Luzardo-Álvarez A. Infliximab microencapsulation: an innovative approach for intra-articular administration of biologics in the management of rheumatoid arthritis-in vitro evaluation. Drug Deliv Transl Res 2023; 13:3030-3058. [PMID: 37294425 PMCID: PMC10624745 DOI: 10.1007/s13346-023-01372-1] [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] [Accepted: 05/20/2023] [Indexed: 06/10/2023]
Abstract
Microencapsulation of the therapeutical monoclonal antibody infliximab (INF) was investigated as an innovative approach to improve its stability and to achieve formulations with convenient features for intra-articular administration. Ultrasonic atomization (UA), a novel alternative to microencapsulate labile drugs, was compared with the conventional emulsion/evaporation method (Em/Ev) using biodegradable polymers, specifically Polyactive® 1000PEOT70PBT30 [poly(ethylene-oxide-terephthalate)/poly(butylene-terephthalate); PEOT-PBT] and its polymeric blends with poly-(D, L-lactide-co-glycolide) (PLGA) RG502 and RG503 (PEOT-PBT:PLGA; 65:35). Six different formulations of spherical core-shell microcapsules were successfully developed and characterized. The UA method achieved a significantly higher encapsulation efficiency (69.7-80.25%) than Em/Ev (17.3-23.0%). Mean particle size, strongly determined by the microencapsulation method and to a lesser extent by polymeric composition, ranged from 26.6 to 49.9 µm for UA and 1.5-2.1 µm for Em/Ev. All formulations demonstrated sustained INF release in vitro for up to 24 days, with release rates modulated by polymeric composition and microencapsulation technique. Both methods preserved INF biological activity, with microencapsulated INF showing higher efficacy than commercial formulations at comparable doses regarding bioactive tumor necrosis factor-alpha (TNF-α) neutralization according to WEHI-13VAR bioassay. Microparticles' biocompatibility and extensive internalization by THP-1-derived macrophages was demonstrated. Furthermore, high in vitro anti-inflammatory activity was achieved after treatment of THP-1 cells with INF-loaded microcapsules, significatively reducing in vitro production of TNF-α and interleucine-6 (Il-6).
Collapse
Affiliation(s)
- Iván Lamela-Gómez
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Faculty of Sciences, Universidade de Santiago de Compostela, Campus Terra, 27002, Lugo, Spain
- Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Lídia M Gonçalves
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - António J Almeida
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Asteria Luzardo-Álvarez
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Faculty of Sciences, Universidade de Santiago de Compostela, Campus Terra, 27002, Lugo, Spain.
- Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain.
| |
Collapse
|
12
|
Oo WM, Hunter DJ. Efficacy, Safety, and Accuracy of Intra-articular Therapies for Hand Osteoarthritis: Current Evidence. Drugs Aging 2023; 40:1-20. [PMID: 36633823 DOI: 10.1007/s40266-022-00994-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2022] [Indexed: 01/13/2023]
Abstract
The lifetime risk of symptomatic hand osteoarthritis (OA) is 39.8%, with one in two women and one in four men developing the disease by age 85 years and no disease-modifying drug (DMOAD) available so far. Intra-articular (IA) therapy is one of the options commonly used for symptomatic alleviation of OA disease as it can circumvent systemic exposure and potential side effects of oral medications. The current narrative review focuses on the efficacy and safety profiles of the currently available IA agents in hand OA (thumb-base OA or interphalangeal OA) such as corticosteroids and hyaluronic acid (HA), as well as the efficacy and safety of IA investigational injectates in phase 2/3 clinical trials such as prolotherapy, platelet-rich plasma, stem cells, infliximab, interferon-? and botulinum toxin, based on the published randomized controlled trials on PubMed database. The limited published literature revealed the short-term symptomatic benefits of corticosteroids in interphalangeal OA while long-term data are lacking. Most of the short-term studies showed no significant difference between corticosteroids and hyaluronic acid in thumb-base OA, usually with a faster onset of pain relief in the corticosteroid group and a slower but greater (statistically insignificant) pain improvement in the HA group. The majority of studies in investigational agents were limited by small sample size, short-term follow-up, and presence of serious side effects. In addition, we reported higher accuracy rates of drug administrations under imaging guidance than landmark guidance (blind method), and then briefly describe challenges for the long-term efficacy and prospects of IA therapeutics.
Collapse
Affiliation(s)
- Win Min Oo
- Department of Physical Medicine and Rehabilitation, Mandalay General Hospital, University of Medicine, Mandalay, Mandalay, Myanmar.
- Rheumatology Department, Faculty of Medicine and Health, Royal North Shore Hospital, and Sydney Musculoskeletal Health, Kolling Institute, The University of Sydney, Sydney, NSW, Australia.
| | - David J Hunter
- Rheumatology Department, Faculty of Medicine and Health, Royal North Shore Hospital, and Sydney Musculoskeletal Health, Kolling Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
13
|
Tenti S, Bruyère O, Cheleschi S, Reginster JY, Veronese N, Fioravanti A. An update on the use of conventional and biological disease-modifying anti-rheumatic drugs in hand osteoarthritis. Ther Adv Musculoskelet Dis 2023; 15:1759720X231158618. [PMID: 36937821 PMCID: PMC10017945 DOI: 10.1177/1759720x231158618] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/02/2023] [Indexed: 03/15/2023] Open
Abstract
Osteoarthritis (OA) is a highly prevalent condition worldwide associated with pain, progressive disability, reduced participation in social activities, and impaired quality of life. Despite its growing burden, the therapeutic options are still limited and almost exclusively addressed to symptoms' management, while no disease-modifying OA drugs able to prevent or retard disease progression are actually available. For these reasons, in the last decades, relevant efforts to find new potential therapeutic targets in OA have been made and a number of existing conventional and biological disease-modifying anti-rheumatic drugs (DMARDs), including hydroxychloroquine (HCQ), methotrexate (MTX), tumor necrosis factor (TNF)-α, interleukin (IL)-1, and IL-6 inhibitors, commonly used to treat inflammatory rheumatic diseases, have been repurposed for the treatment of OA and explored also in hand osteoarthritis (HOA). The current narrative review was aimed to provide a comprehensive and updated understanding of the possibilities and the criticisms related to the treatment of HOA with conventional and biological DMARDs. Unfortunately, therapy with conventional and biologic drugs in HOA has not achieved the expected success, despite a rationale for their use exists. Thus, our findings outline the urgent need to enhance the exploration of HOA basic molecular mechanisms to find new potential therapeutic targets, personalized for each patient, and appropriate for the different subsets of HOA and for the different phases of disease.
Collapse
Affiliation(s)
- Sara Tenti
- Clinic for the Diagnosis and Management of Hand Osteoarthritis, Rheumatology Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Viale Bracci 1, 53100 Siena, Italy
| | - Olivier Bruyère
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Centre for Public Health, Aspects of Musculoskeletal Health and Ageing, University of Liege, Liege, Belgium
| | - Sara Cheleschi
- Clinic for the Diagnosis and Management of Hand Osteoarthritis, Rheumatology Unit, Department of Medicine, Surgery and Neuroscience, Azienda ospedaliero-universitaria Senese, Siena, Italy
| | - Jean-Yves Reginster
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Centre for Public Health, Aspects of Musculoskeletal Health and Ageing, University of Liege, Liege, Belgium
| | - Nicola Veronese
- Geriatric Unit, Department of Internal Medicine and Geriatrics, University of Palermo, Palermo, Italy
| | | |
Collapse
|