1
|
Lee S, Kim B, Lee MJ, Kim D, Park S, Kim J, Arai Y, Lee SH. Curcumin-PLGA NPs coated with targeting biomimetic personalized stem cell membranes for osteoarthritis therapy. J Control Release 2025; 381:113625. [PMID: 40081740 DOI: 10.1016/j.jconrel.2025.113625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Traditional drug delivery systems for OA treatments face limitations due to rapid clearance within the joint and low biocompatibility. Moreover, the inflammation associated with OA exacerbates tissue damage and delays the regenerative capacity of therapeutics. To overcome these limitations, an OA-specific drug delivery system designated dCOL2-CM-Cur-PNPs is developed herein to target OA cartilage for anti-inflammatory and cartilage regeneration purposes. This system is constructed using cell membranes obtained from induced pluripotent stem cell -derived mesenchymal stem cells (iMSC-CMs), poly(D,l-lactide-co-glycolide) (PLGA) nanoparticles loaded with the well-known anti-inflammatory and cartilage-regenerating agent curcumin (Cur-PNPs), and damaged type II collagen (dCOL2)-targeting phospholipids. Coating the Cur-PNPs with iMSC-CMs enhances the sustained release of curcumin and improves its cellular uptake by OA-induced chondrocytes. The dCOL2-CM-Cur-PNPs restores the chondrogenic properties of the OA-induced chondrocytes, inhibit the pro-inflammatory function of M1 macrophages, and promote the anti-inflammatory function of M2 macrophages. The dCOL2-targeting phospholipids integrated on the surface of the iMSC-CMs facilitate specific binding to OA cartilage, as validated by in-vitro and in-vivo experiments. Additionally, the dCOL2-CM-Cur-PNPs alleviate OA progression in a DMM rat model. This drug delivery system based on iMSC-CMs modified with dCOL2-targeting phospholipids demonstrates significant potential as a next-generation platform for promoting cartilage regeneration through OA-specific therapy.
Collapse
Affiliation(s)
- Sunjun Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Bowon Kim
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Min-Ju Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Deogil Kim
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Sunghyun Park
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Jinsik Kim
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea.
| |
Collapse
|
2
|
Mitsou E, Klein J. Liposome-Based Interventions in Knee Osteoarthritis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410060. [PMID: 40143645 PMCID: PMC12036560 DOI: 10.1002/smll.202410060] [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: 10/27/2024] [Revised: 01/21/2025] [Indexed: 03/28/2025]
Abstract
Osteoarthritis (OA) is the most common degenerative disease of the joints, causing significant disability and socio-economic burden in the aging population. Simultaneously, however, it is a common occurrence in younger individuals, initiated by joint injuries or obesity alongside other factors. Intravenous and oral pharmaceutical OA management have both been associated with systemic adverse effects, thereby resulting in a growing interest in intra-articular (IA) treatment. IA-administered drugs circumvent the requirement for high dosage, offering immediate access to the site of interest while minimizing any unfavorable effects. Nonetheless, IA-injected drugs, administered in their free form, present low retention time in the knee joint raising the need for multiple injection dosage regimens, while their capability to target the cartilage or specific cell populations is limited. Liposomes, due to their unique characteristics and tunable nature, have proven to be excellent candidates for the management of knee OA. This review explores the last decade's research on the efficacy of various IA liposomal formulations, investigating their multifaceted properties as pharmaceutical carriers, lubricating agents, and a basis for combinatorial approaches paving the way to novel treatment solutions for OA.
Collapse
Affiliation(s)
- Evgenia Mitsou
- Department of Molecular Chemistry and Materials ScienceWeizmann Institute of ScienceHertzl Street 234Rehovot7610001Israel
- Present address:
Institute of Chemical BiologyNational Hellenic Research Foundation48, Vassileos Constantinou Ave.Athens11635Greece
| | - Jacob Klein
- Department of Molecular Chemistry and Materials ScienceWeizmann Institute of ScienceHertzl Street 234Rehovot7610001Israel
| |
Collapse
|
3
|
Del Río E. Rethinking Osteoarthritis Management: Synergistic Effects of Chronoexercise, Circadian Rhythm, and Chondroprotective Agents. Biomedicines 2025; 13:598. [PMID: 40149577 PMCID: PMC11940269 DOI: 10.3390/biomedicines13030598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
Osteoarthritis (OA) is a chronic and debilitating joint disease characterized by progressive cartilage degeneration for which no definitive cure exists. Conventional management approaches often rely on fragmented and poorly coordinated pharmacological and non-pharmacological interventions that are inconsistently applied throughout the disease course. Persistent controversies regarding the clinical efficacy of chondroprotective agents, frequently highlighted by pharmacovigilance agencies, underscore the need for a structured evidence-based approach. Emerging evidence suggests that synchronizing pharmacotherapy and exercise regimens with circadian biology may optimize therapeutic outcomes by addressing early pathological processes, including low-grade inflammation, oxidative stress, and matrix degradation. Recognizing the influence of the chondrocyte clock on these processes, this study proposes a 'prototype' for a novel framework that leverages the circadian rhythm-aligned administration of traditional chondroprotective agents along with tailored, accessible exercise protocols to mitigate cartilage breakdown and support joint function. In addition, this model-based framework emphasizes the interdependence between cartilage chronobiology and time-of-day-dependent responses to exercise, where strategically timed joint activity enhances nutrient and waste exchange, mitigates mitochondrial dysfunction, supports cellular metabolism, and promotes tissue maintenance, whereas nighttime rest promotes cartilage rehydration and repair. This time-sensitive, comprehensive approach aims to slow OA progression, reduce structural damage, and delay invasive procedures, particularly in weight-bearing joints such as the knee and hip. However, significant challenges remain, including inter-individual variability in circadian rhythms, a lack of reliable biomarkers for pharmacotherapeutic monitoring, and limited clinical evidence supporting chronoexercise protocols. Future large-scale, longitudinal trials are critical to evaluate the efficacy and scalability of this rational integrative strategy, paving the way for a new era in OA management.
Collapse
|
4
|
Chen Y, Zhang Y, Wu C, Zhao X, Zhang H, Li C, Deng Y, Sun L, Zhou Y, Zhang X. High-Throughput Screening Strategy and Metal-Organic Framework-Based Multifunctional Controlled-Release Nanomaterial for Osteoarthritis Therapy. ACS NANO 2025; 19:4802-4819. [PMID: 39829021 DOI: 10.1021/acsnano.4c15740] [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: 01/22/2025]
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease that lacks effective therapy. Oxidative stress is one of the major factors contributing to OA; however, treatments targeting oxidative stress are still lacking. In the current study, we established an oxidative stress-induced cell death model in chondrocytes in vitro and screened drugs that may suppress oxidative stress-induced cell death. Ethyl gallate (EG) was identified as the most potent drug against oxidative stress-induced cell death out of more than 600 drugs in the natural product library. Application of drugs without an appropriate delivery system for OA therapy may have drawbacks such as low bioavailability, short action time, and poor efficacy. Herein, poly-His6-zinc assembly (PZA), a pH-responsive metal-organic framework (MOF) loaded with EG (EG@PZA) was designed for OA therapy. It was demonstrated that EG@PZA may have the lysosome escape property, which dramatically increases the utilization of EG. Furthermore, EG@PZA showed enhanced release capability of EG in the acidic microenvironment. In vitro and in vivo studies demonstrated that EG@PZA effectively suppresses oxidative stress-induced extracellular matrix degradation, ferroptosis, and senescence in chondrocytes and also ameliorates OA in the destabilization of the medial meniscus (DMM) mouse model in vivo. Together, the current study showed that EG@PZA may become a potential controlled-release nanomaterial for effective OA therapy.
Collapse
Affiliation(s)
- Yu Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yekai Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chenyu Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaoying Zhao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Hanwen Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chenchao Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yuxin Deng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Liaojun Sun
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yifei Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
5
|
Zhang Z, Wang G, Zhang Z, Liang X, Wang G, Xu M, Yang X, Zhong X, Li C, Zhou M. Locally administered liposomal drug depot enhances rheumatoid arthritis treatment by inhibiting inflammation and promoting cartilage repair. J Nanobiotechnology 2025; 23:69. [PMID: 39891123 PMCID: PMC11783794 DOI: 10.1186/s12951-025-03110-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: 08/17/2024] [Accepted: 01/11/2025] [Indexed: 02/03/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune condition characterized by synovial hyperplasia, where inflammatory macrophages within the joint synovium produce multiple inflammatory cytokines, leading to cartilage damage. The development of therapeutic strategies that combine anti-inflammatory effects and cartilage repair mechanisms holds great promise for effective RA treatment. To address the limitations associated with the off-target effects of intravenous administration and the risk of synovial cavity infection with repeated local injections, we have innovatively developed a liposomal drug depot through hyaluronic acid (HA)-modified liposomes encapsulating dexamethasone sodium phosphate (DSP)-loaded nanogels, termed HA-Lipo@G/D. The nanogels were prepared by ionic cross-linking of chondroitin sulfate and gelatin, both of which have notable cartilage repair properties. In vitro studies demonstrated that this formulation exhibited sustained drug release, enhanced uptake by inflammatory macrophages, reduced secretion of inflammatory factors (TNF-α, IL-1β), and significantly decreased chondrocyte apoptosis induced by inflammatory factors. Moreover, in vivo assessments in a rat model of collagen-induced arthritis revealed effective accumulation of the liposomal drug depot at the inflamed joint site, resulting in macrophage repolarization and cartilage tissue repair. Our findings provide a synergistic strategy for inhibiting inflammation and mitigating cartilage damage through local joint cavity injection, thereby enhancing the therapeutic efficacy of RA.
Collapse
Affiliation(s)
- Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Guan Wang
- Department of Orthopaedics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Zhuo Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Key Laboratory of Luzhou City for Aging Medicine, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Guoshuang Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Maochang Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiaolin Zhong
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
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
|
Lin X, Zhang Y, Li J, Oliver BG, Wang B, Li H, Yong KT, Li JJ. Biomimetic multizonal scaffolds for the reconstruction of zonal articular cartilage in chondral and osteochondral defects. Bioact Mater 2025; 43:510-549. [PMID: 40115881 PMCID: PMC11923379 DOI: 10.1016/j.bioactmat.2024.10.001] [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: 05/30/2024] [Revised: 09/02/2024] [Accepted: 10/01/2024] [Indexed: 03/23/2025] Open
Abstract
Chondral and osteochondral injuries are frequently encountered in clinical practice. However, articular cartilage has limited self-healing capacity due to its sophisticated zonal structure and avascular nature, introducing significant challenges to the restoration of chondral and osteochondral tissues after injury. Improperly repaired articular cartilage can lead to irreversible joint damage and increase the risk of osteoarthritis progression. Cartilage tissue engineering using stratified scaffolds with multizonal design to match the zonal structure of articular cartilage may help to meet the complex regeneration requirements of chondral and osteochondral tissues, and address the drawbacks experienced with single-phase scaffolds. Navigating the heterogeneity in matrix organisation and cellular composition across cartilage zones is a central consideration in multizonal scaffold design. With emphasis on recent advances in scaffold design and fabrication strategies, this review captures emerging approaches on biomimetic multizonal scaffolds for the reconstruction of zonal articular cartilage, including strategies on replicating native tissue structure through variations in fibre orientation, porous structure, and cell types. Exciting progress in this dynamic field has highlighted the tremendous potential of multizonal scaffolding strategies for regenerative medicine in the recreation of functional tissues.
Collapse
Affiliation(s)
- Xiaoqi Lin
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, NSW, 2007, Australia
| | - Ye Zhang
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, 2007, Australia
| | - Jiarong Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, NSW, 2007, Australia
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, 2007, Australia
- Woolcock Institute of Medical Research, Macquarie University, Macquarie Park, NSW, 2113, Australia
| | - Bin Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Haiyan Li
- Chemical and Environmental Engineering Department, School of Engineering, STEM College, RMIT University, Melbourne, VIC, 3000, Australia
| | - Ken-Tye Yong
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, NSW, 2007, Australia
- Woolcock Institute of Medical Research, Macquarie University, Macquarie Park, NSW, 2113, Australia
| |
Collapse
|
8
|
Liu X, Chen X, Fei Y, Zhang J, Yue O, Wang X, Jiang H. Locally Injectable, ROS-Scavenging, and ROS-/pH-Responsive Polymeric-Micelles-Embedded Hydrogels for Precise Minimally Invasive and Long-Lasting Rheumatoid Therapy. Adv Healthc Mater 2025; 14:e2403579. [PMID: 39629502 DOI: 10.1002/adhm.202403579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/16/2024] [Indexed: 01/29/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by synovitis, bone-erosion, and joint-destruction. Here, we developed a locally injectable, ROS-scavenging, and ROS-/pH-responsive drug-delivery platform (HC@PTM) by bio-compositing of aldolizing hyaluronic acid (HA) crosslinked with chitosan (CS), and ROS-triggered/eliminated micelles (PTM) coupled with the drug methotrexate(MTX). The PTM efficiently eradicate excessive ROS in RA-joints, precisely triggering drug-release within inflamed arthritic-sites and further confer exceptional antioxidant origins to HC@PTM. HC@PTM with outstanding shape-adaptability and self-repairing properties effectively conformed to irregular articular cartilage while resisting joint-induced deformations. Further, the platform's pH-responsive nature enables on-demand drug-release within acidic inflamed synovium, serving as a drug-reservoir for precise and sustained therapeutic effects. Extensive in vitro and in vivo investigations confirm HC@PTM's ability to induce M2 macrophage polarization, downregulate inflammatory factor expression, and ameliorate the RA-microenvironment, ultimately achieving synergistic therapeutic outcomes. This study represents significant advancements in precise and long-term RA-treatment through a minimally invasive approach, offering potential strategies for novel precision medicine.
Collapse
Affiliation(s)
- Xinhua Liu
- Institute of Biomass & Functional Materials, Shaanxi University of Science &Technology, Xi'an, 710021, China
| | - Xing Chen
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
- Institute of Biomass & Functional Materials, Shaanxi University of Science &Technology, Xi'an, 710021, China
| | - Yifan Fei
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
- Institute of Biomass & Functional Materials, Shaanxi University of Science &Technology, Xi'an, 710021, China
| | - Jiamin Zhang
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
- Institute of Biomass & Functional Materials, Shaanxi University of Science &Technology, Xi'an, 710021, China
| | - Ouyang Yue
- Institute of Biomass & Functional Materials, Shaanxi University of Science &Technology, Xi'an, 710021, China
| | - Xuechuan Wang
- Institute of Biomass & Functional Materials, Shaanxi University of Science &Technology, Xi'an, 710021, China
| | - Huie Jiang
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| |
Collapse
|
9
|
Martínez-Borrajo R, Rouco H, Virzì NF, Diaz-Rodriguez P, Landin M. Modulation of IFN-γ induced macrophage inflammatory responses via indomethacin-loaded NLCs for OA management. Int J Pharm 2024; 666:124823. [PMID: 39396655 DOI: 10.1016/j.ijpharm.2024.124823] [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: 07/21/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Macrophages are the main cells present in the synovial membrane. They play an important role in the development and progression of osteoarthritis (OA). After the establishment of the disease macrophages mostly adopt a pro-inflammatory secretory phenotype (OA phenotype) further inducing cartilage degradation. Indomethacin (IND) is a non-steroidal anti-inflammatory drug (NSAID) able to inhibit the synthesis of prostaglandins mediated by both cyclooxygenase isoforms depicting a potent anti-inflammatory capacity. However, the lack of specificity and short half-like of free drugs within the joint cavity limits its utility in controlling inflammation after intra-articular administration. This study aims at developing IND loaded glycosylated nanostructured lipid carriers (NLCs) to selectively target macrophages and promote their reprogramming to an anti-inflammatory phenotype. This approach focused on the local administration of the NLCs, offers a promising therapeutic strategy for treating OA by modulating the inflammatory environment within the joint. NLCs will be designed by combining experimental and in silico docking analyses, and thoroughly characterized to obtain drug delivery systems with high stability and suitable physicochemical properties. The proposed mannose-functionalized systems exhibited adequate particle sizes (≈ 70 nm) and positive surface charges (> 20 mV) to be efficiently retained in the joint cavity. Moreover, the developed NLCs demonstrated effective and specific uptake by OA-like macrophages leading to a significant decrease in the secretion of the pro-inflammatory cytokines IL-6, IL-8 and TNF-α similarly to the free drug. Therefore, these systems effectively reprogrammed OA-associated macrophages to adopt a more regenerative phenotype, offering a promising strategy for managing inflammation in OA.
Collapse
Affiliation(s)
- Rebeca Martínez-Borrajo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Grupo I+D Farma (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; Instituto de Materiais da Universidade de Santiago de Compostela (iMATUS), 15782 Santiago de Compostela, Spain
| | - Helena Rouco
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Grupo I+D Farma (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; Instituto de Materiais da Universidade de Santiago de Compostela (iMATUS), 15782 Santiago de Compostela, Spain
| | - Nicola Filippo Virzì
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Grupo I+D Farma (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Department of Drug and Health Science, University of Catania, Viale A. Doria 6, 95125 Catania, Italy; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; Instituto de Materiais da Universidade de Santiago de Compostela (iMATUS), 15782 Santiago de Compostela, Spain
| | - Patricia Diaz-Rodriguez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Grupo I+D Farma (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; Instituto de Materiais da Universidade de Santiago de Compostela (iMATUS), 15782 Santiago de Compostela, Spain.
| | - Mariana Landin
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Grupo I+D Farma (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; Instituto de Materiais da Universidade de Santiago de Compostela (iMATUS), 15782 Santiago de Compostela, Spain
| |
Collapse
|
10
|
Pouyanfar N, Anvari Z, Davarikia K, Aftabi P, Tajik N, Shoara Y, Ahmadi M, Ayyoubzadeh SM, Shahbazi MA, Ghorbani-Bidkorpeh F. Machine learning-assisted rheumatoid arthritis formulations: A review on smart pharmaceutical design. MATERIALS TODAY COMMUNICATIONS 2024; 41:110208. [DOI: 10.1016/j.mtcomm.2024.110208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
Yi X, Leng P, Wang S, Liu L, Xie B. Functional Nanomaterials for the Treatment of Osteoarthritis. Int J Nanomedicine 2024; 19:6731-6756. [PMID: 38979531 PMCID: PMC11230134 DOI: 10.2147/ijn.s465243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/15/2024] [Indexed: 07/10/2024] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease, affecting more than 595 million people worldwide. Nanomaterials possess superior physicochemical properties and can influence pathological processes due to their unique structural features, such as size, surface interface, and photoelectromagnetic thermal effects. Unlike traditional OA treatments, which suffer from short half-life, low stability, poor bioavailability, and high systemic toxicity, nanotherapeutic strategies for OA offer longer half-life, enhanced targeting, improved bioavailability, and reduced systemic toxicity. These advantages effectively address the limitations of traditional therapies. This review aims to inspire researchers to develop more multifunctional nanomaterials and promote their practical application in OA treatment.
Collapse
Affiliation(s)
- Xinyue Yi
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, People’s Republic of China
| | - Pengyuan Leng
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| | - Supeng Wang
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, People’s Republic of China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| | - Bingju Xie
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
12
|
Saeedi T, Prokopovich P. Screening of poly-beta amino ester coated emulsion of ketorolac for cartilage delivery. J Mater Chem B 2024; 12:5930-5939. [PMID: 38804519 PMCID: PMC11186506 DOI: 10.1039/d4tb00313f] [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] [Received: 02/15/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Osteoarthritis (OA) is a prevalent chronic health condition necessitating effective treatment strategies. Globally, there were 86 million people with incident knee osteoarthritis in 2020. Pain management remains the primary approach to OA as the nature of cartilage poses challenges for drug delivery. An emulsion-based delivery system, using a class of positively charged and hydrolysable polymers (poly-beta-amino-esters) to coat oil droplets containing drugs, has been shown to enhance and prolong drug localization in ex vivo cartilage models. As the properties of the polymers used in this technology strongly depend on the monomers used in the synthesis, this study presents the screening of a wide range of PBAEs as droplet coating agents and using ketorolac as a model of nonsteroidal anti-inflammatory drugs. The emulsions prepared with this PBAE library were characterized, and drug localisation and retention were evaluated in both native and glycosaminoglycan (GAG) depleted cartilage ex vivo models. Optimal candidates were identified and tested in an ex vivo model showing the ability to protect chondrocyte cell viability and increase both GAG and collagen contents in cartilage exposed to cytokine (IL-1α) simulating acute cartilage damage. This work demonstrates the potential of PBAE coated emulsion as a delivery system for effective drug delivery in OA treatment.
Collapse
Affiliation(s)
- Tahani Saeedi
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff, UK.
| | | |
Collapse
|
13
|
Zong L, Wang Q, Sun H, Wu Q, Xu Y, Yang H, Lv S, Zhang L, Geng D. Intra-Articular Injection of PLGA/Polydopamine Core-Shell Nanoparticle Attenuates Osteoarthritis Progression. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21450-21462. [PMID: 38649157 DOI: 10.1021/acsami.3c18464] [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: 04/25/2024]
Abstract
Osteoarthritis (OA) is a common joint disease characterized by progressive cartilage degeneration. Unfortunately, currently available clinical drugs are mainly analgesics and cannot alleviate the development of OA. Kartogenin (KGN) has been found to promote the differentiation of bone marrow mesenchymal stem cells (BMSCs) into chondrocytes for the treatment of cartilage damage in early OA. However, KGN, as a small hydrophobic molecule, is rapidly cleared from the synovial fluid after intra-articular injection. This study synthesized a KGN-loaded nanocarrier based on PLGA/polydopamine core/shell structure to treat OA. The fluorescence signal of KGN@PLGA/PDA-PEG-E7 nanoparticles lasted for 4 weeks, ensuring long-term sustained release of KGN from a single intra-articular injection. In addition, the polyphenolic structure of PDA enables it to effectively scavenge reactive oxygen species, and the BMSC-targeting peptide E7 (EPLQLKM) endows KGN@PLGA/PDA-PEG-E7 NPs with an effective affinity for BMSCs. As a result, the KGN@PLGA/PDA-PEG-E7 nanoparticles could effectively induce cartilage in vitro and protect the cartilage and subchondral bone in a rat ACLT model. This therapeutic strategy could also be extended to the delivery of other drugs, targeting other tissues to treat joint diseases.
Collapse
Affiliation(s)
- Lujie Zong
- Department of Orthopaedics, The First People's Hospital of Changzhou, Soochow University, Changzhou, Jiangsu 213000, China
| | - Qing Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215000, China
| | - Houyi Sun
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong 250000, China
| | - Qian Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215000, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215000, China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215000, China
| | - Shujun Lv
- Department of Orthopedics, Hai'an People's Hospital, Hai'an, Jiangsu 226000, China
| | - Liang Zhang
- Department of Orthopaedics, Beijing Friendship Hospital, Capital Medical University, Beijing 100000, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215000, China
| |
Collapse
|
14
|
Singh H, Dan A, Kumawat MK, Pawar V, Chauhan DS, Kaushik A, Bhatia D, Srivastava R, Dhanka M. Pathophysiology to advanced intra-articular drug delivery strategies: Unravelling rheumatoid arthritis. Biomaterials 2023; 303:122390. [PMID: 37984246 DOI: 10.1016/j.biomaterials.2023.122390] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 10/29/2023] [Accepted: 11/04/2023] [Indexed: 11/22/2023]
Abstract
Rheumatoid arthritis (RA) is one of the most prevalent life-long autoimmune diseases with an unknown genesis. It primarily causes chronic inflammation, pain, and synovial joint-associated cartilage and bone degradation. Unfortunately, limited information is available regarding the etiology and pathogenesis of this chronic joint disorder. In the last few decades, an improved understanding of RA pathophysiology about key immune cells, antibodies, and cytokines has inspired the development of several anti-rheumatic drugs and biopharmaceuticals to act on RA-affected joints. However, life-long frequent systemic high doses of commercially available drugs are currently a limiting factor in the efficient management of RA. To address this issue, various single and double-barrier intra-articular drug delivery systems (IA-DDSs) such as nanocarriers, microparticles, hydrogels, and particles-hybrid hydrogel composite have been developed which can exclusively target the RA-affected joint cavity and release the precisely controlled therapeutic drug concentration for prolonged time whilst avoiding the systemic toxicity. This review provides a comprehensive overview of the pathogenesis of RA and discusses the rational design and development of biomaterials-based novel IA-DDs, ranging from conventional to advanced systems, for improved treatment of RA. Therefore, this review aims to unravel the pathophysiology of rheumatoid arthritis and explore cutting-edge IA-DD strategies exploiting biomaterials. It offers researchers a consolidated and up-to-date resource platform to analyze existing knowledge, identify research gaps, and contribute to the scientific literature.
Collapse
Affiliation(s)
- Hemant Singh
- Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, 382055, Gujarat, India; Department of Biology, Khalifa University, Main Campus, Abu Dhabi, 127788, United Arab Emirates
| | - Aniruddha Dan
- Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, 382055, Gujarat, India
| | - Mukesh Kumar Kumawat
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Vaishali Pawar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Deepak S Chauhan
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, H3C 3J7, Canada
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL- 33805, USA
| | - Dhiraj Bhatia
- Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, 382055, Gujarat, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Mukesh Dhanka
- Biological Sciences and Engineering, Indian Institute of Technology, Gandhinagar, 382055, Gujarat, India.
| |
Collapse
|
15
|
Partain BD, Bracho-Sanchez E, Farhadi SA, Yarmola EG, Keselowsky BG, Hudalla GA, Allen KD. Intra-articular delivery of an indoleamine 2,3-dioxygenase galectin-3 fusion protein for osteoarthritis treatment in male Lewis rats. Arthritis Res Ther 2023; 25:173. [PMID: 37723593 PMCID: PMC10506271 DOI: 10.1186/s13075-023-03153-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/27/2023] [Indexed: 09/20/2023] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is driven by low-grade inflammation, and controlling local inflammation may offer symptomatic relief. Here, we developed an indoleamine 2,3-dioxygenase and galectin-3 fusion protein (IDO-Gal3), where IDO increases the production of local anti-inflammatory metabolites and Gal3 binds carbohydrates to extend IDO's joint residence time. In this study, we evaluated IDO-Gal3's ability to alter OA-associated inflammation and pain-related behaviors in a rat model of established knee OA. METHODS Joint residence was first evaluated with an analog Gal3 fusion protein (NanoLuc™ and Gal3, NL-Gal3) that produces luminescence from furimazine. OA was induced in male Lewis rats via a medial collateral ligament and medial meniscus transection (MCLT + MMT). At 8 weeks, NL or NL-Gal3 were injected intra-articularly (n = 8 per group), and bioluminescence was tracked for 4 weeks. Next, IDO-Gal3s's ability to modulate OA pain and inflammation was assessed. Again, OA was induced via MCLT + MMT in male Lewis rats, with IDO-Gal3 or saline injected into OA-affected knees at 8 weeks post-surgery (n = 7 per group). Gait and tactile sensitivity were then assessed weekly. At 12 weeks, intra-articular levels of IL6, CCL2, and CTXII were assessed. RESULTS The Gal3 fusion increased joint residence in OA and contralateral knees (p < 0.0001). In OA-affected animals, both saline and IDO-Gal3 improved tactile sensitivity (p = 0.008), but IDO-Gal3 also increased walking velocities (p ≤ 0.033) and improved vertical ground reaction forces (p ≤ 0.04). Finally, IDO-Gal3 decreased intra-articular IL6 levels within the OA-affected joint (p = 0.0025). CONCLUSION Intra-articular IDO-Gal3 delivery provided long-term modulation of joint inflammation and pain-related behaviors in rats with established OA.
Collapse
Affiliation(s)
- Brittany D Partain
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Biomedical Sciences Building, Gainesville, FL, 32610, USA
| | - Evelyn Bracho-Sanchez
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Biomedical Sciences Building, Gainesville, FL, 32610, USA
| | - Shaheen A Farhadi
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Biomedical Sciences Building, Gainesville, FL, 32610, USA
| | - Elena G Yarmola
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Biomedical Sciences Building, Gainesville, FL, 32610, USA
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Biomedical Sciences Building, Gainesville, FL, 32610, USA
| | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Biomedical Sciences Building, Gainesville, FL, 32610, USA
| | - Kyle D Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Biomedical Sciences Building, Gainesville, FL, 32610, USA.
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
16
|
Partain BD, Bracho-Sanchez E, Farhadi SA, Yarmola EG, Keselowsky BG, Hudalla GA, Allen KD. Intra-Articular Delivery of an Indoleamine 2,3-Dioxygenase Galectin-3 Fusion Protein for Osteoarthritis Treatment in Male Lewis Rats. RESEARCH SQUARE 2023:rs.3.rs-2753443. [PMID: 37131836 PMCID: PMC10153358 DOI: 10.21203/rs.3.rs-2753443/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Objective : Controlling joint inflammation can improve osteoarthritis (OA) symptoms; however, current treatments often fail to provide long-term effects. We have developed an indoleamine 2,3-dioxygenase and galectin-3 fusion protein (IDO-Gal3). IDO converts tryptophan to kynurenines, directing the local environment toward an anti-inflammatory state; Gal3 binds carbohydrates and extends IDO's joint residence time. In this study, we evaluated IDO-Gal3's ability to alter OA-associated inflammation and pain-related behaviors in a rat model of established knee OA. Methods : Joint residence was first evaluated with an analog Gal3 fusion protein (NanoLuc™ and Gal3, NL-Gal3) that produces luminescence from furimazine. OA was induced in male Lewis rats via a medial collateral ligament and medial meniscus transection (MCLT+MMT). At 8 weeks, NL or NL-Gal3 were injected intra-articularly (n=8 per group), and bioluminescence was tracked for 4 weeks. Next, IDO-Gal3's ability to modulate OA pain and inflammation was assessed. Again, OA was induced via MCLT+MMT in male Lewis rats, with IDO-Gal3 or saline injected into OA-affected knees at 8 weeks post-surgery (n=7 per group). Gait and tactile sensitivity were then assessed weekly. At 12 weeks, intra-articular levels of IL6, CCL2, and CTXII were assessed. Results : The Gal3 fusion increased joint residence in OA and contralateral knees (p<0.0001). In OA-affected animals, IDO-Gal3 improved tactile sensitivity (p=0.002), increased walking velocities (p≤0.033), and improved vertical ground reaction forces (p≤0.04). Finally, IDO-Gal3 decreased intra-articular IL6 levels within the OA-affected joint (p=0.0025). Conclusion : Intra-articular IDO-Gal3 delivery provided long-term modulation of joint inflammation and pain-related behaviors in rats with established OA.
Collapse
|
17
|
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
|
18
|
Avendaño-Godoy J, Poblete MG, Ramos RG, Gómez-Gaete C. Evaluation of the Effects of Gamma Radiation Sterilization on Rhein-Loaded Biodegradable Microparticles for the Treatment of Osteoarthritis. J Pharm Sci 2023; 112:837-843. [PMID: 36347357 DOI: 10.1016/j.xphs.2022.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/02/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
In previous work, we reported on the design of biodegradable rhein-loaded PLGA microparticles for the treatment of osteoarthritis. Considering that a formulation designed for intra-articular administration must meet sterility requirements to guarantee its safety, in this study the effect of gamma radiation sterilization on these microparticles was evaluated. The size, morphology, and surface characteristics of the microparticles and the encapsulation efficiency of rhein were not affected by the sterilization process. Although DSC and PXRD analyses suggested otherwise, rhein release profiles were not altered by gamma radiation. The release of rhein from the microparticles was fitted to a Gompertz model. In conclusion, the results of this study suggest that gamma radiation is a suitable method for the sterilization of rhein-loaded PLGA microparticles to enable their intra-articular administration in order to provide a therapeutic solution to patients suffering from chronic joint diseases.
Collapse
Affiliation(s)
- Javier Avendaño-Godoy
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Maria Gabriela Poblete
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Ricardo Godoy Ramos
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Carolina Gómez-Gaete
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
19
|
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: 0] [Impact Index Per Article: 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
|
20
|
Bruno MC, Cristiano MC, Celia C, d'Avanzo N, Mancuso A, Paolino D, Wolfram J, Fresta M. Injectable Drug Delivery Systems for Osteoarthritis and Rheumatoid Arthritis. ACS NANO 2022; 16:19665-19690. [PMID: 36512378 DOI: 10.1021/acsnano.2c06393] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Joint diseases are one of the most common causes of morbidity and disability worldwide. The main diseases that affect joint cartilage are osteoarthritis and rheumatoid arthritis, which require chronic treatment focused on symptomatic relief. Conventional drugs administered through systemic or intra-articular routes have low accumulation and/or retention in articular cartilage, causing dose-limiting toxicities and reduced efficacy. Therefore, there is an urgent need to develop improved strategies for drug delivery, in particular, the use of micro- and nanotechnology-based methods. Encapsulation of therapeutic agents in delivery systems reduces drug efflux from the joint and protects against rapid cellular and enzymatic clearance following intra-articular injection. Consequently, the use of drug delivery systems decreases side effects and increases therapeutic efficacy due to enhanced drug retention in the intra-articular space. Additionally, the frequency of intra-articular administration is reduced, as delivery systems enable sustained drug release. This review summarizes various advanced drug delivery systems, such as nano- and microcarriers, developed for articular cartilage diseases.
Collapse
Affiliation(s)
- Maria Chiara Bruno
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Maria Chiara Cristiano
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307, Kaunas, Lithuania
| | - Nicola d'Avanzo
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
| | - Antonia Mancuso
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Massimo Fresta
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| |
Collapse
|
21
|
Emulsion Gel: a Dual Drug Delivery Platform for Osteoarthritis Treatment. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00282-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
22
|
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]
|
23
|
Hall ME, Wang AS, Gold GE, Levenston ME. Contrast solution properties and scan parameters influence the apparent diffusivity of computed tomography contrast agents in articular cartilage. J R Soc Interface 2022; 19:20220403. [PMID: 35919981 PMCID: PMC9346352 DOI: 10.1098/rsif.2022.0403] [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: 05/27/2022] [Accepted: 07/08/2022] [Indexed: 11/12/2022] Open
Abstract
The inability to detect early degenerative changes to the articular cartilage surface that commonly precede bulk osteoarthritic degradation is an obstacle to early disease detection for research or clinical diagnosis. Leveraging a known artefact that blurs tissue boundaries in clinical arthrograms, contrast agent (CA) diffusivity can be derived from computed tomography arthrography (CTa) scans. We combined experimental and computational approaches to study protocol variations that may alter the CTa-derived apparent diffusivity. In experimental studies on bovine cartilage explants, we examined how CA dilution and transport direction (absorption versus desorption) influence the apparent diffusivity of untreated and enzymatically digested cartilage. Using multiphysics simulations, we examined mechanisms underlying experimental observations and the effects of image resolution, scan interval and early scan termination. The apparent diffusivity during absorption decreased with increasing CA concentration by an amount similar to the increase induced by tissue digestion. Models indicated that osmotically-induced fluid efflux strongly contributed to the concentration effect. Simulated changes to spatial resolution, scan spacing and total scan time all influenced the apparent diffusivity, indicating the importance of consistent protocols. With careful control of imaging protocols and interpretations guided by transport models, CTa-derived diffusivity offers promise as a biomarker for early degenerative changes.
Collapse
Affiliation(s)
- Mary E. Hall
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Adam S. Wang
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Garry E. Gold
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Marc E. Levenston
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University, Stanford, CA, USA
| |
Collapse
|
24
|
Pape E, Pinzano A, Henrionnet C, Scala-Bertola J, Gillet P, Gambier N. Rat synovial tissue and blood rapamycin pharmacokinetics after intra-articular injection of free solution or nanoparticles vs free rapamycin intravenous shot. Int J Pharm 2022; 624:122026. [PMID: 35863592 DOI: 10.1016/j.ijpharm.2022.122026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/09/2022] [Accepted: 07/13/2022] [Indexed: 10/17/2022]
Abstract
Intra-articular (IA) injection of a chondroprotective candidate may delay the osteoarthritis (OA) course, but its rapid absorption into systemic circulation may limit efficacy and produce untoward effects. We compared the pharmacokinetics (PK) of IA rapamycin injected as sustained release in nanoparticles (NPs) versus a free rapamycin suspension in the rat knee compared to an intravenous (IV) free rapamycin shot taken as a reference. Rats received either a single IV injection of free rapamycin (10 µM) or an IA of free or NPs-loaded rapamycin. After sequential exsanguination (15, 30, 60, 180, 360 min, D1, and D7), knee synovial tissue (ST) and cartilage histology were performed. Blood and ST concentrations (LC-MS/MS), PK parameters (area under the curve: AUC; mean residence time: MRT; elimination half-life: T1/2), and IA biocompatibility were assessed. AUCIV was significantly higher for IV than for both IA injections (AUCIA free and AUCIA NPs), with 4248 vs 28 and 74 µg.min.L-1. For ST parameters, we observed a significant difference between AUCIA free and AUCIA NPs with 3735 and 10513 µg.min.L-1 correspondingly. Articular T1/2 and MRT were higher after NPs than after free rapamycin injection: 57.8 and 5.0 h for T1/2 and 80.6 and 5.5 h for MRT, respectively. Histological analysis revealed no chondral injuries and slight transient synovitis only 3 h after the administration of NPs. In the rat knee, rapamycin-loaded NPs delivery via a single IA injection is biocompatible and prolongs synovium joint residency, diminishes blood levels, and reduces detrimental systemic exposure.
Collapse
Affiliation(s)
- Elise Pape
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie-Toxicologie, Pharmacovigilance & CEIPA, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Astrid Pinzano
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France.
| | | | - Julien Scala-Bertola
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie-Toxicologie, Pharmacovigilance & CEIPA, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Pierre Gillet
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie-Toxicologie, Pharmacovigilance & CEIPA, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Nicolas Gambier
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie-Toxicologie, Pharmacovigilance & CEIPA, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Co-formulations of adalimumab with hyaluronic acid / polyvinylpyrrolidone to combine intraarticular drug delivery and viscosupplementation. Eur J Pharm Biopharm 2022; 177:39-49. [PMID: 35691537 DOI: 10.1016/j.ejpb.2022.06.002] [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: 02/01/2022] [Revised: 05/01/2022] [Accepted: 06/06/2022] [Indexed: 11/24/2022]
Abstract
Polymer-based formulations present an attractive strategy in intraarticular drug-delivery to refrain biologicals from early leakage from the joint. In this study, co-formulations of hyaluronic acid and polyvinylpyrrolidone were investigated for their potential as viscosupplements and their influence on the transsynovial loss of adalimumab. For this purpose, polymer mixtures were evaluated for their viscosity and elasticity behavior while their influence on the permeation of adalimumab across a porcine ex-vivo synovial membrane was determined. Hyaluronic acid showed strong shear thinning behavior and exhibited high viscosity and elasticity at low motions, while combinations with polyvinylpyrrolidone provided absorption and stiffness at high mechanical stress, so that they can potentially restore the rheological properties of the synovial fluid over the range of joint motion. In addition, the formulations showed significant influence on transsynovial permeation kinetics of adalimumab and hyaluronic acid, which could be decelerated up to 5- and 3-fold, respectively. Besides viscosity effects, adalimumab was retained primarily by an electrostatic interaction with hyaluronic acid, as detected by isothermal calibration calorimetry. Furthermore, polymer-mediated stabilization of the antibody activity was detected. In summary, hyaluronic acid - polyvinylpyrrolidone combinations can be efficiently used to prolong the residence of adalimumab in the joint cavity while simultaneously supplying viscosupplementation.
Collapse
|
28
|
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]
|
29
|
Simón-Vázquez R, Tsapis N, Lorscheider M, Rodríguez A, Calleja P, Mousnier L, de Miguel Villegas E, González-Fernández Á, Fattal E. Improving dexamethasone drug loading and efficacy in treating arthritis through a lipophilic prodrug entrapped into PLGA-PEG nanoparticles. Drug Deliv Transl Res 2022; 12:1270-1284. [PMID: 34993924 PMCID: PMC8734546 DOI: 10.1007/s13346-021-01112-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2021] [Indexed: 01/01/2023]
Abstract
Targeted delivery of dexamethasone to inflamed tissues using nanoparticles is much-needed to improve its efficacy while reducing side effects. To drastically improve dexamethasone loading and prevent burst release once injected intravenously, a lipophilic prodrug dexamethasone palmitate (DXP) was encapsulated into poly(DL-lactide-co-glycolide)-polyethylene glycol (PLGA-PEG) nanoparticles (NPs). DXP-loaded PLGA-PEG NPs (DXP-NPs) of about 150 nm with a drug loading as high as 7.5% exhibited low hemolytic profile and cytotoxicity. DXP-NPs were able to inhibit the LPS-induced release of inflammatory cytokines in macrophages. After an intravenous injection to mice, dexamethasone (DXM) pharmacokinetic profile was also significantly improved. The concentration of DXM in the plasma of healthy mice remained high up to 18 h, much longer than the commercial soluble drug dexamethasone phosphate (DSP). Biodistribution studies showed lower DXM concentrations in the liver, kidneys, and lungs when DXP-NPs were administered as compared with the soluble drug. Histology analysis revealed an improvement in the knee structure and reduction of cell infiltration in animals treated with the encapsulated DXP compared with the soluble DSP or non-treated animals. In summary, the encapsulation of a lipidic prodrug of dexamethasone into PLGA-PEG NPs appears as a promising strategy to improve the pharmacological profile and reduce joint inflammation in a murine model of rheumatoid arthritis.
Collapse
Affiliation(s)
- Rosana Simón-Vázquez
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296, Châtenay-Malabry, France
- Immunology Group, Centro de Investigaciones Biomédicas, CINBIO, Universidade de Vigo, Campus Universitario Lagoas Marcosende, 36310, Vigo, Spain
- Instituto de Investigación Sanitaria Galicia Sur (IIS-GS), Pontevedra, Spain
| | - Nicolas Tsapis
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296, Châtenay-Malabry, France
| | - Mathilde Lorscheider
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296, Châtenay-Malabry, France
| | - Ainhoa Rodríguez
- Histology service, CINBIO, Universidade de Vigo, Campus Universitario Lagoas Marcosende, 36310, Vigo, Spain
| | - Patricia Calleja
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296, Châtenay-Malabry, France
| | - Ludivine Mousnier
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296, Châtenay-Malabry, France
| | - Encarnación de Miguel Villegas
- Immunology Group, Centro de Investigaciones Biomédicas, CINBIO, Universidade de Vigo, Campus Universitario Lagoas Marcosende, 36310, Vigo, Spain
- Histology service, CINBIO, Universidade de Vigo, Campus Universitario Lagoas Marcosende, 36310, Vigo, Spain
| | - África González-Fernández
- Immunology Group, Centro de Investigaciones Biomédicas, CINBIO, Universidade de Vigo, Campus Universitario Lagoas Marcosende, 36310, Vigo, Spain
- Instituto de Investigación Sanitaria Galicia Sur (IIS-GS), Pontevedra, Spain
| | - Elias Fattal
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296, Châtenay-Malabry, France.
| |
Collapse
|
30
|
Ma L, Zheng X, Lin R, Sun AR, Song J, Ye Z, Liang D, Zhang M, Tian J, Zhou X, Cui L, Liu Y, Liu Y. Knee Osteoarthritis Therapy: Recent Advances in Intra-Articular Drug Delivery Systems. Drug Des Devel Ther 2022; 16:1311-1347. [PMID: 35547865 PMCID: PMC9081192 DOI: 10.2147/dddt.s357386] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/17/2022] [Indexed: 12/12/2022] Open
Abstract
Drug delivery for osteoarthritis (OA) treatment is a continuous challenge because of their poor bioavailability and rapid clearance in joints. Intra-articular (IA) drug delivery is a common strategy and its therapeutic effects depend mainly on the efficacy of the drug-delivery system used for OA therapy. Different types of IA drug-delivery systems, such as microspheres, nanoparticles, and hydrogels, have been rapidly developed over the past decade to improve their therapeutic effects. With the continuous advancement in OA mechanism research, new drugs targeting specific cell/signaling pathways in OA are rapidly evolving and effective drug delivery is critical for treating OA. In this review, recent advances in various IA drug-delivery systems for OA treatment, OA targeted strategies, and related signaling pathways in OA treatment are summarized and analyzed based on current publications.
Collapse
Affiliation(s)
- Luoyang Ma
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
- Marine Medical Research Institute of Zhanjiang, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Xiaoyan Zheng
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
- Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang city, Guangdong province, 524045, People's Republic of China
| | - Rui Lin
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Antonia RuJia Sun
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen City, Guangdong Province, 518055, People’s Republic of China
| | - Jintong Song
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Zhiqiang Ye
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Dahong Liang
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Min Zhang
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Jia Tian
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Xin Zhou
- Marine Medical Research Institute of Zhanjiang, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Liao Cui
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Yuyu Liu
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
| | - Yanzhi Liu
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang City, Guangdong Province, 524023, People’s Republic of China
- Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang city, Guangdong province, 524045, People's Republic of China
- Shenzhen Osteomore Biotechnology Co., Ltd., Shenzhen city, Guangdong Province, 518118, People’s Republic of China
- Correspondence: Yanzhi Liu; Yuyu Liu, Tel +86-759-2388405; +86-759-2388588, Email ;
| |
Collapse
|
31
|
Di Francesco M, Fragassi A, Pannuzzo M, Ferreira M, Brahmachari S, Decuzzi P. Management of osteoarthritis: From drug molecules to nano/micromedicines. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1780. [PMID: 35253405 PMCID: PMC9285805 DOI: 10.1002/wnan.1780] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/29/2021] [Accepted: 01/21/2022] [Indexed: 12/12/2022]
Abstract
With the change in lifestyle and aging of the population, osteoarthritis (OA) is emerging as a major medical burden globally. OA is a chronic inflammatory and degenerative disease initially manifesting with joint pain and eventually leading to permanent disability. To date, there are no drugs available for the definitive treatment of osteoarthritis and most therapies have been palliative in nature by alleviating symptoms rather than curing the disease. This coupled with the vague understanding of the early symptoms and methods of diagnosis so that the disease continues as a global problem and calls for concerted research efforts. A cascade of events regulates the onset and progression of osteoarthritis starting with the production of proinflammatory cytokines, including interleukin (IL)‐1β, IL‐6, tumor necrosis factor (TNF)‐α; catabolic enzymes, such as matrix metalloproteinases (MMPs)‐1, ‐3, and ‐13, culminating into cartilage breakdown, loss of lubrication, pain, and inability to load the joint. Although intra‐articular injections of small and macromolecules are often prescribed to alleviate symptoms, low residence times within the synovial cavity severely impair their efficacy. This review will briefly describe the factors dictating the onset and progression of the disease, present the current clinically approved methods for its treatment and diagnosis, and finally elaborate on the main challenges and opportunities for the application of nano/micromedicines in the treatment of osteoarthritis. Thus, future treatment regimens will benefit from simultaneous consideration of the mechanobiological, the inflammatory, and tissue degradation aspects of the disease. This article is categorized under:Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement
Collapse
Affiliation(s)
- Martina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Agnese Fragassi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.,Department of Chemistry and Industrial Chemistry, University of Genova, Genoa, Italy
| | - Martina Pannuzzo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Miguel Ferreira
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Sayanti Brahmachari
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
32
|
Chitosan-Based Nanogels: Synthesis and Toxicity Profile for Drug Delivery to Articular Joints. NANOMATERIALS 2022; 12:nano12081337. [PMID: 35458048 PMCID: PMC9027118 DOI: 10.3390/nano12081337] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/06/2022] [Accepted: 04/09/2022] [Indexed: 12/20/2022]
Abstract
One important challenge in treating avascular-degraded cartilage is the development of new drugs for both pain management and joint preservation. Considerable efforts have been invested in developing nanosystems using biomaterials, such as chitosan, a widely used natural polymer exhibiting numerous advantages, i.e., non-toxic, biocompatible and biodegradable. However, even if chitosan is generally recognized as safe, the safety and biocompatibility of such nanomaterials must be addressed because of potential for greater interactions between nanomaterials and biological systems. Here, we developed chitosan-based nanogels as drug-delivery platforms and established an initial biological risk assessment for osteocartilaginous applications. We investigated the influence of synthesis parameters on the physicochemical characteristics of the resulting nanogels and their potential impact on the biocompatibility on all types of human osteocartilaginous cells. Monodisperse nanogels were synthesized with sizes ranging from 268 to 382 nm according to the acidic solution used (i.e., either citric or acetic acid) with overall positive charge surface. Our results demonstrated that purified chitosan-based nanogels neither affected cell proliferation nor induced nitric oxide production in vitro. However, nanogels were moderately genotoxic in a dose-dependent manner but did not significantly induce acute embryotoxicity in zebrafish embryos, up to 100 µg∙mL−1. These encouraging results hold great promise for the intra-articular delivery of drugs or diagnostic agents for joint pathologies.
Collapse
|
33
|
In Situ Gelling System for Sustained Intraarticular Delivery of Bupivacaine and Ketorolac in Sheep. Eur J Pharm Biopharm 2022; 174:35-46. [DOI: 10.1016/j.ejpb.2022.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 11/18/2022]
|
34
|
Lin W, Goldberg R, Klein J. Poly-phosphocholination of liposomes leads to highly-extended retention time in mice joints. J Mater Chem B 2022; 10:2820-2827. [PMID: 35099493 PMCID: PMC9007059 DOI: 10.1039/d1tb02346b] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Surface-attached layers of phosphatidylcholine (PC) lipid vesicles (liposomes) may reduce the friction coefficient μ (= force-to-slide/load) between the sliding surfaces down to μ ≈ 10−3–10−4 up to tens of atm contact pressures, as high as those in the major joints (hips or knees). Such friction reduction is attributed to hydration lubrication by the highly-hydrated phosphocholine head-groups exposed at the outer vesicle surfaces. It has been suggested therefore that intra-articular (IA) administration of liposomes as potential boundary lubricants may alleviate degenerative, friction-associated joint conditions such as osteoarthritis (OA), which is associated with insufficient lubrication at the articular cartilage surface. To overcome the problem, common to all nanoparticles, of rapid removal by the mononuclear phagocyte system, as well as to ensure long-term colloidal stability during storage, functionalizing liposomes with poly(ethylene glycol) moieties, PEGylation, is often used. Here we describe a different liposome functionalization approach, using poly(2-methacryloyloxyethyl phosphorylcholine), PMPC, moieties (strictly, lipid–PMPC conjugates), and compare the retention time in mice joints of such PMPCylated liposomes with otherwise-identical but PEGylated vesicles following IA administration. We find, using fluorescence labeling and in vivo optical imaging, that when PMPC-stabilized liposomes are injected into mice knee joints, there is a massive increase of the vesicles’ retention half-life in the joints of about (4–5)-fold (ca. 300–400% increase in retention time) compared with the PEGylated liposomes (and some 100-fold longer than the retention time of intra-articularly injected hyaluronan or HA). Such PMPCylated liposomes are therefore promising candidates as potential long-lived boundary lubricants at the articular cartilage surface, with implication for friction-associated pathologies. Moreover, as lipid vesicles are well known to be efficient drug carriers, such long retention in the joints may enable analgesic or anti-inflammatory agents for joint pathologies to be more efficiently delivered via IA administration using PMPCylated liposomal vehicles relative to PEGylated ones. PMPCylated liposomes injected into mice joints show a massive increase in retention half-life compared with PEGylated liposomes (or hyaluronan, HA), making them promising candidates as boundary lubricants at articular cartilage, or as drug carriers.![]()
Collapse
Affiliation(s)
- Weifeng Lin
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Ronit Goldberg
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Jacob Klein
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
35
|
Zewail M, Nafee N, Helmy MW, Boraie N. Synergistic and receptor-mediated targeting of arthritic joints via intra-articular injectable smart hydrogels containing leflunomide-loaded lipid nanocarriers. Drug Deliv Transl Res 2021; 11:2496-2519. [PMID: 34013458 DOI: 10.1007/s13346-021-00992-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2021] [Indexed: 11/30/2022]
Abstract
Intra-articular drug delivery represents a tempting strategy for local treatment of rheumatoid arthritis. Targeting drugs to inflamed joints bypasses systemic-related side effects. Albeit, rapid drug clearance and short joint residence limit intra-articular administration. Herein, injectable smart hydrogels comprising free/nanoencapsulated leflunomide (LEF) were developed. Nanostructured lipid carriers (NLCs), 200-300 nm, were coated with either chondroitin sulfate (CHS), hyaluronic acid (HA), or chitosan (CS) to provide joint targetability. Coated NLCs were incorporated in either hyaluronic/pluronic (HP) or chitosan/β-glycerophosphate (CS/βGP) hydrogels. Optimized systems ensured convenient gelation time (14-100 s), injectability (5-15 s), formulation-dependent mechanical strength, and extended LEF release up to 51 days. In vivo intra-articular injection in induced arthritis rat model revealed that rats treated with HA-coated NLCs showed the fastest recovery. Histopathological examination demonstrated perfect joint healing in case of HA-coated LEF-NLCs in CS/βGP thermogel manifested as minor erosion of subchondral bone, improved intensity of extracellular matrix, cartilage thickness, and chondrocyte number. Both HA- and CHS-coated NLCs reduced TNF-α level 4-5-fold relative to positive control. The feat would be achieved via active targeting to CD44 receptors overexpressed in the articular tissue, limiting chondrocyte apoptosis together with innate synergistic targetability by promoting chondrocyte proliferation and neovascularization, inhibiting the production of pro-inflammatory cytokines, thus enhancing cartilaginous tissue repair.
Collapse
Affiliation(s)
- Mariam Zewail
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Noha Nafee
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
- Department of Pharmaceutics, Faculty of Pharmacy, Kuwait University, POB 24923, 13110, Safat, Kuwait.
| | - Maged W Helmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Nabila Boraie
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
36
|
Zhang Z, Jiang W, Xie X, Liang H, Chen H, Chen K, Zhang Y, Xu W, Chen M. Recent Developments of Nanomaterials in Hydrogels: Characteristics, Influences, and Applications. ChemistrySelect 2021. [DOI: 10.1002/slct.202103528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Zongzheng Zhang
- School of Chemistry and Materials Science Ludong University Yantai 264025 China
| | - Wenqing Jiang
- School of Chemistry and Materials Science Ludong University Yantai 264025 China
| | - Xinmin Xie
- School of Chemistry and Materials Science Ludong University Yantai 264025 China
| | - Haiqing Liang
- School of Chemistry and Materials Science Ludong University Yantai 264025 China
| | - Hao Chen
- School of Chemistry and Materials Science Ludong University Yantai 264025 China
| | - Kun Chen
- School of Chemistry and Materials Science Ludong University Yantai 264025 China
| | - Ying Zhang
- School of Chemistry and Materials Science Ludong University Yantai 264025 China
| | - Wenlong Xu
- School of Chemistry and Materials Science Ludong University Yantai 264025 China
| | - Mengjun Chen
- School of Qilu Transportation Shandong University Jinan 250002 China
| |
Collapse
|
37
|
Pape E, Parent M, Pinzano A, Sapin-Minet A, Henrionnet C, Gillet P, Scala-Bertola J, Gambier N. Rapamycin-loaded Poly(lactic-co-glycolic) acid nanoparticles: Preparation, characterization, and in vitro toxicity study for potential intra-articular injection. Int J Pharm 2021; 609:121198. [PMID: 34662644 DOI: 10.1016/j.ijpharm.2021.121198] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/29/2021] [Accepted: 10/10/2021] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease. Rapamycin is a potential candidate for OA treatment by increasing the autophagy process implicated in its physiopathology. To optimize Rapamycin profit and avoid systemic side effects, intra-articular (i.a.) administration appeared helpful. However, Rapamycin's highly hydrophobic nature and low bioavailability made it challenging to develop purpose-made drug delivery systems to overcome these limitations. We developed Rapamycin-loaded nanoparticles (NPs) using poly (lactic-co-glycolic acid) by emulsion/evaporation method. We evaluated these NPs' cytocompatibility towards cartilage (chondrocytes) and synovial membrane cells (synoviocytes) for a potential i.a. administration. The in vitro characterization of Rapamycin-loaded NPs had shown a suitable profile for an i.a. administration. In vitro biocompatibility of NPs was highlighted to 10 µM of Rapamycin for both synoviocytes and chondrocytes, but significant toxicity was observed with higher concentrations. Besides, synoviocytes are more sensitive to Rapamycin-loaded NPs than chondrocytes. Finally, we observed in vitro that an adapted formulated Rapamycin-loaded NPs could be safe at suitable i.a. injection concentrations. The toxic effect of Rapamycin encapsulated in these NPs on both articular cells was dose-dependent. After Rapamycin-loaded NPs i.a. administration, local retention, in situ safety, and systemic release should be evaluated with experimental in vivo models.
Collapse
Affiliation(s)
- Elise Pape
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | | | - Astrid Pinzano
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France.
| | | | | | - Pierre Gillet
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Julien Scala-Bertola
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Nicolas Gambier
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| |
Collapse
|
38
|
Feng K, Xie X, Yuan J, Gong L, Zhu Z, Zhang J, Li H, Yang Y, Wang Y. Reversing the surface charge of MSC-derived small extracellular vesicles by εPL-PEG-DSPE for enhanced osteoarthritis treatment. J Extracell Vesicles 2021; 10:e12160. [PMID: 34724347 PMCID: PMC8559985 DOI: 10.1002/jev2.12160] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/27/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) possess a great therapeutical potential for osteoarthritis (OA) treatment. However, the steric and electrostatic hindrance of cartilage matrix leads to very limited distribution of MSC-sEVs in cartilage and low bioavailability of MSC-sEVs after intra-articular injection. To overcome this, a strategy to reverse the surface charge of MSC-sEVs by modifying the MSC-sEVs with a novel cationic amphiphilic macromolecule namely ε-polylysine-polyethylene-distearyl phosphatidylethanolamine (PPD) was developed in this study. Through incubation with 100 μg/ml PPD, positively charged MSC-sEVs (PPD-sEVs) were obtained, and the modification process showed nearly no disturbance to the integrity and contents of sEVs and exhibited good stability under the interference of anionic macromolecules. A more effective cellular uptake and homeostasis modulation ability of PPD-sEVs than unmodified MSC-sEVs to chondrocytes was demonstrated. More importantly, PPD-sEVs demonstrated significantly enhanced cartilage uptake, cartilage penetration, and joint retention capacity as compared to MSC-sEVs. Intra-articular injection of PPD-sEVs into a mouse OA model showed significantly improved bioavailability than MSC-sEVs, which resulted in enhanced therapeutic efficacy with reduced injection frequency. In general, this study provides a facile and effective strategy to improve the intra-articular bioavailability of MSC-sEVs and has a great potential to accelerate the clinical practice of MSC-sEVs based OA therapy.
Collapse
Affiliation(s)
- Kai Feng
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Xuetao Xie
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Ji Yuan
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Liangzhi Gong
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Zhaochen Zhu
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Juntao Zhang
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Haiyan Li
- Chemical and Environmental EngineeringSchool of EngineeringRMIT UniversityMelbourneAustralia
| | - Yunlong Yang
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Yang Wang
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| |
Collapse
|
39
|
Oo WM, Little C, Duong V, Hunter DJ. The Development of Disease-Modifying Therapies for Osteoarthritis (DMOADs): The Evidence to Date. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2921-2945. [PMID: 34262259 PMCID: PMC8273751 DOI: 10.2147/dddt.s295224] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/16/2021] [Indexed: 12/16/2022]
Abstract
Osteoarthritis (OA) is a complex heterogeneous articular disease with multiple joint tissue involvement of varying severity and no regulatory-agency-approved disease-modifying drugs (DMOADs). In this review, we discuss the reasons necessitating the development of DMOADs for OA management, the classifications of clinical phenotypes or molecular/mechanistic endotypes from the viewpoint of targeted drug discovery, and then summarize the efficacy and safety profile of a range of targeted drugs in Phase 2 and 3 clinical trials directed to cartilage-driven, bone-driven, and inflammation-driven endotypes. Finally, we briefly put forward the reasons for failures in OA clinical trials and possible steps to overcome these barriers.
Collapse
Affiliation(s)
- Win Min Oo
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Department of Physical Medicine and Rehabilitation, Mandalay General Hospital, University of Medicine, Mandalay, Mandalay, Myanmar
| | - Christopher Little
- Raymond Purves Bone and Joint Research Laboratories, Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Vicky Duong
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - David J Hunter
- Rheumatology Department, Royal North Shore Hospital, and Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| |
Collapse
|
40
|
Yang Y, Zhu Z, Gao R, Yuan J, Zhang J, Li H, Xie Z, Wang Y. Controlled release of MSC-derived small extracellular vesicles by an injectable Diels-Alder crosslinked hyaluronic acid/PEG hydrogel for osteoarthritis improvement. Acta Biomater 2021; 128:163-174. [PMID: 33862283 DOI: 10.1016/j.actbio.2021.04.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 03/24/2021] [Accepted: 04/02/2021] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) show great therapeutic potential for osteoarthritis (OA). However, their low bioavailability through intraarticular injection inhibits the process of clinical application. In the present study, an injectable Diels-Alder crosslinked hyaluronic acid/PEG (DAHP) hydrogel was developed as an intraarticular delivery platform for MSC-sEVs. Our results showed that the DAHP hydrogel could be prepared easily and that its gelation properties were suitable for intraarticular administration. In vitro studies demonstrated that the DAHP hydrogel could achieve sustained release of MSC-sEVs mainly by degradation control and preserve the therapeutic functions of sEVs. An in vivo experiment revealed that the DAHP hydrogel could enhance the efficacy of MSC-sEVs for OA improvement. This study provides a suitable delivery platform for MSC-sEVs-based OA therapy. STATEMENT OF SIGNIFICANCE: Mesenchymal stem cell (MSC)-derived small extracellular vesicles (MSC-sEVs) have shown a high potential as a cell-free therapeutic factor for treating osteoarthritis (OA). The sustained release of these MSC-sEVs in the joint space is essential for their clinical application. Herein, an injectable Diels-Alder crosslinked hyaluronic acid/PEG (DAHP) hydrogel was developed for intraarticular release of MSC-sEVs. The properties of the DAHP hydrogel, namely gelation features, cytocompatibility, sustained release, and functional maintenance of MSC-sEVs, make it suitable for intraarticular injection and delivery of sEVs. The efficacy of MSC-sEVs was enhanced by the intraarticularly injected DAHP hydrogel. Our present study provides a promising sustained delivery platform for MSC-sEVs for treating OA.
Collapse
Affiliation(s)
- Yunlong Yang
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China
| | - Zhaochen Zhu
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China; Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China
| | - Renzhi Gao
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China; Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China
| | - Ji Yuan
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China
| | - Juntao Zhang
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China
| | - Haiyan Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800#, Dongchuan Road, Shanghai 200240, China; Chemical and Environmental Engineering, School of Engineering, RMIT University, 124 La Trobe St, Melbourne VIC 3000, Australia
| | - Zongping Xie
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China.
| | - Yang Wang
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People' Hospital, 600# Yishan Road, Shanghai 200233, China
| |
Collapse
|
41
|
Parra-Coca A, Boix-Montañés A, Calpena-Campmany AC, Colom H. In vivo pharmacokinetic evaluation of carprofen delivery from intra-articular nanoparticles in rabbits: A population modelling approach. Res Vet Sci 2021; 137:235-242. [PMID: 34049110 DOI: 10.1016/j.rvsc.2021.05.012] [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: 03/17/2020] [Revised: 05/09/2021] [Accepted: 05/18/2021] [Indexed: 11/16/2022]
Abstract
Osteoarthritis is treated with COX or fosfolipase A2 inhibitors such as carprofen, a propionic acid NSAID. The enhancement of its action over the articular cartilage is mandatory to facilitate its therapeutic application. Drug uptake into the cartilage requires high synovial fluid concentrations, anticipating its rapid distribution towards bloodstream. Thus, intraarticular administration improves local targeting of the drug, lining with the site of action. A pharmacokinetic study in rabbits has been performed to evaluate carprofen nanoparticles after intraarticular administration. Pharmacokinetic analysis of plasma profiles through a modelling approach, has demonstrated the rapid distribution of drug outside of synovial chamber but mainly remaining in plasma. The data modelling has demonstrated the existence of two release-absorption processes when the nanoparticles are administered in the synovial space. Additionally, results are predictive of the PK profile of some other species such as cat, dogs or humans.
Collapse
Affiliation(s)
- A Parra-Coca
- Department of Pharmacy and Pharmaceutical technology and Physical-Chemistry, Faculty of Pharmacy and Food sciences, University of Barcelona, ES, Spain; Department of Veterinary Medicine and Zootechnic, Faculty of Agricultural Sciences, University of Applied and Environmental Sciences (U.D.C.A.), Bogotá, CO, Colombia
| | - A Boix-Montañés
- Department of Pharmacy and Pharmaceutical technology and Physical-Chemistry, Faculty of Pharmacy and Food sciences, University of Barcelona, ES, Spain.
| | - A C Calpena-Campmany
- Department of Pharmacy and Pharmaceutical technology and Physical-Chemistry, Faculty of Pharmacy and Food sciences, University of Barcelona, ES, Spain
| | - H Colom
- Department of Pharmacy and Pharmaceutical technology and Physical-Chemistry, Faculty of Pharmacy and Food sciences, University of Barcelona, ES, Spain
| |
Collapse
|
42
|
Alves JC, Santos A, Jorge P, Lavrador C, Carreira LM. Evaluation of digital thermography imaging to assess and monitor treatment of police working dogs with naturally occurring hip osteoarthritis. BMC Vet Res 2021; 17:180. [PMID: 33933090 PMCID: PMC8088695 DOI: 10.1186/s12917-021-02876-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 04/11/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In dogs, thermal imaging has been documented only recently, but a growing interest in this modality has led to studies using thermography to assess pathologies in the canine hip, stifle, elbow, intervertebral disc, and bone neoplasia. This study aimed to evaluate the use of digital thermography in assessing and evaluating treatment response in dogs with hip osteoarthritis (OA) and comparing its results with an objective measure and two clinical metrology instruments. In an experimental, randomized, double-blinded study, one hundred hip joints of fifty police working dogs with bilateral hip OA were evaluated. A dorsoventral and lateral thermographic image were obtained on days 0, 8, 15, 30, 90, and 180. Mean and maximal temperatures were determined. Additionally, the animal's weight-bearing distribution and radiographic examination of the hip joint (extended legs ventrodorsal view) were performed. Copies of the Canine Brief Pain Inventory (CBPI) and Canine Orthopaedic Index (COI) were obtained. Results were analyzed by ANOVA, followed by an LSD post-hoc test, and correlations were assessed with Spearman correlation coefficient, with p < 0.05. RESULTS Values recorded on the lateral view were higher than those on the dorsoventral view. No differences or correlations were found between Orthopedic Foundation for Animals hip grades and temperature. Digital thermographic images showed a weak significant correlation with weight-bearing evaluations (r = 0.13, p < 0.01) and different clinical metrology instruments scores (r = - 0.25, p < 0.01 for pain severity score, and r = - 0.21, p = 0.04 for gait). It also correlated with radiographic findings, specifically the circumferential femoral head osteophyte and caudolateral curvilinear osteophyte. CONCLUSION To our knowledge, this is the first study presenting the digital thermography assessment of Police working dogs submitted to treatment for hip OA. Digital thermography, mainly based on a lateral view evaluation, showed a weak significant correlation with stance analysis and clinical metrology instruments scores.
Collapse
Affiliation(s)
- J C Alves
- Divisão de Medicina Veterinária, Guarda Nacional Republicana (GNR), Rua Presidente Arriaga, 9, 1200-771, Lisbon, Portugal. .,MED - Mediterranean Institute for Agriculture, Environment and Development, Instituto de Investigação e Formação Avançada, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554, Évora, Portugal.
| | - A Santos
- Divisão de Medicina Veterinária, Guarda Nacional Republicana (GNR), Rua Presidente Arriaga, 9, 1200-771, Lisbon, Portugal
| | - P Jorge
- Divisão de Medicina Veterinária, Guarda Nacional Republicana (GNR), Rua Presidente Arriaga, 9, 1200-771, Lisbon, Portugal
| | - C Lavrador
- MED - Mediterranean Institute for Agriculture, Environment and Development, Instituto de Investigação e Formação Avançada, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554, Évora, Portugal
| | - L Miguel Carreira
- Faculty of Veterinary Medicine, University of Lisbon (FMV/ULisboa), Lisbon, Portugal.,Interdisciplinary Centre for Research in Animal Health (CIISA) - University of Lisbon, (FMV/ULisboa), Lisbon, Portugal.,Anjos of Assis Veterinary Medicine Centre (CMVAA), Barreiro, Portugal
| |
Collapse
|
43
|
Siefen T, Lokhnauth J, Liang A, Larsen CC, Lamprecht A. An ex-vivo model for transsynovial drug permeation of intraarticular injectables in naive and arthritic synovium. J Control Release 2021; 332:581-591. [PMID: 33705826 DOI: 10.1016/j.jconrel.2021.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/23/2021] [Accepted: 03/04/2021] [Indexed: 01/17/2023]
Abstract
Estimation of joint residence time of a drug is a key requirement for rational development of intraarticular therapeutics. There is a great need for a predictive model to reduce the high number of animal experiments in early stage development. Here, a Franz-cell based porcine ex-vivo permeation model is proposed, and transsynovial permeation of fluorescently-labeled dextrans in the range of potential drug candidates (10-150 kDa), as well as a small molecule (fluorescein sodium) and charged dextran derivates, have been determined. In addition, a lipopolysaccharide (LPS) -induced synovitis model was assessed for inflammatory biomarker levels and its effect on permeation of the solutes. Size-dependent permeability was observed for the analytes, which distinctly differed from findings with an artificial polycarbonate membrane, which is a widely used model. LPS was found to successfully stimulate an inflammatory response and led to a reduced size selectivity of the synovial membrane. 150 kDa dextran flux was accelerated approximately 2.5-fold in the inflamed state, whereas the permeation of smaller molecules was little affected. Moreover, by varying the LPS concentrations, the ex-vivo model was shown to produce varying degrees of synovitis-like inflammation. A simple and highly relevant ex-vivo tool for investigation of transsynovial permeation was developed, offering the further advantage of mimicking synovitis-induced permeability changes. Thus, this model provides a promising method for formulation screening, while reducing the need for animal experiments.
Collapse
Affiliation(s)
- Tobias Siefen
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany
| | | | - Alfred Liang
- Ferring Pharmaceuticals Inc, Parsippany, NJ, USA
| | | | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; PEPITE (EA4267), University of Burgundy/Franche-Comté, Besançon, France.
| |
Collapse
|
44
|
Rahimi M, Charmi G, Matyjaszewski K, Banquy X, Pietrasik J. Recent developments in natural and synthetic polymeric drug delivery systems used for the treatment of osteoarthritis. Acta Biomater 2021; 123:31-50. [PMID: 33444800 DOI: 10.1016/j.actbio.2021.01.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/15/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA), is a common musculoskeletal disorder that will progressively increase in older populations and is expected to be the most dominant cause of disability in the world population by 2030. The progression of OA is controlled by a multi-factorial pathway that has not been completely elucidated and understood yet. However, over the years, research efforts have provided a significant understanding of some of the processes contributing to the progression of OA. Both cartilage and bone degradation processes induce articular cells to produce inflammatory mediators that produce proinflammatory cytokines that block the synthesis of collagen type II and aggrecan, the major components of cartilage. Systemic administration and intraarticular injection of anti-inflammatory agents are the first-line treatments of OA. However, small anti-inflammatory molecules are rapidly cleared from the joint cavity which limits their therapeutic efficacy. To palliate this strong technological drawback, different types of polymeric materials such as microparticles, nanoparticles, and hydrogels, have been examined as drug carriers for the delivery of therapeutic agents to articular joints. The main purpose of this review is to provide a summary of recent developments in natural and synthetic polymeric drug delivery systems for the delivery of anti-inflammatory agents to arthritic joints. Furthermore, this review provides an overview of the design rules that have been proposed so far for the development of drug carriers used in OA therapy. Overall it is difficult to state clearly which polymeric platform is the most efficient one because many advantages and disadvantages could be pointed to both natural and synthetic formulations. That requires further research in the near future.
Collapse
|
45
|
Sun Z, Li Z, Qu K, Zhang Z, Niu Y, Xu W, Ren C. A review on recent advances in gel adhesion and their potential applications. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2020.115254] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
46
|
Dhanka M, Pawar V, Chauhan DS, Jain NK, R S P, Shetty C, Kumawat MK, Prasad R, Srivastava R. Synthesis and characterization of an injectable microparticles integrated hydrogel composite biomaterial: In-vivo biocompatibility and inflammatory arthritis treatment. Colloids Surf B Biointerfaces 2021; 201:111597. [PMID: 33609936 DOI: 10.1016/j.colsurfb.2021.111597] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/11/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022]
Abstract
Polymeric hydrogels and microparticles have been widely used for localized drug delivery applications for the treatment of arthritis. Nonetheless, owing to initial burst drug release, non-specific biodistribution and low retention time at the target site in body, these polymeric drug delivery systems have been found with low in-vivo performance. Hence, the above limitations need to be resolved by designing a smart novel drug delivery system which is the current need in biomedicine. Herein, a novel localized injectable thermoresponsive microparticles embedded hydrogel composite drug delivery system has been developed for the treatment of inflammatory arthritis. In the current study, methotrexate (MTX) loaded alginate microparticles (MTX-Microparticles) are embedded into thermoreversible hydrogel matrix (MTX-MPs-H) prepared by physical blending of sodium hyaluronate and methylcellulose (SHMC). Microparticles-hydrogel composite system exhibited appropriate in-vitro thermoreversibility (sol at 4 °C and gel at 37 °C), biocompatibility (>80 %), hemocompatibility, and controlled drug release profile. The in-vivo biocompatibility studies for 10 days revealed that composite system is non-toxic in nature. The developed MTX-MPs-H composite drug delivery system effectively decreased the swelling/ inflammation of the arthritis affected paw in wistar rats in comparison to only alginate microparticles and pure MTX up to 30 days.
Collapse
Affiliation(s)
- Mukesh Dhanka
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | - Vaishali Pawar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | - Deepak S Chauhan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | - Nishant Kumar Jain
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | - Prabhuraj R S
- Center for Research in Nanotechnology & Science (CRNTS), IIT Bombay, Mumbai, India
| | - Chaitra Shetty
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | - Mukesh K Kumawat
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India
| | - Rajendra Prasad
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India; Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT Bombay), Mumbai, India.
| |
Collapse
|
47
|
Alves JC, Santos A, Jorge P, Lavrador C, Carreira LM. The intra-articular administration of triamcinolone hexacetonide in the treatment of osteoarthritis. Its effects in a naturally occurring canine osteoarthritis model. PLoS One 2021; 16:e0245553. [PMID: 33471857 PMCID: PMC7816979 DOI: 10.1371/journal.pone.0245553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/03/2021] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE To evaluate the effect of an intra-articular (IA) administration of triamcinolone hexacetonide, compared with saline. PATIENTS AND METHODS Forty (N = 40) hip joints were randomly assigned to a treatment group (THG, n = 20, receiving IA triamcinolone hexacetonide) and a control group (CG, n = 20, receiving IA saline). On treatment day (T0), and at 8, 15, 30, 90 and 180 days post-treatment, weight distribution, joint range of motion, thigh girth, digital thermography, radiographic signs, synovial fluid interleukin-1 and C-reactive protein levels were evaluated. Data from four Clinical Metrology Instruments was also gathered. Results were compared Repeated Measures ANOVA, with a Huynh-Feldt correction, Paired Samples T-Test or Wilcoxon Signed Ranks Test. A Kaplan-Meier test was performed to compare both groups, with p<0.05. RESULTS Joints were graded as mild (65%), moderate (20%) and severe (15%). Patients of both sexes, with a mean age of 6.5±2.4 years and bodyweight of 26.7±5.2kg, were included. No differences were found between groups at T0. Comparing THG to CG, weight distribution showed significant improvements in THG from 8 (p = 0.05) up to 90 days (p = 0.01). THG showed lower values during thermographic evaluation in the Lt view (p<0.01). Pain and function scores also improved from 30 to 180 days. Increasing body weight, age, and presence of caudolateral curvilinear osteophyte corresponded to worse response to treatment. Results of the Kaplan Meier test showed significant differences between groups, with THG performing better considering several evaluations and scores. CONCLUSION THG recorded significant improvements in weight-bearing and in with the considered CMIs, particularly pain scores. Lower thermographic values were registered in THG up to the last evaluation day. Age, sex, and radiographic findings did significantly influenced response to treatment.
Collapse
Affiliation(s)
- João C. Alves
- Divisão de Medicina Veterinária, Guarda Nacional Republicana (GNR), Lisbon, Portugal
- MED – Mediterranean Institute for Agriculture, Environment and Development, Instituto de Investigação e Formação Avançada, Universidade de Évora, Pólo da Mitra, Évora, Portugal
| | - Ana Santos
- Divisão de Medicina Veterinária, Guarda Nacional Republicana (GNR), Lisbon, Portugal
| | - Patrícia Jorge
- Divisão de Medicina Veterinária, Guarda Nacional Republicana (GNR), Lisbon, Portugal
| | - Catarina Lavrador
- MED – Mediterranean Institute for Agriculture, Environment and Development, Instituto de Investigação e Formação Avançada, Universidade de Évora, Pólo da Mitra, Évora, Portugal
| | - L. Miguel Carreira
- Faculty of Veterinary Medicine, University of Lisbon (FMV/ULisboa), Lisbon, Portugal
- Interdisciplinary Centre for Research in Animal Health (CIISA) – University of Lisbon, (FMV/ULisboa), Lisbon, Portugal
- Anjos of Assis Veterinary Medicine Centre (CMVAA), Barreiro, Portugal
| |
Collapse
|
48
|
Alves JC, Santos A, Jorge P, Lavrador C, Carreira LM. Intra-articular Injections With Either Triamcinolone Hexacetonide, Stanozolol, Hylan G-F 20, or a Platelet Concentrate Improve Clinical Signs in Police Working Dogs With Bilateral Hip Osteoarthritis. Front Vet Sci 2021; 7:609889. [PMID: 33537353 PMCID: PMC7849590 DOI: 10.3389/fvets.2020.609889] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Objectives: To compare the effect of intra-articular treatment with triamcinolone hexacetonide (TH), stanozolol, hyaluronan, and a platelet concentrate in police working dogs with bilateral hip osteoarthritis (OA). Study Design: Prospective, longitudinal, double-blinded, negative controlled study. Sample Population: Fifty police working dogs with naturally occurring hip OA. Methods: Animals were randomly assigned to a control group (CG, n = 10), TH group (THG, n = 10), platelet concentrate group (PCG, n = 10), stanozolol group (SG, n = 10), and Hylan G-F 20 group (HG). On days 0 (T0), 8, 15, 30, 90, and 180 days post-treatment, weight-bearing distribution was evaluated. In those days, and on days 60, 120, and 150, four clinical metrology instruments were completed. Kaplan-Meier estimators were conducted and compared with the log-rank test. Cox proportional hazard regression analysis was performed to determine treatment survival. Significance was set at p < 0.05. Results: Patients had a mean age of 6.5 ± 2.4 years and body weight of 26.7 ± 5.2 kg. At T0, hips were classified as mild (n = 35), moderate (n = 10), and severe (n = 5), according to the Orthopedic Foundation for Animals grading scheme. No differences were found between groups at that moment considering age, body weight, OFA hip score, and all assessments performed. All treatments improved clinical signs in various OA dimensions in some groups, with a broad effect interval. PCG showed a lower range of variation while maintaining a positive result for more extended periods (p < 0.01 for symmetry index and 0.01 < p < 0.04 in the majority of scores). Breed, age, sex, and OFA grade did not significantly influence response to treatment. Conclusions and Clinical Relevance: This is the first prospective, negative controlled, double-blinded study to compare the effect of a single administration of these IA treatments in dogs with hip OA. HG and PCG recorded more significant improvements throughout the 180-day follow-up. In particular, PCG also registered a lower variation in results, seemingly the best therapeutic option. Nevertheless, improvements were still observed in THG and SG, and these treatment options can be considered, mainly when the first two treatments are not available.
Collapse
Affiliation(s)
- João C. Alves
- Divisão de Medicina Veterinária, Guarda Nacional Republicana (GNR), Lisbon, Portugal,MED - Mediterranean Institute for Agriculture, Environment and Development, Instituto de Investigação e Forma Avana, Universidade de Évora, Évora, Portugal,*Correspondence: João C. Alves
| | - Ana Santos
- Divisão de Medicina Veterinária, Guarda Nacional Republicana (GNR), Lisbon, Portugal
| | - Patrícia Jorge
- Divisão de Medicina Veterinária, Guarda Nacional Republicana (GNR), Lisbon, Portugal
| | - Catarina Lavrador
- MED - Mediterranean Institute for Agriculture, Environment and Development, Instituto de Investigação e Forma Avana, Universidade de Évora, Évora, Portugal
| | - L. Miguel Carreira
- Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal,Interdisciplinary Centre for Research in Animal Health (CIISA), University of Lisbon, Lisbon, Portugal,Anjos of Assis Veterinary Medicine Centre (CMVAA), Barreiro, Portugal
| |
Collapse
|
49
|
Aminkov KB, Mehandzhiyski NH, Aminkov BY, Zlateva-Panayotova NZ. Application of platelet-rich plasma for canine osteoarthritis treatment - a clinical series. BULGARIAN JOURNAL OF VETERINARY MEDICINE 2021. [DOI: 10.15547/bjvm.2019-0095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Osteoarthritis, also known as degenerative joint disease (DJD), is defined as a progressive and permanent long-term deterioration of the cartilage surrounding the joints. There is no known cause for primary DJD. However, there are a wide variety of causes for secondary DJD, such as trauma, abnormal wear of joints and cartilage, or a congenital defect present at birth such as an improperly formed hip. One of the most popular methods used to biologically enhance healing in the fields of orthopaedic surgery and medicine includes the use of autologous blood products, namely, platelet rich plasma (PRP). Reports suggest that PRP, presumably containing high levels of platelet growth factors, may promote the recovery of the affected cartilage. This case series presents clinical and radiographic findings of three dogs with osteoarthritis of the elbow and knee joints. Pain score were assessed by CBPI (Canine Brief Pain Inventory). Treatment with three-fold intra-articular application of PRP, obtained by double centrifugation method, resulted in significant improvement in the function of the affected joint. Therefore, it could be concluded that PRP was clinically effective in the treatment of osteoarthritis in these three cases.
Collapse
Affiliation(s)
- K. B Aminkov
- Department of Surgery, Radiology, Anesthesiology, Obstetrics and Gynecology, Faculty of Veterinary Medicine, University of Forestry, Sofia, Bulgaria
| | - N. H. Mehandzhiyski
- Department of Surgery, Radiology, Anesthesiology, Obstetrics and Gynecology, Faculty of Veterinary Medicine, University of Forestry, Sofia, Bulgaria
| | - B. Y. Aminkov
- Department of Surgery, Radiology, Anesthesiology, Obstetrics and Gynecology, Faculty of Veterinary Medicine, University of Forestry, Sofia, Bulgaria
| | - N. Z. Zlateva-Panayotova
- Department of Surgery, Radiology, Anesthesiology, Obstetrics and Gynecology, Faculty of Veterinary Medicine, University of Forestry, Sofia, Bulgaria
| |
Collapse
|
50
|
Chen W, Li Z, Wang Z, Gao H, Ding J, He Z. Intraarticular Injection of Infliximab-Loaded Thermosensitive Hydrogel Alleviates Pain and Protects Cartilage in Rheumatoid Arthritis. J Pain Res 2020; 13:3315-3329. [PMID: 33324092 PMCID: PMC7733037 DOI: 10.2147/jpr.s283518] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/19/2020] [Indexed: 12/16/2022] Open
Abstract
Purpose Pain and cartilage destruction caused by rheumatoid arthritis (RA) are major challenges during clinical treatment. Traditional systemic administration not only has obvious side effects but also provides limited relief for local symptoms in major joints. Local delivery of therapeutics for RA treatment is a potential strategy but is limited by rapid intraarticular release. Materials and Methods In this study, we prepared a thermoresponsive injectable hydrogel by mixing pluronic F127 (F127) and hyaluronic acid (HA) with poly (γ-glutamic acid) (PGA) incorporating infliximab (IFX), a new generation monoclonal antibody drug. We investigated the biocompatibility of the hydrogel and its IFX release profile. In vivo, we studied the clinical manifestations (articular skin temperature and joint diameter), detected cytokines in the synovial fluid and cartilage, performed behavioral studies on pain relief, and evaluated the cartilage protection effect. Results A thermoresponsive hydrogel was successfully prepared by mixing F127, HA, and PGA with injectable properties. The F127-HA-PGA hydrogel had a porous structure with interconnected pores. The infliximab-loaded thermosensitive hydrogel exhibited good biocompatibility and biodegradability and sustained release properties. Intraarticular injection of the IFX-loaded F127-HA-PGA hydrogel could alleviate the expression of inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and interleukin-17 (IL-17), in the synovial fluid and cartilage as well as relieve pain and inhibit cartilage destruction in RA. Conclusion The double effect on pain relief and cartilage protection indicated the significant potential of the IFX-loaded injectable hydrogel for RA treatment in major joint lesions.
Collapse
Affiliation(s)
- Weiying Chen
- School of Anesthesiology, Third Affiliated Hospital of Guizhou Medical University, Guiyang 550004, People's Republic of China.,Department of Anesthesiology, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201112, People's Republic of China
| | - Zuhao Li
- Department of Anesthesiology, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201112, People's Republic of China.,Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| | - Zhenhong Wang
- Department of Anesthesiology, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201112, People's Republic of China
| | - Hong Gao
- School of Anesthesiology, Third Affiliated Hospital of Guizhou Medical University, Guiyang 550004, People's Republic of China
| | - Junyun Ding
- Department of Anesthesiology, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201112, People's Republic of China
| | - Zhenzhou He
- School of Anesthesiology, Third Affiliated Hospital of Guizhou Medical University, Guiyang 550004, People's Republic of China.,Department of Anesthesiology, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201112, People's Republic of China
| |
Collapse
|