1
|
Tang X, Zhou F, Wang S, Wang G, Bai L, Su J. Bioinspired injectable hydrogels for bone regeneration. J Adv Res 2024:S2090-1232(24)00486-7. [PMID: 39505143 DOI: 10.1016/j.jare.2024.10.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 09/28/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024] Open
Abstract
The effective regeneration of bone/cartilage defects remains a significant clinical challenge, causing irreversible damage to millions annually.Conventional therapies such as autologous or artificial bone grafting often yield unsatisfactory outcomes, emphasizing the urgent need for innovative treatment methods. Biomaterial-based strategies, including hydrogels and active scaffolds, have shown potential in promoting bone/cartilage regeneration. Among them, injectable hydrogels have garnered substantial attention in recent years on account of their minimal invasiveness, shape adaptation, and controlled spatiotemporal release. This review systematically discusses the synthesis of injectable hydrogels, bioinspired approaches-covering microenvironment, structural, compositional, and bioactive component-inspired strategies-and their applications in various bone/cartilage disease models, highlighting bone/cartilage regeneration from an innovative perspective of bioinspired design. Taken together, bioinspired injectable hydrogels offer promising and feasible solutions for promoting bone/cartilage regeneration, ultimately laying the foundations for clinical applications. Furthermore, insights into further prospective directions for AI in injectable hydrogels screening and organoid construction are provided.
Collapse
Affiliation(s)
- Xuan Tang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Fengjin Zhou
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an 710000, China
| | - Sicheng Wang
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Department of Orthopedics Trauma, Shanghai Zhongye Hospital, Shanghai 201900, China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Wenzhou Institute of Shanghai University, Wenzhou 325000, China.
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
2
|
Zhu R, Liao HY, Huang YC, Shen HL. Application of Injectable Hydrogels as Delivery Systems in Osteoarthritis and Rheumatoid Arthritis. Br J Hosp Med (Lond) 2024; 85:1-41. [PMID: 39212571 DOI: 10.12968/hmed.2024.0347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Osteoarthritis and rheumatoid arthritis, though etiologically distinct, are both inflammatory joint diseases that cause progressive joint injury, chronic pain, and loss of function. Therefore, long-term treatment with a focus on relieving symptoms is needed. At present, the primary treatment for arthritis is drug therapy, both oral and intravenous. Although significant progress has been achieved for these treatment methods in alleviating symptoms, certain prominent drawbacks such as the substantial side effects and limited absorption of medications call for an urgent need for improved drug delivery methods. Injected hydrogels can be used as a delivery system to deliver drugs to the joint cavity in a controlled manner and continuously release them, thereby enhancing drug retention in the joint cavity to improve therapeutic effectiveness, which is attributed to the desirable attributes of the delivery system such as low immunogenicity, good biodegradability and biocompatibility. This review summarizes the types of injectable hydrogels and analyzes their applications as delivery systems in arthritis treatment. We also explored how hydrogels counteract inflammation, bone and cartilage degradation, and oxidative stress, while promoting joint cartilage regeneration in the treatment of osteoarthritis (OA) and rheumatoid arthritis (RA). This review also highlights new approaches to developing injectable hydrogels as delivery systems for OA and RA.
Collapse
Affiliation(s)
- Rong Zhu
- Department of Rheumatology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Hai-Yang Liao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yi-Chen Huang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Hai-Li Shen
- Department of Rheumatology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
3
|
Kalairaj MS, Pradhan R, Saleem W, Smith MM, Gaharwar AK. Intra-Articular Injectable Biomaterials for Cartilage Repair and Regeneration. Adv Healthc Mater 2024; 13:e2303794. [PMID: 38324655 PMCID: PMC11468459 DOI: 10.1002/adhm.202303794] [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: 10/31/2023] [Revised: 12/29/2023] [Indexed: 02/09/2024]
Abstract
Osteoarthritis is a degenerative joint disease characterized by cartilage deterioration and subsequent inflammatory changes in the underlying bone. Injectable hydrogels have emerged as a promising approach for controlled drug delivery in cartilage therapies. This review focuses on the latest developments in utilizing injectable hydrogels as vehicles for targeted drug delivery to promote cartilage repair and regeneration. The pathogenesis of osteoarthritis is discussed to provide a comprehensive understanding of the disease progression. Subsequently, the various types of injectable hydrogels used for intra-articular delivery are discussed. Specifically, physically and chemically crosslinked injectable hydrogels are critically analyzed, with an emphasis on their fabrication strategies and their capacity to encapsulate and release therapeutic agents in a controlled manner. Furthermore, the potential of incorporating growth factors, anti-inflammatory drugs, and cells within these injectable hydrogels are discussed. Overall, this review offers a comprehensive guide to navigating the landscape of hydrogel-based therapeutics in osteoarthritis.
Collapse
Affiliation(s)
| | - Ridhi Pradhan
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Waqas Saleem
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Morgan M. Smith
- Department of Veterinary Integrative BiosciencesSchool of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTX77843USA
| | - Akhilesh K. Gaharwar
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Department of Material Science and EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Genetics and Genomics Interdisciplinary ProgramTexas A&M UniversityCollege StationTX77843USA
| |
Collapse
|
4
|
Wang Y, Yang C, Liu W, Zhang Y, Wang Q, Cheng H, Shi J, Yang X, Yang S, Yao X, Wang Y, Song X. Enhanced efficacy of brucine dissolving-microneedles as a targeted delivery system in rheumatoid arthritis treatment: a comprehensive pharmacokinetic-pharmacodynamic analysis. Drug Deliv Transl Res 2024:10.1007/s13346-024-01606-w. [PMID: 38705909 DOI: 10.1007/s13346-024-01606-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
Our previous studies have shown the therapeutic efficacy of brucine dissolving-microneedles (Bru-DMNs) in treating rheumatoid arthritis (RA). Bru delivered via the DMNs can bypass some of the issues related to oral and systemic delivery, including extensive enzymatic activity, liver metabolism and in the case of systemic delivery via hypodermic needles, pain resulting from injections and needle stick injury. However, the underlying mechanism of Bru-DMNs against RA has not been investigated in depth at the pharmacokinetic-pharmacodynamic (PK-PD) level. In this study, a microdialysis-based method combined with ultra-performance liquid chromatography-tandem mass spectrometry was developed for the simultaneous and continuous sampling and quantitative analysis of blood and joint cavities in fully awake RA rats. The acquired data were analyzed by the PK-PD analysis method. Bru delivered via microneedles showed enhanced distribution and prolonged retention in the joint cavity compared to its administration in blood. The correlation between the effect of Bru and its concentration at the action site was indirect. In this study, we explored the mechanism of Bru-DMNs against RA and established a visualization method to express the PK-PD relationship of Bru-DMNs against RA. This study provides insights into the mechanism of action of drugs with potential side effects administered transdermally for RA treatment.
Collapse
Affiliation(s)
- Yunxia Wang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
- National Engineering Research Center of Miao's Medicines, Guiyang, 550025, China
| | - Changfu Yang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Wen Liu
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
- School of Pharmacy, Guizhou Medical University, Guiyang, 561113, China
| | - Yongping Zhang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Qun Wang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Huanhuan Cheng
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Jianan Shi
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Xiaoshuang Yang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Shenglei Yang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Xueming Yao
- The Second Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Yonglin Wang
- Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, 550004, China.
| | - Xinli Song
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
- National Engineering Research Center of Miao's Medicines, Guiyang, 550025, China.
| |
Collapse
|
5
|
Nie R, Zhang QY, Feng ZY, Huang K, Zou CY, Fan MH, Zhang YQ, Zhang JY, Li-Ling J, Tan B, Xie HQ. Hydrogel-based immunoregulation of macrophages for tissue repair and regeneration. Int J Biol Macromol 2024; 268:131643. [PMID: 38643918 DOI: 10.1016/j.ijbiomac.2024.131643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/10/2024] [Accepted: 04/14/2024] [Indexed: 04/23/2024]
Abstract
The rational design of hydrogel materials to modulate the immune microenvironment has emerged as a pivotal approach in expediting tissue repair and regeneration. Within the immune microenvironment, an array of immune cells exists, with macrophages gaining prominence in the field of tissue repair and regeneration due to their roles in cytokine regulation to promote regeneration, maintain tissue homeostasis, and facilitate repair. Macrophages can be categorized into two types: classically activated M1 (pro-inflammatory) and alternatively activated M2 (anti-inflammatory and pro-repair). By regulating the physical and chemical properties of hydrogels, the phenotypic transformation and cell behavior of macrophages can be effectively controlled, thereby promoting tissue regeneration and repair. A full understanding of the interaction between hydrogels and macrophages can provide new ideas and methods for future tissue engineering and clinical treatment. Therefore, this paper reviews the effects of hydrogel components, hardness, pore size, and surface morphology on cell behaviors such as macrophage proliferation, migration, and phenotypic polarization, and explores the application of hydrogels based on macrophage immune regulation in skin, bone, cartilage, and nerve tissue repair. Finally, the challenges and future prospects of macrophage-based immunomodulatory hydrogels are discussed.
Collapse
Affiliation(s)
- Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Qing-Yi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Zi-Yuan Feng
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Kai Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Chen-Yu Zou
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ming-Hui Fan
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yue-Qi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ji-Ye Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jesse Li-Ling
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Medical Genetics, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Bo Tan
- Department of Orthopedic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, PR China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, PR China.
| |
Collapse
|
6
|
Zhang J, Song Y, Zhu L, You Y, Hu J, Xu X, Wang C, Lu J, Shen Q, Xu X, Teng C, Du Y. An injectable thermosensitive hyaluronic acid/pluronic F-127 hydrogel for deep penetration and combination therapy of frozen shoulder. Int J Biol Macromol 2024; 263:130342. [PMID: 38395289 DOI: 10.1016/j.ijbiomac.2024.130342] [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: 09/25/2023] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024]
Abstract
Frozen shoulder (FS) is a common and progressive shoulder disorder that causes glenohumeral joint stiffness, characterized by inflammation and fibrosis. The treatment options are quite limited, and the therapeutic response is hindered by the fibrous membrane formed by excessive collagen and the rapid removal by synovial fluid. To address these challenges, we designed a hyaluronic acid/Pluronic F-127 (HP)-based injectable thermosensitive hydrogel as a drug carrier loaded with dexamethasone and collagenase (HPDC). We screened for an optimal HP hydrogel that can sustain drug release for approximately 10 days both in vitro and in vivo. In the meanwhile, we found that HP hydrogel could inhibit the proliferation and diminish the adhesion capacity of rat synovial cells induced by transforming growth factor-β1. Furthermore, using an established immobilization rat model of FS, intra-articular injection of HPDC significantly improved joint range of motion compared to medication alone. Relying on sustained drug release, the accumulated collagen fibers were degraded by collagenase to promote the deep delivery of dexamethasone. These findings showed a positive combined treatment effect of HPDC, providing a novel idea for the comprehensive treatment of FS.
Collapse
Affiliation(s)
- Jucong Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yanling Song
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Luwen Zhu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuchan You
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiahao Hu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinyi Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chen Wang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jingyi Lu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiying Shen
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China.
| | - Chong Teng
- Department of Orthopedic Surgery, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu 32200, China.
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China; Innovation Center of Translational Pharmacy, Jinhua Institute of Zhejiang University, Jinhua 321299, China.
| |
Collapse
|
7
|
Baig MMFA, Wong LK, Zia AW, Wu H. Development of biomedical hydrogels for rheumatoid arthritis treatment. Asian J Pharm Sci 2024; 19:100887. [PMID: 38419762 PMCID: PMC10900807 DOI: 10.1016/j.ajps.2024.100887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/16/2023] [Accepted: 11/05/2023] [Indexed: 03/02/2024] Open
Abstract
Rheumatoid Arthritis (RA) is an autoimmune disorder that hinders the normal functioning of bones and joints and reduces the quality of human life. Every year, millions of people are diagnosed with RA worldwide, particularly among elderly individuals and women. Therefore, there is a global need to develop new biomaterials, medicines and therapeutic methods for treating RA. This will improve the Healthcare Access and Quality Index and also relieve administrative and financial burdens on healthcare service providers at a global scale. Hydrogels are soft and cross-linked polymeric materials that can store a chunk of fluids, drugs and biomolecules for hydration and therapeutic applications. Hydrogels are biocompatible and exhibit excellent mechanical properties, such as providing elastic cushions to articulating joints by mimicking the natural synovial fluid. Hence, hydrogels create a natural biological environment within the synovial cavity to reduce autoimmune reactions and friction. Hydrogels also lubricate the articulating joint surfaces to prevent degradation of synovial surfaces of bones and cartilage, thus exhibiting high potential for treating RA. This work reviews the progress in injectable and implantable hydrogels, synthesis methods, types of drugs, advantages and challenges. Additionally, it discusses the role of hydrogels in targeted drug delivery, mechanistic behaviour and tribological performance for RA treatment.
Collapse
Affiliation(s)
| | - Lee Ki Wong
- Department of Chemistry, Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Abdul Wasy Zia
- Institute of Mechanical, Process and Energy Engineering (IMPEE), School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom
| | - Hongkai Wu
- Department of Chemistry, Hong Kong University of Science and Technology, Hong Kong 999077, China
| |
Collapse
|
8
|
Steplewski A, Fertala J, Cheng L, Wang ML, Rivlin M, Beredjiklian P, Fertala A. Evaluating the Efficacy of a Thermoresponsive Hydrogel for Delivering Anti-Collagen Antibodies to Reduce Posttraumatic Scarring in Orthopedic Tissues. Gels 2023; 9:971. [PMID: 38131957 PMCID: PMC10742524 DOI: 10.3390/gels9120971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Excessive posttraumatic scarring in orthopedic tissues, such as joint capsules, ligaments, tendons, muscles, and peripheral nerves, presents a significant medical problem, resulting in pain, restricted joint mobility, and impaired musculoskeletal function. Current treatments for excessive scarring are often ineffective and require the surgical removal of fibrotic tissue, which can aggravate the problem. The primary component of orthopedic scars is collagen I-rich fibrils. Our research team has developed a monoclonal anti-collagen antibody (ACA) that alleviates posttraumatic scarring by inhibiting collagen fibril formation. We previously established the safety and efficacy of ACA in a rabbit-based arthrofibrosis model. In this study, we evaluate the utility of a well-characterized thermoresponsive hydrogel (THG) as a delivery vehicle for ACA to injury sites. Crucial components of the hydrogel included N-isopropylacrylamide, poly(ethylene glycol) diacrylate, and hyaluronic acid. Our investigation focused on in vitro ACA release kinetics, stability, and activity. Additionally, we examined the antigen-binding characteristics of ACA post-release from the THG in an in vivo context. Our preliminary findings suggest that the THG construct exhibits promise as a delivery platform for antibody-based therapeutics to reduce excessive scarring in orthopedic tissues.
Collapse
Affiliation(s)
- Andrzej Steplewski
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jolanta Fertala
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Lan Cheng
- Department of Neurosciences, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mark L. Wang
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Michael Rivlin
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Pedro Beredjiklian
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Andrzej Fertala
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
9
|
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: 0] [Impact Index Per Article: 0] [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
|
10
|
Fani N, Peshkova M, Bikmulina P, Golroo R, Timashev P, Vosough M. Fabricating the cartilage: recent achievements. Cytotechnology 2023; 75:269-292. [PMID: 37389132 PMCID: PMC10299965 DOI: 10.1007/s10616-023-00582-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 05/09/2023] [Indexed: 07/01/2023] Open
Abstract
This review aims to describe the most recent achievements and provide an insight into cartilage engineering and strategies to restore the cartilage defects. Here, we discuss cell types, biomaterials, and biochemical factors applied to form cartilage tissue equivalents and update the status of fabrication techniques, which are used at all stages of engineering the cartilage. The actualized concept to improve the cartilage tissue restoration is based on applying personalized products fabricated using a full cycle platform: a bioprinter, a bioink consisted of ECM-embedded autologous cell aggregates, and a bioreactor. Moreover, in situ platforms can help to skip some steps and enable adjusting the newly formed tissue in the place during the operation. Only some achievements described have passed first stages of clinical translation; nevertheless, the number of their preclinical and clinical trials is expected to grow in the nearest future.
Collapse
Affiliation(s)
- Nesa Fani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maria Peshkova
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Polina Bikmulina
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, Moscow, Russia
| | - Reihaneh Golroo
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, Moscow, Russia
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
11
|
Chi XK, Xu XL, Chen BY, Su J, Du YZ. Combining nanotechnology with monoclonal antibody drugs for rheumatoid arthritis treatments. J Nanobiotechnology 2023; 21:105. [PMID: 36964609 PMCID: PMC10039584 DOI: 10.1186/s12951-023-01857-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/15/2023] [Indexed: 03/26/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic immune disease characterized by synovial inflammation. Patients with RA commonly experience significant damage to their hand and foot joints, which can lead to joint deformities and even disability. Traditional treatments have several clinical drawbacks, including unclear pharmacological mechanisms and serious side effects. However, the emergence of antibody drugs offers a promising approach to overcome these limitations by specifically targeting interleukin-1 (IL-1), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and other cytokines that are closely related to the onset of RA. This approach reduces the incidence of adverse effects and contributes to significant therapeutic outcomes. Furthermore, combining these antibody drugs with drug delivery nanosystems (DDSs) can improve their tissue accumulation and bioavailability.Herein, we provide a summary of the pathogenesis of RA, the available antibody drugs and DDSs that improve the efficacy of these drugs. However, several challenges need to be addressed in their clinical applications, including patient compliance, stability, immunogenicity, immunosupression, target and synergistic effects. We propose strategies to overcome these limitations. In summary, we are optimistic about the prospects of treating RA with antibody drugs, given their specific targeting mechanisms and the potential benefits of combining them with DDSs.
Collapse
Affiliation(s)
- Xiao-Kai Chi
- College of Pharmacy, Jiamusi University, 258 Xuefu Road, Jiamusi, 154007, China
- Shulan International Medical College, Zhejiang Shuren University), 8 Shuren Street, Hangzhou, 310015, China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University), 8 Shuren Street, Hangzhou, 310015, China.
| | - Bang-Yao Chen
- Shulan International Medical College, Zhejiang Shuren University), 8 Shuren Street, Hangzhou, 310015, China
| | - Jin Su
- College of Pharmacy, Jiamusi University, 258 Xuefu Road, Jiamusi, 154007, China.
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China.
| |
Collapse
|
12
|
Yi J, Liu Y, Xie H, An H, Li C, Wang X, Chai W. Hydrogels for the treatment of rheumatoid arthritis. Front Bioeng Biotechnol 2022; 10:1014543. [PMID: 36312537 PMCID: PMC9597294 DOI: 10.3389/fbioe.2022.1014543] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/02/2022] [Indexed: 12/03/2022] Open
Abstract
Rheumatoid Arthritis is a universal disease that severely affects the normal function of human joints and the quality of life. Millions of people around the world are diagnosed with rheumatoid arthritis every year, carrying a substantial burden for both the individual and society. Hydrogel is a polymer material with good mechanical properties and biocompatibility, which shows great potential in the treatment of rheumatoid arthritis. With the progress of tissue engineering and biomedical material technology in recent years, more and more studies focus on the application of hydrogels in rheumatoid arthritis. We reviewed the progress of hydrogels applied in rheumatoid arthritis in recent years. Also, the needed comprehensive performance and current applications of therapeutic hydrogels based on the complex pathophysiological characteristics of rheumatoid arthritis are also concluded. Additionally, we proposed the challenges and difficulties in the application of hydrogels in rheumatoid arthritis and put forward some prospects for the future research.
Collapse
Affiliation(s)
- Jiafeng Yi
- Senior Department of Orthopedics, Fourth Medical Center of People’s Liberation Army General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
- National Clinical Research Center for Orthopaedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Yubo Liu
- Senior Department of Orthopedics, Fourth Medical Center of People’s Liberation Army General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
- National Clinical Research Center for Orthopaedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Hongbin Xie
- Senior Department of Orthopedics, Fourth Medical Center of People’s Liberation Army General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
- National Clinical Research Center for Orthopaedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Haoming An
- Senior Department of Orthopedics, Fourth Medical Center of People’s Liberation Army General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
- National Clinical Research Center for Orthopaedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Chao Li
- Senior Department of Orthopedics, Fourth Medical Center of People’s Liberation Army General Hospital, Beijing, China
- National Clinical Research Center for Orthopaedics, Sports Medicine and Rehabilitation, Beijing, China
- *Correspondence: Chao Li, ; Xing Wang, ; Wei Chai,
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Chao Li, ; Xing Wang, ; Wei Chai,
| | - Wei Chai
- Senior Department of Orthopedics, Fourth Medical Center of People’s Liberation Army General Hospital, Beijing, China
- National Clinical Research Center for Orthopaedics, Sports Medicine and Rehabilitation, Beijing, China
- *Correspondence: Chao Li, ; Xing Wang, ; Wei Chai,
| |
Collapse
|
13
|
Alternative Routes of Administration for Therapeutic Antibodies—State of the Art. Antibodies (Basel) 2022; 11:antib11030056. [PMID: 36134952 PMCID: PMC9495858 DOI: 10.3390/antib11030056] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/11/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Background: For the past two decades, there has been a huge expansion in the development of therapeutic antibodies, with 6 to 10 novel entities approved each year. Around 70% of these Abs are delivered through IV injection, a mode of administration allowing rapid and systemic delivery of the drug. However, according to the evidence presented in the literature, beyond the reduction of invasiveness, a better efficacy can be achieved with local delivery. Consequently, efforts have been made toward the development of innovative methods of administration, and in the formulation and engineering of novel Abs to improve their therapeutic index. Objective: This review presents an overview of the routes of administration used to deliver Abs, different from the IV route, whether approved or in the clinical evaluation stage. We provide a description of the physical and biological fundamentals for each route of administration, highlighting their relevance with examples of clinically-relevant Abs, and discussing their strengths and limitations. Methods: We reviewed and analyzed the current literature, published as of the 1 April 2022 using MEDLINE and EMBASE databases, as well as the FDA and EMA websites. Ongoing trials were identified using clinicaltrials.gov. Publications and data were identified using a list of general keywords. Conclusions: Apart from the most commonly used IV route, topical delivery of Abs has shown clinical successes, improving drug bioavailability and efficacy while reducing side-effects. However, additional research is necessary to understand the consequences of biological barriers associated with local delivery for Ab partitioning, in order to optimize delivery methods and devices, and to adapt Ab formulation to local delivery. Novel modes of administration for Abs might in fine allow a better support to patients, especially in the context of chronic diseases, as well as a reduction of the treatment cost.
Collapse
|
14
|
Thermosensitive Injectable Hydrogels for Intra-Articular Delivery of Etanercept for the Treatment of Osteoarthritis. Gels 2022; 8:gels8080488. [PMID: 36005089 PMCID: PMC9407145 DOI: 10.3390/gels8080488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022] Open
Abstract
The intra-articular administration of drugs has attracted great interest in recent decades for the treatment of osteoarthritis. The use of modified drugs has also attracted interest in recent years because their intra-articular administration has demonstrated encouraging results. The objective of this work was to prepare injectable-thermosensitive hydrogels for the intra-articular administration of Etanercept (ETA), an inhibitor of tumor necrosis factor-α. Hydrogels were prepared from the physical mixture of chitosan and Pluronic F127 with β-glycerolphosphate (BGP). Adding β-glycerolphosphate to the system reduced the gelation time and also modified the morphology of the resulting material. In vitro studies were carried out to determine the cytocompatibility of the prepared hydrogels for the human chondrocyte line C28/I2. The in vitro release study showed that the incorporation of BGP into the system markedly modified the release of ETA. In the in vivo studies, it was verified that the hydrogels remained inside the implantation site in the joint until the end of the study. Furthermore, ETA was highly concentrated in the blood of the study mice 48 h after the loaded material was injected. Histological investigation of osteoarthritic knees showed that the material promotes cartilage recovery in osteoarthritic mice. The results demonstrate the potential of ETA-loaded injectable hydrogels for the localized treatment of joints.
Collapse
|
15
|
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: 0] [Impact Index Per Article: 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
|
16
|
Spinal microglia-derived TNF promotes the astrocytic JNK/CXCL1 pathway activation in a mouse model of burn pain. Brain Behav Immun 2022; 102:23-39. [PMID: 35143878 DOI: 10.1016/j.bbi.2022.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/28/2022] [Accepted: 02/04/2022] [Indexed: 11/21/2022] Open
Abstract
Burn injury-induced pain (BIP) is an extremely complicated condition usually resistant to analgesic drugs, while its pathogenesis remains unknown. Considerable attention has been attracted to elucidate the glial mechanisms in chronic pain. In this study, we initiatively used a mouse model of second-degree BIP to investigate the underlying non-neuronal mechanisms at the spinal cord level. Our behavioral results showed that hind-paw burn injury caused persistent allodynia and hyperalgesia for 2 weeks in mice. Further studies revealed that both microglia and astrocytes activated in a spatially- and temporally-dependent manner in spinal cord after burn injury. In addition, the phosphorylated p38 mitogen-activated protein kinase (MAPK)-mediated tumor necrosis factor (TNF) release in spinal microglia is essentially attributed to the early stage of BIP, while the c-Jun N-terminal kinase (JNK) MAPK-dependent chemokine CXCL1 expression is mainly involved in the maintenance of pain hypersensitivity. Most strikingly, burn injury-induced pain symptoms and the activation of astrocytes were significantly suppressed by TNF inhibitor Thalidomide. On the contrary, intrathecal injection of TNF caused apparent pain hypersensitivity, accompanied by the activation of astrocytes and the upregulation of CXCL1 via the JNK MAPK signaling pathway, indicating that TNF is the key cytokine in the interaction between microglia and astrocytes at the spinal level. Moreover, treatment with the CXCR2 receptor antagonist SB225002 to block the biological activities of CXCL1 significantly attenuated the mechanical allodynia and thermal hyperalgesia in this BIP model. Taken together, this study indicates that intervention of glial pathways provides a new perspective in the management of BIP.
Collapse
|
17
|
Gupta A, Lee J, Ghosh T, Nguyen VQ, Dey A, Yoon B, Um W, Park JH. Polymeric Hydrogels for Controlled Drug Delivery to Treat Arthritis. Pharmaceutics 2022; 14:540. [PMID: 35335915 PMCID: PMC8948938 DOI: 10.3390/pharmaceutics14030540] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 12/31/2022] Open
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) are disabling musculoskeletal disorders that affect joints and cartilage and may lead to bone degeneration. Conventional delivery of anti-arthritic agents is limited due to short intra-articular half-life and toxicities. Innovations in polymer chemistry have led to advancements in hydrogel technology, offering a versatile drug delivery platform exhibiting tissue-like properties with tunable drug loading and high residence time properties This review discusses the advantages and drawbacks of polymeric materials along with their modifications as well as their applications for fabricating hydrogels loaded with therapeutic agents (small molecule drugs, immunotherapeutic agents, and cells). Emphasis is given to the biological potentialities of hydrogel hybrid systems/micro-and nanotechnology-integrated hydrogels as promising tools. Applications for facile tuning of therapeutic drug loading, maintaining long-term release, and consequently improving therapeutic outcome and patient compliance in arthritis are detailed. This review also suggests the advantages, challenges, and future perspectives of hydrogels loaded with anti-arthritic agents with high therapeutic potential that may alter the landscape of currently available arthritis treatment modalities.
Collapse
Affiliation(s)
- Anuradha Gupta
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Jungmi Lee
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Torsha Ghosh
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Van Quy Nguyen
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Anup Dey
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Been Yoon
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Wooram Um
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
18
|
Ghorbaninezhad F, Leone P, Alemohammad H, Najafzadeh B, Nourbakhsh NS, Prete M, Malerba E, Saeedi H, Tabrizi NJ, Racanelli V, Baradaran B. Tumor necrosis factor‑α in systemic lupus erythematosus: Structure, function and therapeutic implications (Review). Int J Mol Med 2022; 49:43. [PMID: 35137914 DOI: 10.3892/ijmm.2022.5098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/05/2022] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor‑α (TNF‑α) is a pleiotropic pro‑inflammatory cytokine that contributes to the pathophysiology of several autoimmune diseases, such as multiple sclerosis, inflammatory bowel disease, rheumatoid arthritis, psoriatic arthritis and systemic lupus erythematosus (SLE). The specific role of TNF‑α in autoimmunity is not yet fully understood however, partially, in a complex disease such as SLE. Through the engagement of the TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2), both the two variants, soluble and transmembrane TNF‑α, can exert multiple biological effects according to different settings. They can either function as immune regulators, impacting B‑, T‑ and dendritic cell activity, modulating the autoimmune response, or as pro‑inflammatory mediators, regulating the induction and maintenance of inflammatory processes in SLE. The present study reviews the dual role of TNF‑α, focusing on the different effects that TNF‑α may have on the pathogenesis of SLE. In addition, the efficacy and safety of anti‑TNF‑α therapies in preclinical and clinical trials SLE are discussed.
Collapse
Affiliation(s)
- Farid Ghorbaninezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan 5165665811, Iran
| | - Patrizia Leone
- Department of Biomedical Sciences and Human Oncology, 'Aldo Moro' University of Bari Medical School, I‑70124 Bari, Italy
| | - Hajar Alemohammad
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, East Azerbaijan 5166616471, Iran
| | - Basira Najafzadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, East Azerbaijan 5166616471, Iran
| | - Niloufar Sadat Nourbakhsh
- Department of Genetics, Faculty of Basic Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Fars 7319846451, Iran
| | - Marcella Prete
- Department of Biomedical Sciences and Human Oncology, 'Aldo Moro' University of Bari Medical School, I‑70124 Bari, Italy
| | - Eleonora Malerba
- Department of Biomedical Sciences and Human Oncology, 'Aldo Moro' University of Bari Medical School, I‑70124 Bari, Italy
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan 5165665811, Iran
| | - Neda Jalili Tabrizi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan 5165665811, Iran
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, 'Aldo Moro' University of Bari Medical School, I‑70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan 5165665811, Iran
| |
Collapse
|
19
|
Zhu Y, Ye L, Cai X, Li Z, Fan Y, Yang F. Icariin-Loaded Hydrogel Regulates Bone Marrow Mesenchymal Stem Cell Chondrogenic Differentiation and Promotes Cartilage Repair in Osteoarthritis. Front Bioeng Biotechnol 2022; 10:755260. [PMID: 35223781 PMCID: PMC8864219 DOI: 10.3389/fbioe.2022.755260] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 01/07/2022] [Indexed: 02/06/2023] Open
Abstract
Intra-articular injection of mesenchymal stem cells is a potential therapeutic strategy for cartilage protection and symptom relief for osteoarthritis (OA). However, controlling chondrogenesis of the implanted cells in the articular cavity remains a challenge. In this study, hydrogels containing different concentrations of icariin were prepared by in situ crosslinking of hyaluronic acid and Poloxamer 407. This injectable and thermoresponsive hydrogel, as a 3D cell culture system, showed good biocompatibility with chondrocytes and bone marrow mesenchymal stem cells (BMSCs), as well as promoted proliferation and chondrogenesis of BMSCs through the Wnt/β-catenin signaling pathway. Intra-articular injection of this kind of BMSC-loaded composite hydrogel can significantly prevent cartilage destruction by inducing chondrogenic differentiation of BMSCs, and relieve pain through regulating the expression of inflammatory cytokines (e.g., IL-10 and MMP-13) in the OA model. Incorporating BMSCs into this novel icariin-loaded hydrogel indicates a more superior efficacy than the single BMSC injection, which suggests a great potential for its application in OA.
Collapse
Affiliation(s)
- Yuefeng Zhu
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Le Ye
- Department of Pain, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoxi Cai
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zuhao Li
- Department of Pain, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yongqian Fan
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Fengjian Yang
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
20
|
Singla P, Garg S, McClements J, Jamieson O, Peeters M, Mahajan RK. Advances in the therapeutic delivery and applications of functionalized Pluronics: A critical review. Adv Colloid Interface Sci 2022; 299:102563. [PMID: 34826745 DOI: 10.1016/j.cis.2021.102563] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/15/2021] [Accepted: 11/13/2021] [Indexed: 12/22/2022]
Abstract
Pluronic (PEO-PPO-PEO) block copolymers can form nano-sized micelles with a structure composed of a hydrophobic PPO core and hydrophilic PEO shell layer. Pluronics are U.S. Food and Drug Administration approved polymers, which are widely used for solubilization of drugs and their delivery, gene/therapeutic delivery, diagnostics, and tissue engineering applications due to their non-ionic properties, non-toxicity, micelle forming ability, excellent biocompatibility and biodegradability. Although Pluronics have been employed as drug carrier systems for several decades, numerous issues such as rapid dissolution, shorter residence time in biological media, fast clearance and weak mechanical strength have hindered their efficacy. Pluronics have been functionalized with pH-sensitive, biological-responsive moieties, antibodies, aptamers, folic acid, drugs, different nanoparticles, and photo/thermo-responsive hydrogels. These functionalization strategies enable Pluronics to act as stimuli responsive and targeted drug delivery vehicles. Moreover, Pluronics have emerged in nano-emulsion formulations and have been utilized to improve the properties of cubosomes, dendrimers and nano-sheets, including their biocompatibility and aqueous solubility. Functionalization of Pluronics results in the significant improvement of target specificity, loading capacity, biocompatibility of nanoparticles and stimuli responsive hydrogels for the promising delivery of a range of drugs. Therefore, this review presents an overview of all advancements (from the last 15 years) in functionalized Pluronics, providing a valuable tool for industry and academia in order to optimize their use in drug or therapeutic delivery, in addition to several other biomedical applications.
Collapse
Affiliation(s)
- Pankaj Singla
- School of Engineering, Merz Court, Claremont Road, Newcastle University, Newcastle Upon Tyne NE1 7RU, United Kingdom
| | - Saweta Garg
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, India
| | - Jake McClements
- School of Engineering, Merz Court, Claremont Road, Newcastle University, Newcastle Upon Tyne NE1 7RU, United Kingdom
| | - Oliver Jamieson
- School of Engineering, Merz Court, Claremont Road, Newcastle University, Newcastle Upon Tyne NE1 7RU, United Kingdom
| | - Marloes Peeters
- School of Engineering, Merz Court, Claremont Road, Newcastle University, Newcastle Upon Tyne NE1 7RU, United Kingdom.
| | - Rakesh Kumar Mahajan
- Department of Chemistry, UGC-Centre for Advanced Studies-I, Guru Nanak Dev University, Amritsar 143005, India.
| |
Collapse
|
21
|
Tu AB, Lewis JS. Biomaterial-based immunotherapeutic strategies for rheumatoid arthritis. Drug Deliv Transl Res 2021; 11:2371-2393. [PMID: 34414564 PMCID: PMC8376117 DOI: 10.1007/s13346-021-01038-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2021] [Indexed: 02/08/2023]
Abstract
Rheumatoid arthritis (RA) is an extremely painful autoimmune disease characterized by chronic joint inflammation leading to the erosion of adjacent cartilage and bone. Rheumatoid arthritis pathology is primarily driven by inappropriate infiltration and activation of immune cells within the synovium of the joint. There is no cure for RA. As such, manifestation of symptoms entails lifelong management via various therapies that aim to generally dampen the immune system or impede the function of immune mediators. However, these treatment strategies lead to adverse effects such as toxicity, general immunosuppression, and increased risk of infection. In pursuit of safer and more efficacious therapies, many emerging biomaterial-based strategies are being developed to improve payload delivery, specific targeting, and dose efficacy, and to mitigate adverse reactions and toxicity. In this review, we highlight biomaterial-based approaches that are currently under investigation to circumvent the limitations of conventional RA treatments.
Collapse
Affiliation(s)
- Allen B Tu
- Department of Biomedical Engineering, University of California, 1 Shields Ave, Davis , CA, 95616, USA
| | - Jamal S Lewis
- Department of Biomedical Engineering, University of California, 1 Shields Ave, Davis , CA, 95616, USA.
| |
Collapse
|
22
|
Al Ojaimi Y, Blin T, Lamamy J, Gracia M, Pitiot A, Denevault-Sabourin C, Joubert N, Pouget JP, Gouilleux-Gruart V, Heuzé-Vourc'h N, Lanznaster D, Poty S, Sécher T. Therapeutic antibodies - natural and pathological barriers and strategies to overcome them. Pharmacol Ther 2021; 233:108022. [PMID: 34687769 PMCID: PMC8527648 DOI: 10.1016/j.pharmthera.2021.108022] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 02/06/2023]
Abstract
Antibody-based therapeutics have become a major class of therapeutics with over 120 recombinant antibodies approved or under review in the EU or US. This therapeutic class has experienced a remarkable expansion with an expected acceleration in 2021-2022 due to the extraordinary global response to SARS-CoV2 pandemic and the public disclosure of over a hundred anti-SARS-CoV2 antibodies. Mainly delivered intravenously, alternative delivery routes have emerged to improve antibody therapeutic index and patient comfort. A major hurdle for antibody delivery and efficacy as well as the development of alternative administration routes, is to understand the different natural and pathological barriers that antibodies face as soon as they enter the body up to the moment they bind to their target antigen. In this review, we discuss the well-known and more under-investigated extracellular and cellular barriers faced by antibodies. We also discuss some of the strategies developed in the recent years to overcome these barriers and increase antibody delivery to its site of action. A better understanding of the biological barriers that antibodies have to face will allow the optimization of antibody delivery near its target. This opens the way to the development of improved therapy with less systemic side effects and increased patients' adherence to the treatment.
Collapse
Affiliation(s)
- Yara Al Ojaimi
- UMR 1253, iBrain, Inserm, 37000 Tours, France; University of Tours, 37000 Tours, France
| | - Timothée Blin
- University of Tours, 37000 Tours, France; UMR 1100, CEPR, Inserm, 37000 Tours, France
| | - Juliette Lamamy
- University of Tours, 37000 Tours, France; GICC, EA7501, 37000 Tours, France
| | - Matthieu Gracia
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier F-34298, France
| | - Aubin Pitiot
- University of Tours, 37000 Tours, France; UMR 1100, CEPR, Inserm, 37000 Tours, France
| | | | - Nicolas Joubert
- University of Tours, 37000 Tours, France; GICC, EA7501, 37000 Tours, France
| | - Jean-Pierre Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier F-34298, France
| | | | | | - Débora Lanznaster
- UMR 1253, iBrain, Inserm, 37000 Tours, France; University of Tours, 37000 Tours, France
| | - Sophie Poty
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier F-34298, France
| | - Thomas Sécher
- University of Tours, 37000 Tours, France; UMR 1100, CEPR, Inserm, 37000 Tours, France
| |
Collapse
|
23
|
Sánchez-Robles EM, Girón R, Paniagua N, Rodríguez-Rivera C, Pascual D, Goicoechea C. Monoclonal Antibodies for Chronic Pain Treatment: Present and Future. Int J Mol Sci 2021; 22:ijms221910325. [PMID: 34638667 PMCID: PMC8508878 DOI: 10.3390/ijms221910325] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022] Open
Abstract
Chronic pain remains a major problem worldwide, despite the availability of various non-pharmacological and pharmacological treatment options. Therefore, new analgesics with novel mechanisms of action are needed. Monoclonal antibodies (mAbs) are directed against specific, targeted molecules involved in pain signaling and processing pathways that look to be very effective and promising as a novel therapy in pain management. Thus, there are mAbs against tumor necrosis factor (TNF), nerve growth factor (NGF), calcitonin gene-related peptide (CGRP), or interleukin-6 (IL-6), among others, which are already recommended in the treatment of chronic pain conditions such as osteoarthritis, chronic lower back pain, migraine, or rheumatoid arthritis that are under preclinical research. This narrative review summarizes the preclinical and clinical evidence supporting the use of these agents in the treatment of chronic pain.
Collapse
|
24
|
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: 31] [Impact Index Per Article: 10.3] [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
|
25
|
Ultra-Low Dose Cytokines in Rheumatoid Arthritis, Three Birds with One Stone as the Rationale of the 2LARTH ® Micro-Immunotherapy Treatment. Int J Mol Sci 2021; 22:ijms22136717. [PMID: 34201546 PMCID: PMC8268272 DOI: 10.3390/ijms22136717] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/15/2021] [Accepted: 06/20/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) are two cytokines involved in the perpetuation of the chronic inflammation state characterizing rheumatoid arthritis (RA). Significant advances in the treatment of this pathology have been made over the past ten years, partially through the development of anti-TNF and anti-IL-1 therapies. However, major side effects still persist and new alternative therapies should be considered. The formulation of the micro-immunotherapy medicine (MIM) 2LARTH® uses ultra-low doses (ULD) of TNF-α, IL-1β, and IL-2, in association with other immune factors, to gently restore the body’s homeostasis. The first part of this review aims at delineating the pivotal roles played by IL-1β and TNF-α in RA physiopathology, leading to the development of anti-TNF and anti-IL-1 therapeutic agents. In a second part, an emphasis will be made on explaining the rationale of using multiple therapeutic targets, including both IL-1β and TNF-α in 2LARTH® medicine. Particular attention will be paid to the ULD of those two main pro-inflammatory factors in order to counteract their overexpression through the lens of their molecular implication in RA pathogenesis.
Collapse
|
26
|
Dehshahri A, Kumar A, Madamsetty VS, Uzieliene I, Tavakol S, Azedi F, Fekri HS, Zarrabi A, Mohammadinejad R, Thakur VK. New Horizons in Hydrogels for Methotrexate Delivery. Gels 2020; 7:2. [PMID: 33396629 PMCID: PMC7839000 DOI: 10.3390/gels7010002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022] Open
Abstract
Since its first clinical application, methotrexate (MTX) has been widely used for the treatment of human diseases. Despite great advantages, some properties such as poor absorption, short plasma half-life and unpredictable bioavailability have led researchers to seek novel delivery systems to improve its characteristics for parenteral and oral administration. Recently, great attention has been directed to hydrogels for the preparation of MTX formulations. This review describes the potential of hydrogels for the formulation of MTX to treat cancer, rheumatoid arthritis, psoriasis and central nervous system diseases. We will delineate the state-of-the-art and promising potential of hydrogels for systemic MTX delivery as well as transdermal delivery of the drug-using hydrogel-based formulations.
Collapse
Affiliation(s)
- Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran;
| | - Anuj Kumar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Korea;
| | - Vijay Sagar Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA;
| | - Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08406 Vilnius, Lithuania;
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614525, Iran; (S.T.); (F.A.)
| | - Fereshteh Azedi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614525, Iran; (S.T.); (F.A.)
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Hojjat Samareh Fekri
- Student Research Committee, Kerman University of Medical Sciences, Kerman 7619813159, Iran;
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616911319, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey;
| | - Reza Mohammadinejad
- Research Center for Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman 7618866749, Iran
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Center, Scotland’s Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh EH9 3JG, UK
| |
Collapse
|