1
|
Makled S, Abbas H, Ali ME, Zewail M. Melatonin hyalurosomes in collagen thermosensitive gel as a potential repurposing approach for rheumatoid arthritis management via the intra-articular route. Int J Pharm 2024; 661:124449. [PMID: 38992734 DOI: 10.1016/j.ijpharm.2024.124449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/30/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
Despite the fact that several rheumatoid arthritis treatments have been utilized, none of them achieved complete joint healing and has been accompanied by several side effects that compromise patient compliance. This study aims to provide an effective safe RA treatment with minimum side effects through the encapsulation of melatonin (MEL) in hyalurosomes and loading these hyalurosomes in collagen thermos-sensitive poloxamer 407 (PCO) hydrogels, followed by their intra-articular administration in AIA model rats. In vitro characterization of MEL-hyalurosomes and PCO hydrogel along with in vivo evaluation of the selected formulation were conducted. Particle size, PDI and EE % of the selected formulation were 71.5 nm, 0.09 and 90 %. TEM micrographs demonstrated that the particles had spherical shape with no aggregation signs. Loading PCO hydrogels with MEL-hyalurosomes did not cause significant changes in pH although it increased its viscosity and injection time. FTIR analysis showed that no interactions were noted among the delivery system components. In vivo results revealed the superior effect of MEL-hyalurosomes PCO hydrogel over MEL-PCO hydrogel and blank PCO hydrogels in improving joint healing, cartilage repair, pannus formation and cell infiltrations. Also, MEL-hyalurosomes PCO hydrogel group showed comparable levels of TNF-α, IL1, MDA, NRF2 and HO-1 with the negative control group. These findings highlight the MEL encapsulation role in augmenting its pharmacological effects along with the synergistic effect of hyaluronic acid in hyalurosomes and collagen in PCO hydrogel in promoting joint healing.
Collapse
Affiliation(s)
- Shaimaa Makled
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, 21521, Egypt
| | - Haidy Abbas
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Egypt P.O. Box 22511, Damanhour, Egypt.
| | - Merhan E Ali
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Mariam Zewail
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Egypt P.O. Box 22511, Damanhour, Egypt
| |
Collapse
|
2
|
Ren S, Xu Y, Dong X, Mu Q, Chen X, Yu Y, Su G. Nanotechnology-empowered combination therapy for rheumatoid arthritis: principles, strategies, and challenges. J Nanobiotechnology 2024; 22:431. [PMID: 39034407 PMCID: PMC11265020 DOI: 10.1186/s12951-024-02670-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with multifactorial etiology and intricate pathogenesis. In RA, repeated monotherapy is frequently associated with inadequate efficacy, drug resistance, and severe side effects. Therefore, a shift has occurred in clinical practice toward combination therapy. However, conventional combination therapy encounters several hindrances, including low selectivity to arthritic joints, short half-lives, and varying pharmacokinetics among coupled drugs. Emerging nanotechnology offers an incomparable opportunity for developing advanced combination therapy against RA. First, it allows for co-delivering multiple drugs with augmented physicochemical properties, targeted delivery capabilities, and controlled release profiles. Second, it enables therapeutic nanomaterials development, thereby expanding combination regimens to include multifunctional nanomedicines. Lastly, it facilitates the construction of all-in-one nanoplatforms assembled with multiple modalities, such as phototherapy, sonodynamic therapy, and imaging. Thus, nanotechnology offers a promising solution to the current bottleneck in both RA treatment and diagnosis. This review summarizes the rationale, advantages, and recent advances in nano-empowered combination therapy for RA. It also discusses safety considerations, drug-drug interactions, and the potential for clinical translation. Additionally, it provides design tips and an outlook on future developments in nano-empowered combination therapy. The objective of this review is to achieve a comprehensive understanding of the mechanisms underlying combination therapy for RA and unlock the maximum potential of nanotechnology, thereby facilitating the smooth transition of research findings from the laboratory to clinical practice.
Collapse
Affiliation(s)
- Shujing Ren
- Department of Pharmacy, Affiliated Hospital 2 of Nantong University, Nantong, 226000, PR China
| | - Yuhang Xu
- School of Pharmacy, Nantong University, Nantong, 226000, PR China
| | - Xingpeng Dong
- School of Pharmacy, Nantong University, Nantong, 226000, PR China
| | - Qingxin Mu
- Department of Pharmaceutics, University of Washington, Seattle, WA, 98195, USA
| | - Xia Chen
- Department of Pharmacy, Affiliated Hospital 2 of Nantong University, Nantong, 226000, PR China.
| | - Yanyan Yu
- School of Pharmacy, Nantong University, Nantong, 226000, PR China.
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong, 226000, PR China.
| |
Collapse
|
3
|
An X, Yang J, Cui X, Zhao J, Jiang C, Tang M, Dong Y, Lin L, Li H, Wang F. Advances in local drug delivery technologies for improved rheumatoid arthritis therapy. Adv Drug Deliv Rev 2024; 209:115325. [PMID: 38670229 DOI: 10.1016/j.addr.2024.115325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/25/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease characterized by an inflammatory microenvironment and cartilage erosion within the joint cavity. Currently, antirheumatic agents yield significant outcomes in RA treatment. However, their systemic administration is limited by inadequate drug retention in lesion areas and non-specific tissue distribution, reducing efficacy and increasing risks such as infection due to systemic immunosuppression. Development in local drug delivery technologies, such as nanostructure-based and scaffold-assisted delivery platforms, facilitate enhanced drug accumulation at the target site, controlled drug release, extended duration of the drug action, reduced both dosage and administration frequency, and ultimately improve therapeutic outcomes with minimized damage to healthy tissues. In this review, we introduced pathogenesis and clinically used therapeutic agents for RA, comprehensively summarized locally administered nanostructure-based and scaffold-assisted drug delivery systems, aiming at improving the therapeutic efficiency of RA by alleviating the inflammatory response, preventing bone erosion and promoting cartilage regeneration. In addition, the challenges and future prospects of local delivery for clinical translation in RA are discussed.
Collapse
Affiliation(s)
- Xiaoran An
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Jiapei Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Xiaolin Cui
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Jiaxuan Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Chenwei Jiang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Minglu Tang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Yabing Dong
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Longfei Lin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Hui Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China; Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330000, PR China
| | - Feihu Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China.
| |
Collapse
|
4
|
Li JJ, Li L, Su SS, Liao ML, Gong QZ, Liu M, Jiang S, Zhang ZQ, Zhou H, Liu JX. Anti-inflammatory properties and characterization of water extracts obtained from Callicarpa kwangtungensis Chun using in vitro and in vivo rat models. Sci Rep 2024; 14:11047. [PMID: 38744989 PMCID: PMC11094131 DOI: 10.1038/s41598-024-61892-9] [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: 12/29/2023] [Accepted: 05/10/2024] [Indexed: 05/16/2024] Open
Abstract
Callicarpa kwangtungensis Chun (CK) is a common remedy exhibits anti-inflammatory properties and has been used in Chinese herbal formulations, such as KangGongYan tablets. It is the main component of KangGongYan tablets, which has been used to treat chronic cervicitis caused by damp heat, red and white bands, cervical erosion, and bleeding. However, the anti-inflammatory effects of CK water extract remains unknown. This study assessed the anti-inflammatory effects of CK in vivo and in vitro, characterized its main components in the serum of rats and verified the anti-inflammatory effects of serum containing CK. Nitric oxide (NO), tumour necrosis factor α (TNF-α) and interleukin-6 (IL-6) release by RAW264.7 cells was examined by ELISA and Griess reagents. Inflammation-related protein expression in LPS-stimulated RAW264.7 cells was measured by western blotting. Furthermore, rat model of foot swelling induced by λ-carrageenan and a collagen-induced arthritis (CIA) rat model were used to explore the anti-inflammatory effects of CK. The components of CK were characterized by LC-MS, and the effects of CK-containing serum on proinflammatory factors levels and the expression of inflammation-related proteins were examined by ELISA, Griess reagents and Western blotting. CK suppressed IL-6, TNF-α, and NO production, and iNOS protein expression in LPS-stimulated RAW264.7 cells. Mechanistic studies showed that CK inhibited the phosphorylation of ERK, P38 and JNK in the MAPK signaling pathway, promoted the expression of IκBα in the NF-κB signaling pathway, and subsequently inhibited the expression of iNOS, thereby exerting anti-inflammatory effects. Moreover, CK reduced the swelling rates with λ-carrageenan induced foot swelling, and reduced the arthritis score and incidence in the collagen-induced arthritis (CIA) rat model. A total of 68 compounds in CK water extract and 31 components in rat serum after intragastric administration of CK were characterized. Serum pharmacological analysis showed that CK-containing serum suppressed iNOS protein expression and NO, TNF-α, and IL-6 release. CK may be an anti-inflammatory agent with therapeutic potential for acute and chronic inflammatory diseases, especially inflammatory diseases associated with MAPK activation.
Collapse
Affiliation(s)
- Jun-Jian Li
- School of Pharmaceutical Sciences, School of Basic Medical Sciences, Hunan Provincial Key Laboratory of Dong Medicine, Hunan University of Medicine, Huaihua, China
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Li Li
- School of Pharmaceutical Sciences, School of Basic Medical Sciences, Hunan Provincial Key Laboratory of Dong Medicine, Hunan University of Medicine, Huaihua, China
| | - Shan-Shan Su
- School of Pharmaceutical Sciences, School of Basic Medical Sciences, Hunan Provincial Key Laboratory of Dong Medicine, Hunan University of Medicine, Huaihua, China
| | - Mei-Lan Liao
- School of Pharmaceutical Sciences, School of Basic Medical Sciences, Hunan Provincial Key Laboratory of Dong Medicine, Hunan University of Medicine, Huaihua, China
| | - Qiu-Zi Gong
- School of Pharmaceutical Sciences, School of Basic Medical Sciences, Hunan Provincial Key Laboratory of Dong Medicine, Hunan University of Medicine, Huaihua, China
| | - Mei Liu
- School of Pharmaceutical Sciences, School of Basic Medical Sciences, Hunan Provincial Key Laboratory of Dong Medicine, Hunan University of Medicine, Huaihua, China
- School of Pharmaceutical Science, University of South China, Hengyang, China
| | - Shan Jiang
- School of Pharmaceutical Sciences, School of Basic Medical Sciences, Hunan Provincial Key Laboratory of Dong Medicine, Hunan University of Medicine, Huaihua, China
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Zai-Qi Zhang
- School of Pharmaceutical Sciences, School of Basic Medical Sciences, Hunan Provincial Key Laboratory of Dong Medicine, Hunan University of Medicine, Huaihua, China.
| | - Hua Zhou
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab On Chinese Medicine and Immune Disease Research, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Second Affiliated Hospital of Gzangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Jian-Xin Liu
- School of Pharmaceutical Sciences, School of Basic Medical Sciences, Hunan Provincial Key Laboratory of Dong Medicine, Hunan University of Medicine, Huaihua, China.
| |
Collapse
|
5
|
Dang LH, Vu NQ, Nguyen TT, Do THT, Pham TKT, Tran NQ. Thermally-responsive and reduced glutathione-sensitive folate-targeted nanocarrier based on alginate and pluronic F127 for on-demand release of methotrexate. Int J Biol Macromol 2024; 263:130227. [PMID: 38378121 DOI: 10.1016/j.ijbiomac.2024.130227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/08/2024] [Accepted: 02/14/2024] [Indexed: 02/22/2024]
Abstract
A specific rheumatoid arthritis (RA)-microenvironment-triggered nanocarrier for RA treatment of a first-line antirheumatic drug (Methotrexate, MTX) has been proposed. Reduced glutathione (GSH) responsivity, cystamine, was first introduced on the alginate backbone, which was then used as the bridge to connect pluronic F127 (temperature-responsive factor) and folic acid (targeting factor for active immune cells), resulting in dual-responsive triggered targeting carrier, PCAC-FA. In vitro study demonstrated that PCAC-FA was preferentially taken up by activated macrophage cells rather than normal ones, suggesting the targeting of PCAC-FA to inflamed tissue. The loading capacity of the designed carrier was 21.23 ± 0.91 %. MTX from the PCAC-FA carrier was significantly accelerated release in the presentation of glutathione or in cold shock condition, proposing the efficacy-controlled release. MTX@PCAC-FA showed excellent hemocompatibility, confirming a suitable application with parenteral administration. Notably, the acute and subacute toxicity in the mice model showed that the toxicity of MTX had significantly reduced after encapsulating in the PCAC-FA carrier. These nanoplatforms not only provide an alternative safe strategy for the clinical treatment of rheumatoid arthritis with MTX but also deliver MTX selectively and provide on-demand drug release via external and internal signals, thus emerging as a promising therapeutic option for precise RA therapy.
Collapse
Affiliation(s)
- Le Hang Dang
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, HCMC, Viet Nam; Institute of Applied Materials Science, Vietnam Academy of Science and Technology, HCMC, Viet Nam.
| | - Nhu Quynh Vu
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, HCMC, Viet Nam; School of Medicine -, Vietnam National University, Ho Chi Minh City, Viet Nam
| | - Thuy Tien Nguyen
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, HCMC, Viet Nam; School of Medicine -, Vietnam National University, Ho Chi Minh City, Viet Nam
| | - Thi Hong Tuoi Do
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Viet Nam
| | - Thi Kim Tram Pham
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Viet Nam
| | - Ngoc Quyen Tran
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, HCMC, Viet Nam; Institute of Applied Materials Science, Vietnam Academy of Science and Technology, HCMC, Viet Nam.
| |
Collapse
|
6
|
Shi Y, Xu S, Zhao J, Zhu H, Pan X, Zhao B, Sun Z, Li N, Hou X. Development of injectable in situ hydrogels based on hyaluronic acid via Diels-Alder reaction for their antitumor activities studies. Int J Biol Macromol 2024; 262:129642. [PMID: 38266838 DOI: 10.1016/j.ijbiomac.2024.129642] [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: 10/09/2023] [Revised: 01/09/2024] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
The objective of this study was to develop an injectable hydrogel based on furfuryl amine-conjugated hyaluronic acid (FA-conj-HA) and evaluate the in vivo anti-4 T1 tumor activity of doxorubicin-loaded hydrogel (DOX@FA-conj-HAgel). The cargo-free hydrogel (FA-conj-HAgel) was fabricated through a Diels-Alder reaction at 37 °C with FA-conj-HA as a gel material and four armed poly(ethylene glycol)2000-maleimide (4-arm-PEG2000-Mal) as a cross-linker. The bio-safety of FA-conj-HAgel were assessed, and the in vivo antitumor activity of DOX@FA-conj-HAgel was also investigated. Many 3D network structures were observed from scanning electron microscope (SEM) photograph, confirming the successful preparation of FA-conj-HAgel. The absence of cytotoxicity from FA-conj-HAgel was proved by the high viability of 4 T1 cells. In vivo bio-safety studies suggested that the obtained FA-conj-HAgel did not induce acute toxicity or other lesions in treated mice, confirming its high bio-safety. The reduced tumor volumes, hematoxylin-eosin staining (H&E), and TdT-mediated dUTP-biotin nick end labeling (TUNEL) analysis indicated the potent in vivo anti-4 T1 tumor effects of DOX@FA-conj-HAgel. In conclusion, the favorable bio-safety and potent antitumor activity of DOX@FA-conj-HAgel highlighted its potential application in oncological therapy.
Collapse
Affiliation(s)
- Yongli Shi
- College of pharmacy, Xinxiang Medical University, 453003 Xinxiang, PR China.
| | - Suyue Xu
- College of pharmacy, Xinxiang Medical University, 453003 Xinxiang, PR China
| | - Jingya Zhao
- College of pharmacy, Xinxiang Medical University, 453003 Xinxiang, PR China
| | - Huiqing Zhu
- College of pharmacy, Xinxiang Medical University, 453003 Xinxiang, PR China
| | - Xiaofei Pan
- College of pharmacy, Xinxiang Medical University, 453003 Xinxiang, PR China
| | - Bingqian Zhao
- Basic Medicine College, Xinxiang Medical University, 453003, Xinxiang, PR China
| | - Zeyu Sun
- First Clinical College, Xinxiang Medical University, 453003 Xinxiang, PR China
| | - Na Li
- College of pharmacy, Xinxiang Medical University, 453003 Xinxiang, PR China.
| | - Xueyan Hou
- College of pharmacy, Xinxiang Medical University, 453003 Xinxiang, PR China; Pingyuan Laboratory, Xinxiang, Henan 453007, PR 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
|
Bhoi A, Dwivedi SD, Singh D, Keshavkant S, Singh MR. Plant-Based Approaches for Rheumatoid Arthritis Regulation: Mechanistic Insights on Pathogenesis, Molecular Pathways, and Delivery Systems. Crit Rev Ther Drug Carrier Syst 2024; 41:39-86. [PMID: 38305341 DOI: 10.1615/critrevtherdrugcarriersyst.2023048324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Rheumatoid arthritis (RA) is classified as a chronic inflammatory autoimmune disorder, associated with a varied range of immunological changes, synovial hyperplasia, cartilage destructions, as well as bone erosion. The infiltration of immune-modulatory cells and excessive release of proinflammatory chemokines, cytokines, and growth factors into the inflamed regions are key molecules involved in the progression of RA. Even though many conventional drugs are suggested by a medical practitioner such as DMARDs, NSAIDs, glucocorticoids, etc., to treat RA, but have allied with various side effects. Thus, alternative therapeutics in the form of herbal therapy or phytomedicine has been increasingly explored for this inflammatory disorder of joints. Herbal interventions contribute substantial therapeutic benefits including accessibility, less or no toxicity and affordability. But the major challenge with these natural actives is the need of a tailored approach for treating inflamed tissues by delivering these bioactive agentsat an appropriate dose within the treatment regimen for an extended periodof time. Drug incorporated with wide range of delivery systems such as liposomes, nanoparticles, polymeric micelles, and other nano-vehicles have been developed to achieve this goal. Thus, inclinations of modern treatment are persuaded on the way to herbal therapy or phytomedicines in combination with novel carriers is an alternative approach with less adverse effects. The present review further summarizes the significanceof use of phytocompounds, their target molecules/pathways and, toxicity and challenges associated with phytomolecule-based nanoformulations.
Collapse
Affiliation(s)
- Anita Bhoi
- School of Studies in Biotechnology, Pt. Ravishankar Shukla University, Raipur 492 010, India
| | - Shradha Devi Dwivedi
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492 010, India
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh, 492010, India; National Centre for Natural Resources, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh, 492010, India
| | - S Keshavkant
- School of Studies in Biotechnology, Pt. Ravishankar Shukla University, Raipur 492 010, India
| | - Manju Rawat Singh
- University Institute of pharmacy, Pt.Ravishankar Shukla University, Raipur.(C.G.) 2. National centre for natural resources, Pt. Ravishankar Shukla University, Raipur
| |
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
|
Lin Q, Shan X, Li X, Luo Z, Yu X, Liu H, Wang S, Zhao X, Zhu Y, Zhou H, Luo L, You J. Solvent exchange-motivated and tunable in situ forming implants sustaining triamcinolone acetonide release for arthritis treatment. Int J Pharm 2023; 645:123383. [PMID: 37678476 DOI: 10.1016/j.ijpharm.2023.123383] [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: 05/12/2023] [Revised: 08/17/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Arthritis is a syndrome characterized by inflammation in the joints. Triamcinolone acetonide (TA) was used as an anti-inflammatory agent in the treatment of this disease. However, there are limitations to its clinical application, including rapid clearance from the joint cavity, potential joint damage from multiple injections, and adverse joint events. To address these drawbacks, we developed a tunable in situ forming implant loaded with TA. This injectable polymer solution utilized poly (lactic-co-glycolic acid) (PLGA) as an extended-release material. When injected into the joints, the solution solidifies into implants through a solvent exchange in the aqueous environment. The implants demonstrated robust retention at the injection site and released TA over several weeks even months through diffusion and erosion. By adding different proportions of low water-miscible plasticizers, the release period of the drug could be precisely adjusted. The plasticizers-optimized implants exhibited a tough texture, enhancing the therapeutic efficiency and drug safety in vivo. In arthritic model studies, the tunable TA-loaded implants significantly reduced swelling, pain, and motor discoordination, and also showed suppression of arthritis progression to some extent. These findings suggested that TA-loaded ISFI holds promise for managing inflammatory disorders in individuals with arthritis.
Collapse
Affiliation(s)
- Qing Lin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xinyu Shan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xiang Li
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Zhenyu Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xin Yu
- College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, No. 26 Huatuo Dajie, Benxi, Liaoning 117004, China
| | - Huihui Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xiaoqi Zhao
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Ying Zhu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Huanli Zhou
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China; Jinhua Institute of Zhejiang University, 321299 Jinhua, China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China; Jinhua Institute of Zhejiang University, 321299 Jinhua, China; Zhejiang-California International Nanosystems Institute, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
11
|
Luo X, Xiong H, Jiang Y, Fan Y, Zuo C, Chen D, Chen L, Lin H, Gao J. Macrophage Reprogramming via Targeted ROS Scavenging and COX-2 Downregulation for Alleviating Inflammation. Bioconjug Chem 2023. [PMID: 37330989 DOI: 10.1021/acs.bioconjchem.3c00239] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Inflammation-related diseases affect large populations of people in the world and cause substantial healthcare burdens, which results in significant costs in time, material, and labor. Preventing or relieving uncontrolled inflammation is critical for the treatment of these diseases. Herein, we report a new strategy for alleviating inflammation by macrophage reprogramming via targeted reactive oxygen species (ROS) scavenging and cyclooxygenase-2 (COX-2) downregulation. As a proof of concept, we synthesize a multifunctional compound named MCI containing a mannose-based macrophage targeting moiety, an indomethacin (IMC)-based segment for inhibiting COX-2, and a caffeic acid (CAF)-based section for ROS clearance. As revealed by a series of in vitro experiments, MCI could significantly attenuate the expression of COX-2 and the level of ROS, leading to M1 to M2 macrophage reprogramming, as evidenced by the reduction and the elevation in the levels of pro-inflammatory M1 markers and anti-inflammatory M2 markers, respectively. Furthermore, in vivo experiments show MCI's promising therapeutic effects on rheumatoid arthritis (RA). Our work illustrates the success of targeted macrophage reprogramming for inflammation alleviation, which sheds light on the development of new anti-inflammatory drugs.
Collapse
Affiliation(s)
- Xiangjie Luo
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Hui Xiong
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yuhang Jiang
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yifan Fan
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Cuicui Zuo
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Dongxia Chen
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Limin Chen
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Hongyu Lin
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jinhao Gao
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
12
|
Zhang R, Liu F, Zhang Q, Yang L, Hou X, Du T, Fan J, Hu H, Deng H, Hao L, Guo L, Fu C. Intra-articular delivery system of methotrexate for rheumatoid arthritis therapy: An in-suit thermosensitive comprehensive gel of polysaccharide from Aconitum carmichaelii Debx. Int J Biol Macromol 2023:124822. [PMID: 37257527 DOI: 10.1016/j.ijbiomac.2023.124822] [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: 11/16/2022] [Revised: 04/18/2023] [Accepted: 05/07/2023] [Indexed: 06/02/2023]
Abstract
The polysaccharides (FP) extracted from the lateral roots of Aconitum carmichaelii Debx. (Fuzi) are natural compounds, which have effective therapy for rheumatoid arthritis (RA). Methotrexate (MTX) is the first-line drug for RA, but its application is greatly limited to the toxicity in liver and kidney and drug resistance. In this study, an attempt is made to apply oxidized FP (OFP) as a polymer carrier based on intra-articular delivery system loaded MTX. The FP could be developed and used as comprehensive gel carriers with biocompatibility and degradability for therapy of RA. Firstly, OFP-chitosan-poloxamer 407 (OFP-CS-F407-MTX) gel was prepared by natural non-toxic cross-linking agents. Physicochemical characterization was performed by using 1H NMR and FTIR spectroscopic techniques to assess the successful functionalization of OFP. TGA, SEM and rheological experiment of OFP-CS-F407-MTX gel were investigated. Notably, we loaded MTX into OFP-CS-F407-MTX gel which had remarkable therapeutic efficacy and biosafety for RA. Based on advantages of intra-articular injection of OFP-CS-F407-MTX gel releasing MTX, it modulated proinflammatory cytokines by down-regulating TNF-α, IL-6 and IL-1β expression. Therefore, OFP-CS-F407-MTX in situ gel delivery system can potentially reduce systemic toxicity and irritation of oral administration of MTX but give a controlled release of drug for a long period of time.
Collapse
Affiliation(s)
- Ruiyuan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
| | - Fang Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China.
| | - Qian Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
| | - Luping Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
| | - Xinlian Hou
- Hua Run Sanjiu (Ya'an) Pharmaceutical Co., Ltd., Ya'an 625000, Sichuan Province, China
| | - TaoMing Du
- Department of Radiology, Chengdu Seventh People's Hospital, Chengdu 610000, Sichuan Province, China
| | - Jie Fan
- Department of Radiology, Chengdu Seventh People's Hospital, Chengdu 610000, Sichuan Province, China
| | - Huiling Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
| | - Hongdan Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
| | - Li Hao
- Hua Run Sanjiu (Ya'an) Pharmaceutical Co., Ltd., Ya'an 625000, Sichuan Province, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China.
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province, China
| |
Collapse
|
13
|
Shen Q, Du Y. A comprehensive review of advanced drug delivery systems for the treatment of rheumatoid arthritis. Int J Pharm 2023; 635:122698. [PMID: 36754181 DOI: 10.1016/j.ijpharm.2023.122698] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 01/21/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023]
Abstract
Rheumatoid arthritis (RA), a chronic autoimmune disease, is characterized by articular pain and swelling, synovial hyperplasia, and cartilage and bone destruction. Conventional treatment strategies for RA involve the use of anti-rheumatic drugs, which warrant high-dose, frequent, and long-term administration, resulting in serious adverse effects and poor patient compliance. To overcome these problems and improve clinical efficacy, drug delivery systems (DDS) have been designed for RA treatment. These systems have shown success in animal models of RA. In this review, representative DDS that target RA through passive or active effects on inflammatory cells are discussed and highlighted using examples. In particular, DDS allowing controlled and targeted drug release based on a variety of stimuli, intra-articular DDS, and transdermal DDS for RA treatment are described. Thus, this review provides an improved understanding of these DDS and paves the way for the development of novel DDS for efficient RA treatment.
Collapse
Affiliation(s)
- Qiying Shen
- School of Pharmacy, Hangzhou Normal University, 2318 Yu-HangTang Road, Hangzhou 311121, China; Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-HangTang Road, Hangzhou 310058, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-HangTang Road, Hangzhou 310058, China.
| |
Collapse
|
14
|
Han Y, Huang S. Nanomedicine is more than a supporting role in rheumatoid arthritis therapy. J Control Release 2023; 356:142-161. [PMID: 36863691 DOI: 10.1016/j.jconrel.2023.02.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023]
Abstract
Rheumatoid arthritis(RA) is an autoimmune disorder that affects the joints. Various medications successfully alleviate the symptoms of RA in clinical. Still, few therapy strategies can cure RA, especially when joint destruction begins, and there is currently no effective bone-protective treatment to reverse the articular damage. Furthermore, the RA medications now used in clinical practice accompany various adverse side effects. Nanotechnology can improve the pharmacokinetics of traditional anti-RA drugs and therapeutic precision through targeting modification. Although the clinical application of nanomedicines for RA is in its infancy, preclinical research is rising. Current anti-RA nano-drug studies mainly focus on the following: drug delivery systems, nanomedicines with anti-inflammatory and anti-arthritic properties, biomimetic design with better biocompatibility and therapeutic features, and nanoparticle-dominated energy conversion therapies. These therapies have shown promising therapeutic benefits in animal models, indicating that nanomedicines are a potential solution to the current bottleneck in RA treatment. This review will summarize the present state of anti-RA nano-drug research.
Collapse
Affiliation(s)
- Yu Han
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Shilei Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
15
|
Logesh K, Raj B, Bhaskaran M, Thirumaleshwar S, Gangadharappa H, Osmani R, Asha Spandana K. Nanoparticulate drug delivery systems for the treatment of rheumatoid arthritis: A comprehensive review. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
16
|
Haloi P, Chawla S, Konkimalla VB. Thermosensitive smart hydrogel of PEITC ameliorates the therapeutic efficacy in rheumatoid arthritis. Eur J Pharm Sci 2023; 181:106367. [PMID: 36572358 DOI: 10.1016/j.ejps.2022.106367] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune condition that accompanies chronic inflammation of joints with limited therapeutic options. Phenethyl isothiocyanate (PEITC), a bioactive phytochemical, exerts its chemopreventive, anti-oxidant, and anti-inflammatory activity via the Nrf-2 pathway. However, limited water solubility, short half-life, and instability are reasons for the low bioavailability of PEITC that hampers clinical application. From studies in healthy rats, the performance of PEITC-loaded chitosan/pluronic F-127 smart hydrogel (PH) as a thermosensitive injectable demonstrated adequate thermosensitivity (gel formation), injectability (ease of administration), biocompatibility (with prolonged contact), pharmacokinetics (sustained drug release), and biosafety (nontoxic to major organs). In the adjuvant-induced arthritis (AIA) rat model, PEITC-hydrogel (PH50) injected into the knee joint lowered RA-related symptoms significantly (paw edema and arthritis score). Further, a marked reduction in bone erosion and inflammation-specific biomarkers was observed. Finally, this study demonstrates a smart injectable hydrogel optimally loaded with PEITC which is safe, biocompatible and exhibits significant therapeutic efficacy in RA conditions.
Collapse
Affiliation(s)
- Prakash Haloi
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Jatni, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Saurabh Chawla
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Jatni, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - V Badireenath Konkimalla
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Jatni, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India.
| |
Collapse
|
17
|
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: 49] [Impact Index Per Article: 24.5] [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
|
18
|
Zheng K, Bai J, Yang H, Xu Y, Pan G, Wang H, Geng D. Nanomaterial-assisted theranosis of bone diseases. Bioact Mater 2022; 24:263-312. [PMID: 36632509 PMCID: PMC9813540 DOI: 10.1016/j.bioactmat.2022.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/27/2022] Open
Abstract
Bone-related diseases refer to a group of skeletal disorders that are characterized by bone and cartilage destruction. Conventional approaches can regulate bone homeostasis to a certain extent. However, these therapies are still associated with some undesirable problems. Fortunately, recent advances in nanomaterials have provided unprecedented opportunities for diagnosis and therapy of bone-related diseases. This review provides a comprehensive and up-to-date overview of current advanced theranostic nanomaterials in bone-related diseases. First, the potential utility of nanomaterials for biological imaging and biomarker detection is illustrated. Second, nanomaterials serve as therapeutic delivery platforms with special functions for bone homeostasis regulation and cellular modulation are highlighted. Finally, perspectives in this field are offered, including current key bottlenecks and future directions, which may be helpful for exploiting nanomaterials with novel properties and unique functions. This review will provide scientific guidance to enhance the development of advanced nanomaterials for the diagnosis and therapy of bone-related diseases.
Collapse
Affiliation(s)
- Kai Zheng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China,Corresponding author.Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Huaiyu Wang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China,Corresponding author.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China,Corresponding author. Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
19
|
Koland M, Narayanan Vadakkepushpakath A, John A, Tharamelveliyil Rajendran A, Raghunath I. Thermosensitive In Situ Gels for Joint Disorders: Pharmaceutical Considerations in Intra-Articular Delivery. Gels 2022; 8:723. [PMID: 36354630 PMCID: PMC9689403 DOI: 10.3390/gels8110723] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 09/17/2023] Open
Abstract
The intra-articular administration of conventional drug solutions or dispersions in joint diseases such as osteoarthritis has a relatively short retention time and, therefore, limited therapeutic effect. Thermosensitive polymer solutions that exhibit a sol-gel phase transition near body temperature after injection can prolong drug retention by providing a depot from which the drug release is sustained while relieving inflammation and preventing degradation of the joint complex. Thermosensitive hydrogels have in recent times garnered considerable attention in the intra-articular therapeutics of joint diseases such as osteoarthritis. Among the stimuli-responsive gelling systems, most research has focused on thermosensitive hydrogels. These gels are preferred over other stimuli-sensitive hydrogels since they have well-controlled in situ gelling properties and are also easier to load with drugs. Temperature-sensitive polymers, such as block copolymers or poloxamers, are frequently used to modify their gelation properties, usually in combination with other polymers. They are compatible with most drugs but may pose formulation challenges in terms of their low-response time, highly fragile nature, and low biocompatibility. The stability and biodegradability of implant hydrogels can control the drug release rate and treatment efficacy. This review stresses the application of thermosensitive gels in joint disorders and summarizes recent developments for intra-articular application, including the incorporation of nanoparticles. The hydrogel composition, drug release mechanisms, and the challenges involved in their formulation and storage are also discussed.
Collapse
Affiliation(s)
- Marina Koland
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Mangalore 575018, India
| | | | | | | | | |
Collapse
|
20
|
Pu Q, Wang K, Peng B, Chen K, Gong T, Liu F, Yang Q. In situ Preparation of a Phospholipid Gel Co-Loaded with Methotrexate and Dexamethasone for Synergistic Rheumatoid Arthritis Treatment. Int J Nanomedicine 2022; 17:5153-5162. [DOI: 10.2147/ijn.s384772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
|
21
|
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
|
22
|
Advances in nanoenabled 3D matrices for cartilage repair. Acta Biomater 2022; 150:1-21. [PMID: 35902038 DOI: 10.1016/j.actbio.2022.07.033] [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: 03/03/2022] [Revised: 07/09/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022]
Abstract
Cartilage repair strategies are evolving at a fast pace with technology development. Matrices that offer multifaceted functions and a full adaption to the cartilage defect are of pivotal interest. Current cartilage repair strategies face numerous challenges, mostly related to the development of highly biomimetic materials, non-invasive injectable solutions, and adequate degradation rates. These strategies often fail due to feeble mechanical properties, the inability to sustain cell adhesion, growth, and differentiation or by underestimating other players of cartilage degeneration, such as the installed pro-inflammatory microenvironment. The integration of nanomaterials (NMs) into 3D scaffolds, hydrogels and bioinks hold great potential in the improvement of key features of materials that are currently applied in cartilage tissue engineering strategies. NMs offer a high surface to volume ratio and their multiple applications can be explored to enhance cartilage mechanical properties, biocompatibility, cell differentiation, inflammation modulation, infection prevention and even to function as diagnostic tools or as stimuli-responsive cues in these 3D structures. In this review, we have critically reviewed the latest advances in the development of nanoenabled 3D matrices - enhanced by means of NMs - in the context of cartilage regeneration. We have provided a wide perspective of the synergistic effect of combining 3D strategies with NMs, with emphasis on the benefits brought by NMs in achieving functional and enhanced therapeutic outcomes. STATEMENT OF SIGNIFICANCE: Cartilage is one of the most challenging tissues to treat owing to its limited self-regeneration potential. Novel strategies using nanoenabled 3D matrices have emerged from the need to design more efficient solutions for cartilage repair, that take into consideration its unique mechanical properties and can direct specific cell behaviours. Here we aim to provide a comprehensive review on the synergistic effects of 3D matrices nanoenrichment in the context of cartilage regeneration, with emphasis on the heightening brought by nanomaterials in achieving functional and enhanced therapeutic outcomes. We anticipate this review to provide a wide perspective on the past years' research on the field, demonstrating the great potential of these approaches in the treatment and diagnosis of cartilage-related disorders.
Collapse
|
23
|
Campa-Carranza JN, Paez-Mayorga J, Chua CYX, Nichols JE, Grattoni A. Emerging local immunomodulatory strategies to circumvent systemic immunosuppression in cell transplantation. Expert Opin Drug Deliv 2022; 19:595-610. [PMID: 35588058 DOI: 10.1080/17425247.2022.2076834] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Cell transplantation is a promising curative therapeutic strategy whereby impaired organ functions can be restored without the need for whole organ transplantation. A key challenge in allotransplantation is the requirement for life-long systemic immunosuppression to prevent rejection, which is associated with serious adverse effects such as increased risk of opportunistic infections and the development of neoplasms. This challenge underscores the urgent need for novel strategies to prevent graft rejection while abrogating toxicity-associated adverse events. AREAS COVERED We review recent advances in immunoengineering strategies for localized immunomodulation that aim to support allograft function and provide immune tolerance in a safe and effective manner. EXPERT OPINION Immunoengineering strategies are tailored approaches for achieving immunomodulation of the transplant microenvironment. Biomaterials can be adapted for localized and controlled release of immunomodulatory agents, decreasing the effective dose threshold and frequency of administration. The future of transplant rejection management lies in the shift from systemic to local immunomodulation with suppression of effector and activation of regulatory T cells, to promote immune tolerance.
Collapse
Affiliation(s)
- Jocelyn Nikita Campa-Carranza
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.,School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, NL, Mexico
| | - Jesus Paez-Mayorga
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.,School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, NL, Mexico
| | - Corrine Ying Xuan Chua
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Joan E Nichols
- Center for Tissue Engineering, Houston Methodist Research Institute, Houston, TX, USA.,Department of Surgery, Houston Methodist Hospital, Houston, TX, USA
| | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.,Department of Surgery, Houston Methodist Hospital, Houston, TX, USA.,Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
24
|
Yue S, Bo L. Formulation and Development of Prednisolone Loaded Nanoparticulate Injection for the Treatment of Arthritis. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.611.617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
25
|
Zhao T, Wei Z, Zhu W, Weng X. Recent Developments and Current Applications of Hydrogels in Osteoarthritis. Bioengineering (Basel) 2022; 9:132. [PMID: 35447692 PMCID: PMC9024926 DOI: 10.3390/bioengineering9040132] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/13/2022] [Accepted: 03/21/2022] [Indexed: 01/02/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that causes disability if left untreated. The treatment of OA currently requires a proper delivery system that avoids the loss of therapeutic ingredients. Hydrogels are widely used in tissue engineering as a platform for carrying drugs and stem cells, and the anatomical environment of the limited joint cavity is suitable for hydrogel therapy. This review begins with a brief introduction to OA and hydrogels and illustrates the effects, including the analgesic effects, of hydrogel viscosupplementation on OA. Then, considering recent studies of hydrogels and OA, three main aspects, including drug delivery systems, mesenchymal stem cell entrapment, and cartilage regeneration, are described. Hydrogel delivery improves drug retention in the joint cavity, making it possible to deliver some drugs that are not suitable for traditional injection; hydrogels with characteristics similar to those of the extracellular matrix facilitate cell loading, proliferation, and migration; hydrogels can promote bone regeneration, depending on their own biochemical properties or on loaded proregenerative factors. These applications are interlinked and are often researched together.
Collapse
Affiliation(s)
- Tianhao Zhao
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (T.Z.); (Z.W.); (W.Z.)
| | - Zhanqi Wei
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (T.Z.); (Z.W.); (W.Z.)
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Wei Zhu
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (T.Z.); (Z.W.); (W.Z.)
| | - Xisheng Weng
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (T.Z.); (Z.W.); (W.Z.)
- Department of State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
26
|
Drug Delivery Strategies and Biomedical Significance of Hydrogels: Translational Considerations. Pharmaceutics 2022; 14:pharmaceutics14030574. [PMID: 35335950 PMCID: PMC8950534 DOI: 10.3390/pharmaceutics14030574] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 02/04/2023] Open
Abstract
Hydrogels are a promising and attractive option as polymeric gel networks, which have immensely fascinated researchers across the globe because of their outstanding characteristics such as elevated swellability, the permeability of oxygen at a high rate, good biocompatibility, easy loading, and drug release. Hydrogels have been extensively used for several purposes in the biomedical sector using versatile polymers of synthetic and natural origin. This review focuses on functional polymeric materials for the fabrication of hydrogels, evaluation of different parameters of biocompatibility and stability, and their application as carriers for drugs delivery, tissue engineering and other therapeutic purposes. The outcome of various studies on the use of hydrogels in different segments and how they have been appropriately altered in numerous ways to attain the desired targeted delivery of therapeutic agents is summarized. Patents and clinical trials conducted on hydrogel-based products, along with scale-up translation, are also mentioned in detail. Finally, the potential of the hydrogel in the biomedical sector is discussed, along with its further possibilities for improvement for the development of sophisticated smart hydrogels with pivotal biomedical functions.
Collapse
|
27
|
Bentley ER, Little SR. Local delivery strategies to restore immune homeostasis in the context of inflammation. Adv Drug Deliv Rev 2021; 178:113971. [PMID: 34530013 PMCID: PMC8556365 DOI: 10.1016/j.addr.2021.113971] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/05/2021] [Accepted: 09/07/2021] [Indexed: 12/13/2022]
Abstract
Immune homeostasis is maintained by a precise balance between effector immune cells and regulatory immune cells. Chronic deviations from immune homeostasis, driven by a greater ratio of effector to regulatory cues, can promote the development and propagation of inflammatory diseases/conditions (i.e., autoimmune diseases, transplant rejection, etc.). Current methods to treat chronic inflammation rely upon systemic administration of non-specific small molecules, resulting in broad immunosuppression with unwanted side effects. Consequently, recent studies have developed more localized and specific immunomodulatory approaches to treat inflammation through the use of local biomaterial-based delivery systems. In particular, this review focuses on (1) local biomaterial-based delivery systems, (2) common materials used for polymeric-delivery systems and (3) emerging immunomodulatory trends used to treat inflammation with increased specificity.
Collapse
Affiliation(s)
- Elizabeth R Bentley
- Department of Bioengineering, University of Pittsburgh, 302 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15260, United States.
| | - Steven R Little
- Department of Bioengineering, University of Pittsburgh, 302 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15260, United States; Department of Chemical Engineering, University of Pittsburgh, 940 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15213, United States; Department of Clinical and Translational Science, University of Pittsburgh, Forbes Tower, Suite 7057, Pittsburgh, PA 15213, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA 15219, United States; Department of Immunology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, United States; Department of Pharmaceutical Sciences, University of Pittsburgh, 3501 Terrace Street, Pittsburgh, PA 15213, United States; Department of Ophthalmology, University of Pittsburgh, 203 Lothrop Street, Pittsburgh, PA 15213, United States.
| |
Collapse
|
28
|
Han X, Wu Y, Shan Y, Zhang X, Liao J. Effect of Micro-/Nanoparticle Hybrid Hydrogel Platform on the Treatment of Articular Cartilage-Related Diseases. Gels 2021; 7:gels7040155. [PMID: 34698122 PMCID: PMC8544595 DOI: 10.3390/gels7040155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/18/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023] Open
Abstract
Joint diseases that mainly lead to articular cartilage injury with prolonged severe pain as well as dysfunction have remained unexplained for many years. One of the main reasons is that damaged articular cartilage is unable to repair and regenerate by itself. Furthermore, current therapy, including drug therapy and operative treatment, cannot solve the problem. Fortunately, the micro-/nanoparticle hybrid hydrogel platform provides a new strategy for the treatment of articular cartilage-related diseases, owing to its outstanding biocompatibility, high loading capability, and controlled release effect. The hybrid platform is effective for controlling symptoms of pain, inflammation and dysfunction, and cartilage repair and regeneration. In this review, we attempt to summarize recent studies on the latest development of micro-/nanoparticle hybrid hydrogel for the treatment of articular cartilage-related diseases. Furthermore, some prospects are proposed, aiming to improve the properties of the micro-/nanoparticle hybrid hydrogel platform so as to offer useful new ideas for the effective and accurate treatment of articular cartilage-related diseases.
Collapse
|
29
|
Development of a Polysaccharide-Based Hydrogel Drug Delivery System (DDS): An Update. Gels 2021; 7:gels7040153. [PMID: 34698125 PMCID: PMC8544468 DOI: 10.3390/gels7040153] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/02/2021] [Accepted: 09/14/2021] [Indexed: 12/28/2022] Open
Abstract
Delivering a drug to the target site with minimal-to-no off-target cytotoxicity is the major determinant for the success of disease therapy. While the therapeutic efficacy and cytotoxicity of the drug play the main roles, the use of a suitable drug delivery system (DDS) is important to protect the drug along the administration route and release it at the desired target site. Polysaccharides have been extensively studied as a biomaterial for DDS development due to their high biocompatibility. More usefully, polysaccharides can be crosslinked with various molecules such as micro/nanoparticles and hydrogels to form a modified DDS. According to IUPAC, hydrogel is defined as the structure and processing of sols, gels, networks and inorganic–organic hybrids. This 3D network which often consists of a hydrophilic polymer can drastically improve the physical and chemical properties of DDS to increase the biodegradability and bioavailability of the carrier drugs. The advancement of nanotechnology also allows the construction of hydrogel DDS with enhanced functionalities such as stimuli-responsiveness, target specificity, sustained drug release, and therapeutic efficacy. This review provides a current update on the use of hydrogel DDS derived from polysaccharide-based materials in delivering various therapeutic molecules and drugs. We also highlighted the factors that affect the efficacy of these DDS and the current challenges of developing them for clinical use.
Collapse
|
30
|
Anita C, Munira M, Mural Q, Shaily L. Topical nanocarriers for management of Rheumatoid Arthritis: A review. Biomed Pharmacother 2021; 141:111880. [PMID: 34328101 DOI: 10.1016/j.biopha.2021.111880] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/20/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease manifested by chronic joint inflammation leading to severe disability and premature mortality. With a global prevalence of about 0.3%-1% RA is 3-5 times more prevalent in women than in men. There is no known cure for RA; the ultimate goal for treatment of RA is to provide symptomatic relief. The treatment regimen for RA involves frequent drug administration and high doses of NSAIDs such as indomethacin, diclofenac, ibuprofen, celecoxib, etorcoxib. These potent drugs often have off target effects which drastically decreases patient compliance. Moreover, conventional non-steroidal anti-inflammatory have many formulation challenges like low solubility and permeability, poor bioavailability, degradation by gastrointestinal enzymes, food interactions and toxicity. To overcome these barriers, researchers have turned to topical route of drug administration, which has superior patience compliance and they also bypass the first past effect experienced with conventional oral administration. Furthermore, to enhance the permeation of drug through the layers of the skin and reach the site of inflammation, nanosized carriers have been designed such as liposomes, nanoemulsions, niosomes, ethosomes, solid lipid nanoparticles and transferosomes. These drug delivery systems are non-toxic and have high drug encapsulation efficiency and they also provide sustained release of drug. This review discusses the effect of formulation composition on the physiochemical properties of these nanocarriers in terms of particle size, surface charge, drug entrapment and also drug release profile thus providing a landscape of topically used nanoformulations for symptomatic treatment of RA.
Collapse
Affiliation(s)
- Chando Anita
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai 400056, India
| | - Momin Munira
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai 400056, India; Shri C. B. Patel Research Centre, Vile Parle (West), Mumbai 400056, India.
| | - Quadros Mural
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai 400056, India
| | - Lalka Shaily
- Department of Regulatory Affairs, Rusan Pharma Limited, Charkop, Kandivali (West), Mumbai 400067, India
| |
Collapse
|
31
|
Wang L, Che K, Liu Y. Pharmacokinetics, distribution and efficacy of triptolide PLGA microspheres after intra-articular injection in a rat rheumatoid arthritis model. Xenobiotica 2021; 51:703-715. [PMID: 33938387 DOI: 10.1080/00498254.2021.1923860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The UPLC-MS/MS method was established with good precision, accuracy and stability to determine the concentrations of TPL in biological samples, such as heart, liver, spleen, lung, kidney, plasma and joint.After being made into microspheres, TPL can stay in the joint tissue for a long time, further reducing the number of times joint cavity administration, and its sustained release effect was significantly improved compared with the solution dosage form.The pharmacokinetic parameters, such as AUC(0-t), AUC(0-∞), T1/2, Tmax, MTR(0-t), and MTR(0-∞) of the TPL-PLGA-MS group were significantly increased compared with those of the solution group. The microsphere preparation could significantly slow the release rate of the drug from the joint cavity.TPL-PLGA-MS can significantly reduce the expression of inflammatory factors such as IL-1, IL-6, TNF-α and hs-CRP. TPL-PLGA-MS for articular cavity injection has potential as a new preparation for the treatment of RA.
Collapse
Affiliation(s)
- Lijuan Wang
- Pharmacy College, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Keke Che
- Department of Pharmacy, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Yan Liu
- Pharmacy College, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, China
| |
Collapse
|
32
|
Zewail M, Nafee N, Boraie N. Intra-Articular Dual Drug Delivery for Synergistic Rheumatoid Arthritis Treatment. J Pharm Sci 2021; 110:2808-2822. [PMID: 33848528 DOI: 10.1016/j.xphs.2021.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 11/16/2022]
Abstract
Systemic rheumatoid arthritis (RA) regimens fail to attain effective drug level at the affected joints and are associated with serious side effects. Herein, an attempt made to improve therapeutic outcomes of both leflunomide (LEF) which is a disease modifying antirheumatic and dexamethasone (Dex) through local delivery of combination therapy by intra-articular route. LEF and Dex were encapsulated in nanostructured lipid carriers (NLCs) and PLGA nanoparticles (NPs), respectively. Both nanocarriers were loaded into chitosan/β glycerophosphate (CS/βGP) thermo-sensitive hydrogels and injected intra-articularly in adjuvant induced RA rat model. Particle size of LEF NLCs and selected Dex NPs formulations were 200 and 119 nm, respectively. Dex NPs and LEF NLCs showed a sustained release profile for up to 58 and 17 days, respectively. After 14 days of treatment remarkable joint healing was observed for groups treated with Dex NPs in combination with either free LEF or LEF NLCs in CS/βGP hydrogel. Joint diameter measurements, TNF α levels and histopathological examination of dissected joints showed comparable values to the negative control group. This might be attributed to the synergistic effect of drug combination besides the ability of nanocarriers loaded hydrogel to prolong joint residence time and enhance joint healing potential.
Collapse
Affiliation(s)
- Mariam Zewail
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, El Gomhoria Street, Damanhour, Egypt.
| | - Noha Nafee
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Kuwait University, Kuwait
| | - Nabila Boraie
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
33
|
Deng Z, Liu S. Inflammation-responsive delivery systems for the treatment of chronic inflammatory diseases. Drug Deliv Transl Res 2021; 11:1475-1497. [PMID: 33860447 PMCID: PMC8048351 DOI: 10.1007/s13346-021-00977-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2021] [Indexed: 12/30/2022]
Abstract
Inflammation is the biological response of immune system to protect living organisms from injurious factors. However, excessive and uncontrolled inflammation is implicated in a variety of devastating chronic diseases including atherosclerosis, inflammatory bowel disease (IBD), and rheumatoid arthritis (RA). Improved understanding of inflammatory response has unveiled a rich assortment of anti-inflammatory therapeutics for the treatment and management of relevant chronic diseases. Notwithstanding these successes, clinical outcomes are variable among patients and serious adverse effects are often observed. Moreover, there exist some limitations for clinical anti-inflammatory therapeutics such as aqueous insolubility, low bioavailability, off-target effects, and poor accessibility to subcellular compartments. To address these challenges, the rational design of inflammation-specific drug delivery systems (DDSs) holds significant promise. Moreover, as compared to normal tissues, inflamed tissue-associated pathological milieu (e.g., oxidative stress, acidic pH, and overexpressed enzymes) provides vital biochemical stimuli for triggered delivery of anti-inflammatory agents in a spatiotemporally controlled manner. In this review, we summarize recent advances in the development of anti-inflammatory DDSs with built-in pathological inflammation-specific responsiveness for the treatment of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Zhengyu Deng
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Sciences At the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, Anhui Province, China
| | - Shiyong Liu
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Sciences At the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, Anhui Province, China.
| |
Collapse
|
34
|
Yan T, Ma Z, Liu J, Yin N, Lei S, Zhang X, Li X, Zhang Y, Kong J. Thermoresponsive GenisteinNLC-dexamethasone-moxifloxacin multi drug delivery system in lens capsule bag to prevent complications after cataract surgery. Sci Rep 2021; 11:181. [PMID: 33420301 PMCID: PMC7794611 DOI: 10.1038/s41598-020-80476-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 12/11/2020] [Indexed: 01/29/2023] Open
Abstract
Cataract surgery is the most common intraocular procedure. To decrease postsurgical inflammation, prevent infection and reduce the incidence of secondary cataract, we built a temperature-sensitive drug delivery system carrying dexamethasone, moxifloxacin and genistein nanostructured lipid carrier (GenNLC) modified by mPEG-PLA based on F127/F68 as hydrogel. Characterizations and release profiles of the drug delivery system were studied. In vitro functions were detected by CCK-8 test, immunofluorescence, wound-healing assay, real time-PCR and western blotting. The size of GenNLCs was 39.47 ± 0.69 nm in average with surface charges of - 4.32 ± 0.84 mV. The hydrogel gelation temperature and time were 32 °C, 20 s with a viscosity, hardness, adhesiveness and stringiness of 6.135 Pa.s, 54.0 g, 22.0 g, and 3.24 mm, respectively. Transmittance of the gel-release medium was above 90% (93.44 ± 0.33% to 100%) at range of 430 nm to 800 nm. Moxifloxacin released completely within 10 days. Fifty percent of dexamethasone released at a constant rate in the first week, and then released sustainably with a tapering down rate until day 30. Genistein released slowly but persistently with a cumulative release of 63% at day 40. The thermoresponsive hydrogel inhibited the proliferation, migration and epithelial-mesenchymal transition of SRA 01/04 cells, which were confirmed by testing CCK-8, wound-healing assay, western blot, real time-PCR (RT-PCR) and immunofluorescence. These results support this intracameral thermoresponsive in situ multi-drug delivery system with programmed release amounts and release profiles to cut down the need of eye drops for preventing inflammation or infection and to reduce posterior capsular opacification following cataract surgery.
Collapse
Affiliation(s)
- Tingyu Yan
- grid.412644.1Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, No.11 Xinhua Road, Heping District, Shenyang, 110005 Liaoning Province China
| | - Zhongxu Ma
- grid.265021.20000 0000 9792 1228Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Vision Science, Clinical College of Ophthalmology, Tianjin Medical University, No. 4 Gansu Rd, Heping District, Tianjin, 300020 China
| | - Jingjing Liu
- grid.412644.1Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, No.11 Xinhua Road, Heping District, Shenyang, 110005 Liaoning Province China
| | - Na Yin
- grid.412561.50000 0000 8645 4345Department of Pharmaceutics, Shenyang Pharmaceutical University, No.103 Wen Hua Road, Shenyang, 110016 China
| | - Shizhen Lei
- grid.412644.1Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, No.11 Xinhua Road, Heping District, Shenyang, 110005 Liaoning Province China
| | - Xinxin Zhang
- grid.412644.1Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, No.11 Xinhua Road, Heping District, Shenyang, 110005 Liaoning Province China
| | - Xuedong Li
- grid.412644.1Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, No.11 Xinhua Road, Heping District, Shenyang, 110005 Liaoning Province China
| | - Yu Zhang
- grid.412561.50000 0000 8645 4345Department of Pharmaceutics, Shenyang Pharmaceutical University, No.103 Wen Hua Road, Shenyang, 110016 China
| | - Jun Kong
- grid.412644.1Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, No.11 Xinhua Road, Heping District, Shenyang, 110005 Liaoning Province China
| |
Collapse
|
35
|
Combinatory Effects of Bone Marrow-Derived Mesenchymal Stem Cells and Indomethacin on Adjuvant-Induced Arthritis in Wistar Rats: Roles of IL-1 β, IL-4, Nrf-2, and Oxidative Stress. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8899143. [PMID: 33488761 PMCID: PMC7803402 DOI: 10.1155/2021/8899143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/25/2020] [Accepted: 12/19/2020] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis (RA) is a disorder triggered by autoimmune reactions and related with chronic inflammation and severe disability. Bone Marrow-derived Mesenchymal Stem Cells (BM-MSCs) have shown a hopeful immunomodulatory effect towards repairing cartilage and restoring joint function. Additionally, indomethacin (IMC), a nonsteroidal compound, has been considered as a potent therapeutic agent that exhibits significant antipyretic properties and analgesic effects. The target of the current research is to assess the antiarthritic efficacy of BM-MSCs (106 cells/rat at 1, 6, 12 and 18 days) and IMC (2 mg/kg body weight/day for 3 weeks) either alone or concurrently administered against complete Freund's adjuvant-induced arthritic rats. Changes in paw volume, body weight, gross lesions, and antioxidant defense system, as well as oxidative stress, were assessed. The Th1 cytokine (IL-1β) serum level and Th2 cytokine (IL-4) and Nrf-2 ankle joint expression were detected. In comparison to normal rats, it was found that the CFA-induced arthritic rats exhibited significant leukocytosis and increase in paw volume, LPO level, RF, and IL-1β serum levels. In parallel, arthritic rats that received BM-MSCs and/or IMC efficiently exhibited decrease in paw edema, leukocytosis, and enhancement in the antioxidant enzymatic levels of SOD, GPx, GST, and GSH in serum besides upregulation of Nrf-2 and anti-inflammatory IL-4 expression levels in the ankle articular joint. Likewise, these analyses were more evidenced by the histopathological sections and histological score. The data also revealed that the combined administration of BM-MSC and IMC was more potent in suppressing inflammation and enhancing the anti-inflammatory pathway than each agent alone. Thus, it can be concluded that the combined therapy with BM-MSC and IMC may be used as a promising therapeutic choice after assessing their efficacy and safety in human beings with RA, and the antiarthritic effects may be mediated via modulatory effects on Th1/Th2 cytokines, ozidative stress, and Nrf-2.
Collapse
|
36
|
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
|
37
|
Water/pH dual responsive in situ calcium supplement collaborates simvastatin for osteoblast promotion mediated osteoporosis therapy via oral medication. J Control Release 2020; 329:121-135. [PMID: 33279604 DOI: 10.1016/j.jconrel.2020.11.059] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/22/2020] [Accepted: 11/29/2020] [Indexed: 12/20/2022]
Abstract
Calcium supplement is the most commonly adopted treatment for osteoporosis but usually requires high dose and frequency. The modality of calcium supplement is therefore overlooked by current nanomedicine-based osteoporosis therapies without proper oral formulations. Herein, we proposed a tetracycline (Tc) modified and monostearin (MS) coated amorphous calcium carbonate (ACC) platform (TMA) as oral bone targeted and osteoporosis microenvironment (water/pH) responsive carrier for in situ calcium supplement. Moreover, current osteoporosis therapies also fall short of finding suitable molecular target and effective therapeutic regimen to further increase the therapeutic efficacy over available treatment means. As a result, the simvastatin (Sim) was loaded into TMA to construct drug delivery system (TMA/Sim) capable of synergistically activating the bone morphogenetic proteins (BMPs)-Smad pathway to provide a novel therapeutic regimen for osteoblast promotion mediated osteoporosis therapy. Our results revealed that optimized TMA showed high accessibility and oral availability with targeted drug delivery to bone tissue. Most importantly, benefit from the effective in situ calcium supplement and targeted Sim delivery, this therapeutic regime (TMA/Sim) achieved better synergetic effects than conventional combination strategies with promising osteoporosis reversion performance under low calcium dosage (1/10 of commercial calcium carbonate tablet) and significantly attenuated side effects.
Collapse
|
38
|
Polymer colloids as drug delivery systems for the treatment of arthritis. Adv Colloid Interface Sci 2020; 285:102273. [PMID: 33002783 DOI: 10.1016/j.cis.2020.102273] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/07/2020] [Accepted: 09/15/2020] [Indexed: 11/21/2022]
Abstract
The most common types of arthritis are osteoarthritis (OA) and rheumatoid arthritis (RA) which are themain causes of disability and pain among older people. Current treatment of arthritis mainly consists of oral and intra-articular medications. Despite the efficacy of the intraarticular injections over the oral treatment, it is still limited by the rapid clearance of the injected drug. Therefore, a rational design of drug delivery systems (DDSs) able to delivery drugs in controlled manner and for required period of time to the arthritis joint is a key in developing safe and effective formulations for OA and RA. In this paper various colloidal systems like nanoparticles, liposomes, cationic carriers, hydrogels, and emulsion-based carriers were presented and discussed in light of their use and efficacy as delivery systems to transport therapeutics for arthritis treatment. Factors influencing the delivery efficacy such as size, charge, structure, drug uptake, retention and its release profile alongside with cytocompatibility and safety were addressed. Moreover, the advantages and disadvantages of the different colloidal systems were emphasised.
Collapse
|
39
|
The effects of chemical crosslinking manners on the physical properties and biocompatibility of collagen type I/hyaluronic acid composite hydrogels. Int J Biol Macromol 2020; 160:1201-1211. [DOI: 10.1016/j.ijbiomac.2020.05.208] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/26/2020] [Accepted: 05/23/2020] [Indexed: 02/04/2023]
|
40
|
Wang Q, He L, Fan D, Liang W, Wang X, Fang J. PLA2-Triggered Release of Drugs from Self-Assembled Lipid Tubules for Arthritis Treatments. ACS APPLIED BIO MATERIALS 2020; 3:6488-6496. [DOI: 10.1021/acsabm.0c00883] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Qin Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Liming He
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Donghao Fan
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenlang Liang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiaochen Wang
- Advanced Materials Processing and Analysis Center and Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida 32816, United States
| | - Jiyu Fang
- Advanced Materials Processing and Analysis Center and Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida 32816, United States
| |
Collapse
|