1
|
Zahoor I, Bala R, Wani SN, Chauhan S, Madaan R, Kumar R, Hakeem KR, Malik IA. Potential role of NSAIDs loaded nano-formulations to treat inflammatory diseases. Inflammopharmacology 2025; 33:1189-1207. [PMID: 39953360 DOI: 10.1007/s10787-025-01644-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 09/25/2024] [Indexed: 02/17/2025]
Abstract
Inflammation is a necessary immunological response that promotes survival and preserves tissue homeostasis, a common characteristic linked to various diseases. However, in some circumstances, the inflammatory response is deleterious and contributes to disease pathogenesis. Anti-inflammatory substances have poor affinity for inflamed tissues, resulting in low concentrations in the target tissue and a higher incidence of severe adverse effects. To address this issue, several potential approaches have been proposed, such as chemical modification of drug molecules and the development of nanocarriers for drug delivery. Since the development of nanotechnology at the beginning of the twenty-first century, researchers have been using the pathophysiological characteristics of inflammation, primarily leaky vasculature, and biomarker overexpression to develop nanomedicines that can deliver therapeutics via passive and active targeting mechanisms to sites of inflammation and produce therapeutic effects. Drug carriers based on nanoparticles can enhance the safety and efficacy of drugs by increasing their capacity, enhancing their solubility, combining several drugs, protecting them from metabolism, and regulating their release. An approach that shows promise in the treatment of various inflammatory diseases is the application of nanomedicines. Nanomedicine involves nanoparticles that have been loaded with a therapeutically active component. Nanomedicines can target inflammation by recognizing molecules highly expressed on endothelial cells or activated macrophage surfaces, enhancing the permeability of vessels, or even by biomimicry. A review of the research findings shows significant potential for the use of nanotechnology to enhance the quality of life for people using NSAIDs for chronic disorders by minimizing drug side effects or the duration of administration. After a brief introduction to inflammation, its various forms- acute and chronic inflammation, and the pathophysiology of inflammation, this review highlights the main innovative nanocarriers utilized for carrying various nonsteroidal anti-inflammatory drugs that have been utilized in treating various inflammatory disorders.
Collapse
Affiliation(s)
- Ishrat Zahoor
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133207, Haryana, India.
| | - Rajni Bala
- University School of Pharmaceutical Sciences, Rayat-Bhara University, Kharar, Punjab, India
| | - Shahid Nazir Wani
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
- Aman Pharmacy College, Dholakhera Udaipurwati, Jhunjhunu, Rajasthan, India
| | - Samrat Chauhan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Reecha Madaan
- Adesh College of Pharmacy, NH1 Shahabad Kurukshetra, Haryana, India
| | - Rajesh Kumar
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| | - Khalid Rehman Hakeem
- Department of Biological Sciences, Faculty of Science, King Adualaziz University, 21589, Jeddah, Saudi Arabia
- Department of Public Health, Daffodil International University, Dhaka, 1341, Bangladesh
- Centre of Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Irfan Ahmad Malik
- Department of Pharmacology, Sanjivani College of Pharmaceutical Education and Research, Kopargaon, 423603, Maharashtra, India
| |
Collapse
|
2
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2025; 14:e2402737. [PMID: 39506433 PMCID: PMC11730424 DOI: 10.1002/adhm.202402737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Wei Liu
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Jagadeesh K. Venkatesan
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Henning Madry
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Magali Cucchiarini
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| |
Collapse
|
3
|
Yi X, Leng P, Wang S, Liu L, Xie B. Functional Nanomaterials for the Treatment of Osteoarthritis. Int J Nanomedicine 2024; 19:6731-6756. [PMID: 38979531 PMCID: PMC11230134 DOI: 10.2147/ijn.s465243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/15/2024] [Indexed: 07/10/2024] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease, affecting more than 595 million people worldwide. Nanomaterials possess superior physicochemical properties and can influence pathological processes due to their unique structural features, such as size, surface interface, and photoelectromagnetic thermal effects. Unlike traditional OA treatments, which suffer from short half-life, low stability, poor bioavailability, and high systemic toxicity, nanotherapeutic strategies for OA offer longer half-life, enhanced targeting, improved bioavailability, and reduced systemic toxicity. These advantages effectively address the limitations of traditional therapies. This review aims to inspire researchers to develop more multifunctional nanomaterials and promote their practical application in OA treatment.
Collapse
Affiliation(s)
- Xinyue Yi
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, People’s Republic of China
| | - Pengyuan Leng
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| | - Supeng Wang
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, People’s Republic of China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| | - Bingju Xie
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
4
|
Qaiser R, Pervaiz F, Noreen S, Hanan H, Shoukat H, Mahmood H, Ashraf MA. Optimizing lornoxicam-loaded poly(lactic-co-glycolic acid) and (polyethylene glycol) nanoparticles for transdermal delivery: ex vivo/ in vivo inflammation evaluation. Nanomedicine (Lond) 2024; 19:1471-1485. [PMID: 38953843 PMCID: PMC11318691 DOI: 10.1080/17435889.2024.2359356] [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: 02/12/2024] [Accepted: 05/21/2024] [Indexed: 07/04/2024] Open
Abstract
Aim: This study focused on developing a topical gel incorporating lornoxicam-loaded poly(lactic-co-glycolic acid) and polyethylene glycol (PLGA-PEG) blend nanoparticles to mitigate gastrointestinal (GIT) side effects and enhance therapeutic efficacy. Materials & methods: Synthesized nanoparticles were subjected to in vitro characterization, ex vivo permeation studies, and acute oral toxicity analysis post-incorporation into the gel using a S/O/W double emulsion solvent. Results & conclusion: The nanoparticles displayed a smooth, spherical morphology (170-321 nm) with increased entrapment efficiency (96.2%). LOX exhibited a permeation rate of 70-94% from the nanoparticle-infused gel, demonstrating favorable biocompatibility at the cellular level. The formulated gel, enriched with nanoparticles, holds promising prospects for drug-delivery systems and promising improved therapeutic outcomes for LOX.
Collapse
Affiliation(s)
- Rubina Qaiser
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur63100, Punjab, Pakistan
| | - Fahad Pervaiz
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur63100, Punjab, Pakistan
| | - Sobia Noreen
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur63100, Punjab, Pakistan
- Centre for Chemistry & Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, 6020, Austria
| | - Hanasul Hanan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur63100, Punjab, Pakistan
| | - Hina Shoukat
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur63100, Punjab, Pakistan
| | - Hassan Mahmood
- Linguistics & Literature Department, COMSATS University Islamabad, Lahore Campus54000, Punjab, Pakistan
| | | |
Collapse
|
5
|
Zhen J, Wan T, Sun G, Chen X, Zhang S. A ROS-responsive microsphere capsule encapsulated with NADPH oxidase 4 inhibitor ameliorates macrophage inflammation and ferroptosis. Heliyon 2024; 10:e23589. [PMID: 38187270 PMCID: PMC10770568 DOI: 10.1016/j.heliyon.2023.e23589] [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: 08/13/2023] [Revised: 11/15/2023] [Accepted: 12/07/2023] [Indexed: 01/09/2024] Open
Abstract
Inflammatory macrophages within the synovium play a pivotal role in the progression of arthritis inflammation. Effective drug therapy targeting inflammatory macrophages has long been a goal for clinicians and researchers. The standard approach for treating osteoarthritis (OA) involves systemic treatment and local injection. However, the high incidence of side effects associated with long-term drug administration increases the risk of complications in patients. Additionally, the rapid clearance of the joint cavity poses a biological barrier to the therapeutic effect. NADPH oxidase 4 (NOX4) is an enzyme protein regulating the cellular redox state by generating reactive oxygen species (ROS) within the cell. In this study, we designed and fabricated a hydrogel microsphere consisting of methyl methacrylate (MMA) and polyvinyl acetate (PVA) as the outer layer structure. We then loaded GLX351322 (GLX), a novel selective NOX4 inhibitor, into hydrogel microspheres through self-assembly with the compound polyethylene glycol ketone mercaptan (mPEG-TK) containing a disulfide bond, forming nanoparticles (mPEG-TK-GLX), thus creating a two-layer drug-loaded microspheres capsule with ROS-responsive and slow-releasing capabilities. Our results demonstrate that mPEG-TK-GLX@PVA-MMA effectively suppressed TBHP-induced inflammation, ROS production, and ferroptosis, indicating a promising curative strategy for OA and other inflammatory diseases in the future.
Collapse
Affiliation(s)
- Jinze Zhen
- Department of Oral Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Tianhao Wan
- Department of Oral Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Guangxin Sun
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Oral Diseases Laboratory of Liaoning, Shenyang, 110000, China
| | - Xinwei Chen
- Department of Oral Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Shanyong Zhang
- Department of Oral Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| |
Collapse
|
6
|
Li X, Wu X. The microspheres/hydrogels scaffolds based on the proteins, nucleic acids, or polysaccharides composite as carriers for tissue repair: A review. Int J Biol Macromol 2023; 253:126611. [PMID: 37652329 DOI: 10.1016/j.ijbiomac.2023.126611] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/31/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
There are many studies on specific macromolecules and their contributions to tissue repair. Macromolecules have supporting and protective effects in organisms and can help regrow, reshape, and promote self-repair and regeneration of damaged tissues. Macromolecules, such as proteins, nucleic acids, and polysaccharides, can be constructed into hydrogels for the preparation of slow-release drug agents, carriers for cell culture, and platforms for gene delivery. Hydrogels and microspheres are fabricated by chemical crosslinking or mixed co-deposition often used as scaffolds, drug carriers, or cell culture matrix, provide proper mechanical support and nutrient delivery, a well-conditioned environment that to promote the regeneration and repair of damaged tissues. This review provides a comprehensive overview of recent developments in the construction of macromolecules into hydrogels and microspheres based on the proteins, nucleic acids, polysaccharides and other polymer and their application in tissue repair. We then discuss the latest research trends regarding the advantages and disadvantages of these composites in repair tissue. Further, we examine the applications of microspheres/hydrogels in different tissue repairs, such as skin tissue, cartilage, tumor tissue, synovial, nerve tissue, and cardiac repair. The review closes by highlighting the challenges and prospects of microspheres/hydrogels composites.
Collapse
Affiliation(s)
- Xian Li
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Xinlin Wu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China.
| |
Collapse
|
7
|
Gomes JM, Silva SS, Rodrigues LLC, Reis RL. Alginate/acemannan-based beads loaded with a biocompatible ionic liquid as a bioactive delivery system. Int J Biol Macromol 2023:125026. [PMID: 37244345 DOI: 10.1016/j.ijbiomac.2023.125026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/08/2023] [Accepted: 05/20/2023] [Indexed: 05/29/2023]
Abstract
Combining biomacromolecules with green chemistry principles and clean technologies has proven to be an effective approach for drug delivery, providing a prolonged and sustained release of the encapsulated material. The current study investigates the potential of cholinium caffeate (Ch[Caffeate]), a phenolic-based biocompatible ionic liquid (Bio-IL) entrapped in alginate/acemannan beads, as a drug delivery system able to reduce local joint inflammation on osteoarthritis (OA) treatment. The synthesized Bio-IL has antioxidant and anti-inflammatory actions that, combined with biopolymers as 3D architectures, promote the entrapment and sustainable release of the bioactive molecules over time. The physicochemical and morphological characterization of the beads (ALC, ALAC0,5, ALAC1, and ALAC3, containing 0, 0.5, 1, and 3 %(w/v) of Ch[Caffeate], respectively) revealed a porous and interconnected structure, with medium pore sizes ranging from 209.16 to 221.30 μm, with a high swelling ability (up 2400 %). Ch[Caffeate] significantly improved the antioxidant activities of the constructs by 95 % and 97 % for ALAC1 and ALAC3, respectively, when compared to ALA (56 %). Besides, the structures provided the environment for ATDC5 cell proliferation, and cartilage-like ECM formation, supported by the increased GAGs in ALAC1 and ALAC3 formulations after 21 days. Further, the ability to block the secretion of pro-inflammatory cytokines (TNF-α and IL-6), from differentiated THP-1 was evidenced by ChAL-Ch[Caffeate] beads. These outcomes suggest that the established strategy based on using natural and bioactive macromolecules to develop 3D constructs has great potential to be used as therapeutic tools for patients with OA.
Collapse
Affiliation(s)
- Joana M Gomes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - Simone S Silva
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - Luísa L C Rodrigues
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
8
|
Avendaño-Godoy J, Poblete MG, Ramos RG, Gómez-Gaete C. Evaluation of the Effects of Gamma Radiation Sterilization on Rhein-Loaded Biodegradable Microparticles for the Treatment of Osteoarthritis. J Pharm Sci 2023; 112:837-843. [PMID: 36347357 DOI: 10.1016/j.xphs.2022.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/02/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
In previous work, we reported on the design of biodegradable rhein-loaded PLGA microparticles for the treatment of osteoarthritis. Considering that a formulation designed for intra-articular administration must meet sterility requirements to guarantee its safety, in this study the effect of gamma radiation sterilization on these microparticles was evaluated. The size, morphology, and surface characteristics of the microparticles and the encapsulation efficiency of rhein were not affected by the sterilization process. Although DSC and PXRD analyses suggested otherwise, rhein release profiles were not altered by gamma radiation. The release of rhein from the microparticles was fitted to a Gompertz model. In conclusion, the results of this study suggest that gamma radiation is a suitable method for the sterilization of rhein-loaded PLGA microparticles to enable their intra-articular administration in order to provide a therapeutic solution to patients suffering from chronic joint diseases.
Collapse
Affiliation(s)
- Javier Avendaño-Godoy
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Maria Gabriela Poblete
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Ricardo Godoy Ramos
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Carolina Gómez-Gaete
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
9
|
Pape E, Pinzano A, Henrionnet C, Scala-Bertola J, Gillet P, Gambier N. Rat synovial tissue and blood rapamycin pharmacokinetics after intra-articular injection of free solution or nanoparticles vs free rapamycin intravenous shot. Int J Pharm 2022; 624:122026. [PMID: 35863592 DOI: 10.1016/j.ijpharm.2022.122026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/09/2022] [Accepted: 07/13/2022] [Indexed: 10/17/2022]
Abstract
Intra-articular (IA) injection of a chondroprotective candidate may delay the osteoarthritis (OA) course, but its rapid absorption into systemic circulation may limit efficacy and produce untoward effects. We compared the pharmacokinetics (PK) of IA rapamycin injected as sustained release in nanoparticles (NPs) versus a free rapamycin suspension in the rat knee compared to an intravenous (IV) free rapamycin shot taken as a reference. Rats received either a single IV injection of free rapamycin (10 µM) or an IA of free or NPs-loaded rapamycin. After sequential exsanguination (15, 30, 60, 180, 360 min, D1, and D7), knee synovial tissue (ST) and cartilage histology were performed. Blood and ST concentrations (LC-MS/MS), PK parameters (area under the curve: AUC; mean residence time: MRT; elimination half-life: T1/2), and IA biocompatibility were assessed. AUCIV was significantly higher for IV than for both IA injections (AUCIA free and AUCIA NPs), with 4248 vs 28 and 74 µg.min.L-1. For ST parameters, we observed a significant difference between AUCIA free and AUCIA NPs with 3735 and 10513 µg.min.L-1 correspondingly. Articular T1/2 and MRT were higher after NPs than after free rapamycin injection: 57.8 and 5.0 h for T1/2 and 80.6 and 5.5 h for MRT, respectively. Histological analysis revealed no chondral injuries and slight transient synovitis only 3 h after the administration of NPs. In the rat knee, rapamycin-loaded NPs delivery via a single IA injection is biocompatible and prolongs synovium joint residency, diminishes blood levels, and reduces detrimental systemic exposure.
Collapse
Affiliation(s)
- Elise Pape
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie-Toxicologie, Pharmacovigilance & CEIPA, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Astrid Pinzano
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France.
| | | | - Julien Scala-Bertola
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie-Toxicologie, Pharmacovigilance & CEIPA, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Pierre Gillet
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie-Toxicologie, Pharmacovigilance & CEIPA, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Nicolas Gambier
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie-Toxicologie, Pharmacovigilance & CEIPA, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| |
Collapse
|
10
|
The synthetic artificial stem cell (SASC): Shifting the paradigm of cell therapy in regenerative engineering. Proc Natl Acad Sci U S A 2022; 119:2116865118. [PMID: 34987101 PMCID: PMC8764679 DOI: 10.1073/pnas.2116865118] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 12/14/2022] Open
Abstract
This paper presents the synthetic artificial stem cell (SASC) system: a versatile therapy which provides the ability to tailor paracrine responses of different cells and provide a more potent regenerative effect for targeted tissues. Upon challenging the SASC system against an osteoarthritis model, we demonstrate that the factors combined tailored for chondrogenesis have a potent antiinflammatory and chondroprotective effect. This paper also demonstrates the in vivo capacity of SASC to attenuate proteoglycan depletion in the cartilage extracellular matrix while also improving biomechanical properties of the resulting cartilage. We report the first of many applications of the SASC system, which provides a promising step toward clinical translation of a minimally immunogenic stem cell with many commercial advantages over its biological counterpart. Stem cells are of great interest in tissue regeneration due to their ability to modulate the local microenvironment by secreting bioactive factors (collectively, secretome). However, secretome delivery through conditioned media still requires time-consuming cell isolation and maintenance and also may contain factors antagonistic to targeted tissue regeneration. We have therefore engineered a synthetic artificial stem cell (SASC) system which mimics the paracrine effect of the stem cell secretome and provides tailorability of the composition for targeted tissue regeneration. We report the first of many applications of the SASC system we have formulated to treat osteoarthritis (OA). Choosing growth factors important to chondrogenesis and encapsulating respective recombinant proteins in poly (lactic-coglycolic acid) 85:15 (PLGA) we fabricated the SASC system. We compared the antiinflammatory and chondroprotective effects of SASC to that of adipose-derived stem cells (ADSCs) using in vitro interleukin 1B-induced and in vivo collagenase-induced osteoarthritis rodent models. We have designed SASC as an injectable therapy with controlled release of the formulated secretome. In vitro, SASC showed significant antiinflammatory and chondroprotective effects as seen by the up-regulation of SOX9 and reduction of nitric oxide, ADAMTS5, and PRG4 genes compared to ADSCs. In vivo, treatment with SASC and ADSCs significantly attenuated cartilage degeneration and improved the biomechanical properties of the articular cartilage in comparison to OA control. This SASC system demonstrates the feasibility of developing a completely synthetic, tailorable stem cell secretome which reinforces the possibility of developing a new therapeutic strategy that provides better control over targeted tissue engineering applications.
Collapse
|
11
|
Sui L, Xu G, Hao Y, Wang X, Tang K. Engineering of marizomib loaded polymeric nanoparticles: In vivo safety profile and In vitro proliferation in hepatocellular carcinoma. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
12
|
Pape E, Parent M, Pinzano A, Sapin-Minet A, Henrionnet C, Gillet P, Scala-Bertola J, Gambier N. Rapamycin-loaded Poly(lactic-co-glycolic) acid nanoparticles: Preparation, characterization, and in vitro toxicity study for potential intra-articular injection. Int J Pharm 2021; 609:121198. [PMID: 34662644 DOI: 10.1016/j.ijpharm.2021.121198] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/29/2021] [Accepted: 10/10/2021] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease. Rapamycin is a potential candidate for OA treatment by increasing the autophagy process implicated in its physiopathology. To optimize Rapamycin profit and avoid systemic side effects, intra-articular (i.a.) administration appeared helpful. However, Rapamycin's highly hydrophobic nature and low bioavailability made it challenging to develop purpose-made drug delivery systems to overcome these limitations. We developed Rapamycin-loaded nanoparticles (NPs) using poly (lactic-co-glycolic acid) by emulsion/evaporation method. We evaluated these NPs' cytocompatibility towards cartilage (chondrocytes) and synovial membrane cells (synoviocytes) for a potential i.a. administration. The in vitro characterization of Rapamycin-loaded NPs had shown a suitable profile for an i.a. administration. In vitro biocompatibility of NPs was highlighted to 10 µM of Rapamycin for both synoviocytes and chondrocytes, but significant toxicity was observed with higher concentrations. Besides, synoviocytes are more sensitive to Rapamycin-loaded NPs than chondrocytes. Finally, we observed in vitro that an adapted formulated Rapamycin-loaded NPs could be safe at suitable i.a. injection concentrations. The toxic effect of Rapamycin encapsulated in these NPs on both articular cells was dose-dependent. After Rapamycin-loaded NPs i.a. administration, local retention, in situ safety, and systemic release should be evaluated with experimental in vivo models.
Collapse
Affiliation(s)
- Elise Pape
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | | | - Astrid Pinzano
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France.
| | | | | | - Pierre Gillet
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Julien Scala-Bertola
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| | - Nicolas Gambier
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France; Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511 Vandœuvre-Lès-Nancy, France.
| |
Collapse
|
13
|
Han Y, Yang J, Zhao W, Wang H, Sun Y, Chen Y, Luo J, Deng L, Xu X, Cui W, Zhang H. Biomimetic injectable hydrogel microspheres with enhanced lubrication and controllable drug release for the treatment of osteoarthritis. Bioact Mater 2021; 6:3596-3607. [PMID: 33869900 PMCID: PMC8022850 DOI: 10.1016/j.bioactmat.2021.03.022] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/23/2022] Open
Abstract
The occurrence of osteoarthritis (OA) is highly associated with the reduced lubrication property of the joint, where a progressive and irreversible damage of the articular cartilage and consecutive inflammatory response dominate the mechanism. In this study, bioinspired by the super-lubrication property of cartilage and catecholamine chemistry of mussel, we successfully developed injectable hydrogel microspheres with enhanced lubrication and controllable drug release for OA treatment. Particularly, the lubricating microspheres (GelMA@DMA-MPC) were fabricated by dip coating a self-adhesive polymer (DMA-MPC, synthesized by free radical copolymerization) on superficial surface of photo-crosslinked methacrylate gelatin hydrogel microspheres (GelMA, prepared via microfluidic technology), and encapsulated with an anti-inflammatory drug of diclofenac sodium (DS) to achieve the dual-functional performance. The tribological test and drug release test showed the enhanced lubrication and sustained drug release of the GelMA@DMA-MPC microspheres. In addition, the functionalized microspheres were intra-articularly injected into the rat knee joint with an OA model, and the biological tests including qRT-PCR, immunofluorescence staining assay, X-ray radiography and histological staining assay all revealed that the biocompatible microspheres provided significant therapeutic effect against the development of OA. In summary, the injectable hydrogel microspheres developed herein greatly improved lubrication and achieved sustained local drug release, therefore representing a facile and promising technique for the treatment of OA.
Collapse
Affiliation(s)
- Ying Han
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Jielai Yang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Weiwei Zhao
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Haimang Wang
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Yulong Sun
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Yuji Chen
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Jing Luo
- Beijing Research Institute of Automation for Machinery Industry Co., Ltd, Beijing, 100120, China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiangyang Xu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongyu Zhang
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
14
|
Mao L, Wu W, Wang M, Guo J, Li H, Zhang S, Xu J, Zou J. Targeted treatment for osteoarthritis: drugs and delivery system. Drug Deliv 2021; 28:1861-1876. [PMID: 34515606 PMCID: PMC8439249 DOI: 10.1080/10717544.2021.1971798] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The management of osteoarthritis (OA) is a clinical challenge due to the particular avascular, dense, and occluded tissue structure. Despite numerous clinical reports and animal studies, the pathogenesis and progression of OA are still not fully understood. On the basis of traditional drugs, a large number of new drugs have been continuously developed. Intra-articular (IA) administration for OA hastens the development of targeted drug delivery systems (DDS). OA drugs modification and the synthesis of bioadaptive carriers contribute to a qualitative leap in the efficacy of IA treatment. Nanoparticles (NPs) are demonstrated credible improvement of drug penetration and retention in OA. Targeted nanomaterial delivery systems show the prominent biocompatibility and drug loading-release ability. This article reviews different drugs and nanomaterial delivery systems for IA treatment of OA, in an attempt to resolve the inconsonance between in vitro and in vivo release, and explore more interactions between drugs and nanocarriers, so as to open up new horizons for the treatment of OA.
Collapse
Affiliation(s)
- Liwei Mao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Wei Wu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Miao Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jianmin Guo
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Hui Li
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Shihua Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
15
|
Gambaro FM, Ummarino A, Torres Andón F, Ronzoni F, Di Matteo B, Kon E. Drug Delivery Systems for the Treatment of Knee Osteoarthritis: A Systematic Review of In Vivo Studies. Int J Mol Sci 2021; 22:ijms22179137. [PMID: 34502046 PMCID: PMC8431358 DOI: 10.3390/ijms22179137] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/21/2021] [Accepted: 08/22/2021] [Indexed: 12/12/2022] Open
Abstract
Many efforts have been made in the field of nanotechnology to improve the local and sustained release of drugs, which may be helpful to overcome the present limitations in the treatment of knee OA. Nano-/microparticles and/or hydrogels can be now engineered to improve the administration and intra-articular delivery of specific drugs, targeting molecular pathways and pathogenic mechanisms involved in OA progression and remission. In order to summarize the current state of this field, a systematic review of the literature was performed and 45 relevant studies were identified involving both animal models and humans. We found that polymeric nanoparticles loaded with anti-inflammatory drugs (i.e., dexamethasone or celecoxib) are the most frequently investigated drug delivery systems, followed by microparticles and hydrogels. In particular, the nanosystem most frequently used in preclinical research consists of PLGA-nanoparticles loaded with corticosteroids and non-steroidal anti-inflammatory drugs. Overall, improvement in histological features, reduction in joint inflammation, and improvement in clinical scores in patients were observed. The last advances in the field of nanotechnology could offer new opportunities to treat patients affected by knee OA, including those with previous meniscectomy. New smart drug delivery approaches, based on nanoparticles, microparticles, and hydrogels, may enhance the therapeutic potential of intra-articular agents by increasing the permanence of selected drugs inside the joint and better targeting specific receptors and tissues.
Collapse
Affiliation(s)
- Francesco Manlio Gambaro
- Department of Biomedical Sciences, Humanitas University Pieve Emanuele, 20090 Milan, Italy; (A.U.); (F.T.A.); (F.R.); (E.K.)
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
- Correspondence:
| | - Aldo Ummarino
- Department of Biomedical Sciences, Humanitas University Pieve Emanuele, 20090 Milan, Italy; (A.U.); (F.T.A.); (F.R.); (E.K.)
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| | - Fernando Torres Andón
- Department of Biomedical Sciences, Humanitas University Pieve Emanuele, 20090 Milan, Italy; (A.U.); (F.T.A.); (F.R.); (E.K.)
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Flavio Ronzoni
- Department of Biomedical Sciences, Humanitas University Pieve Emanuele, 20090 Milan, Italy; (A.U.); (F.T.A.); (F.R.); (E.K.)
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
| | - Berardo Di Matteo
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
- Department of Traumatology, Orthopaedics and Disaster Surgery, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Elizaveta Kon
- Department of Biomedical Sciences, Humanitas University Pieve Emanuele, 20090 Milan, Italy; (A.U.); (F.T.A.); (F.R.); (E.K.)
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| |
Collapse
|
16
|
Chen L, Wang Y, Sun L, Yan J, Mao H. Nanomedicine Strategies for Anti-Inflammatory Treatment of Noninfectious Arthritis. Adv Healthc Mater 2021; 10:e2001732. [PMID: 33870656 DOI: 10.1002/adhm.202001732] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/09/2021] [Indexed: 02/06/2023]
Abstract
Noninfectious arthritis (NIA) comprises a class of chronic and progressive inflammatory disorders that require early-stage management to prevent disease progression. The most common forms include osteoarthritis, rheumatoid arthritis, ankylosing spondylitis, and gouty arthritis. Current treatments involve nonsteroidal anti-inflammatory drugs, disease-modifying antirheumatic drugs and glucocorticoids to alleviate clinical symptoms, although regular use of these can result in a high risk of chronic kidney disease and heart failure, as well as severe adverse gastrointestinal effects. Nanomedicine offers unique opportunities to address these challenges and improve therapeutic efficacy due to its ability to deliver therapeutics locally in a sustained manner, thus extending the half-life, improving bioavailability, and reducing the side effects of these agents. This review includes a comprehensive analysis of the mechanisms of various treatment options for NIA and highlights recent progress and emerging strategies in treating NIA with nanomedicine platforms, particularly related to long-term biosafety and nonspecific targeting in designing nanomedicine delivery systems.
Collapse
Affiliation(s)
- Long Chen
- Department of Orthopedics Guizhou Provincial People's Hospital Guiyang Guizhou 550000 China
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
| | - Yuanzheng Wang
- Department of Orthopedics Guizhou Provincial People's Hospital Guiyang Guizhou 550000 China
| | - Li Sun
- Department of Orthopedics Guizhou Provincial People's Hospital Guiyang Guizhou 550000 China
| | - Jerry Yan
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
- Department of Biomedical Engineering School of Medicine Johns Hopkins University Baltimore MD 21205 USA
| | - Hai‐Quan Mao
- Translational Tissue Engineering Center Johns Hopkins School of Medicine Baltimore MD 21287 USA
- Institute for NanoBioTechnology Johns Hopkins University Baltimore MD 21218 USA
- Department of Biomedical Engineering School of Medicine Johns Hopkins University Baltimore MD 21205 USA
- Department of Materials Science and Engineering Whiting School of Engineering Johns Hopkins University Baltimore MD 21218 USA
| |
Collapse
|
17
|
Parra-Coca A, Boix-Montañés A, Calpena-Campmany AC, Colom H. In vivo pharmacokinetic evaluation of carprofen delivery from intra-articular nanoparticles in rabbits: A population modelling approach. Res Vet Sci 2021; 137:235-242. [PMID: 34049110 DOI: 10.1016/j.rvsc.2021.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/09/2021] [Accepted: 05/18/2021] [Indexed: 11/16/2022]
Abstract
Osteoarthritis is treated with COX or fosfolipase A2 inhibitors such as carprofen, a propionic acid NSAID. The enhancement of its action over the articular cartilage is mandatory to facilitate its therapeutic application. Drug uptake into the cartilage requires high synovial fluid concentrations, anticipating its rapid distribution towards bloodstream. Thus, intraarticular administration improves local targeting of the drug, lining with the site of action. A pharmacokinetic study in rabbits has been performed to evaluate carprofen nanoparticles after intraarticular administration. Pharmacokinetic analysis of plasma profiles through a modelling approach, has demonstrated the rapid distribution of drug outside of synovial chamber but mainly remaining in plasma. The data modelling has demonstrated the existence of two release-absorption processes when the nanoparticles are administered in the synovial space. Additionally, results are predictive of the PK profile of some other species such as cat, dogs or humans.
Collapse
Affiliation(s)
- A Parra-Coca
- Department of Pharmacy and Pharmaceutical technology and Physical-Chemistry, Faculty of Pharmacy and Food sciences, University of Barcelona, ES, Spain; Department of Veterinary Medicine and Zootechnic, Faculty of Agricultural Sciences, University of Applied and Environmental Sciences (U.D.C.A.), Bogotá, CO, Colombia
| | - A Boix-Montañés
- Department of Pharmacy and Pharmaceutical technology and Physical-Chemistry, Faculty of Pharmacy and Food sciences, University of Barcelona, ES, Spain.
| | - A C Calpena-Campmany
- Department of Pharmacy and Pharmaceutical technology and Physical-Chemistry, Faculty of Pharmacy and Food sciences, University of Barcelona, ES, Spain
| | - H Colom
- Department of Pharmacy and Pharmaceutical technology and Physical-Chemistry, Faculty of Pharmacy and Food sciences, University of Barcelona, ES, Spain
| |
Collapse
|
18
|
Rahimi M, Charmi G, Matyjaszewski K, Banquy X, Pietrasik J. Recent developments in natural and synthetic polymeric drug delivery systems used for the treatment of osteoarthritis. Acta Biomater 2021; 123:31-50. [PMID: 33444800 DOI: 10.1016/j.actbio.2021.01.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/15/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA), is a common musculoskeletal disorder that will progressively increase in older populations and is expected to be the most dominant cause of disability in the world population by 2030. The progression of OA is controlled by a multi-factorial pathway that has not been completely elucidated and understood yet. However, over the years, research efforts have provided a significant understanding of some of the processes contributing to the progression of OA. Both cartilage and bone degradation processes induce articular cells to produce inflammatory mediators that produce proinflammatory cytokines that block the synthesis of collagen type II and aggrecan, the major components of cartilage. Systemic administration and intraarticular injection of anti-inflammatory agents are the first-line treatments of OA. However, small anti-inflammatory molecules are rapidly cleared from the joint cavity which limits their therapeutic efficacy. To palliate this strong technological drawback, different types of polymeric materials such as microparticles, nanoparticles, and hydrogels, have been examined as drug carriers for the delivery of therapeutic agents to articular joints. The main purpose of this review is to provide a summary of recent developments in natural and synthetic polymeric drug delivery systems for the delivery of anti-inflammatory agents to arthritic joints. Furthermore, this review provides an overview of the design rules that have been proposed so far for the development of drug carriers used in OA therapy. Overall it is difficult to state clearly which polymeric platform is the most efficient one because many advantages and disadvantages could be pointed to both natural and synthetic formulations. That requires further research in the near future.
Collapse
|
19
|
Tryfonidou MA, de Vries G, Hennink WE, Creemers LB. "Old Drugs, New Tricks" - Local controlled drug release systems for treatment of degenerative joint disease. Adv Drug Deliv Rev 2020; 160:170-185. [PMID: 33122086 DOI: 10.1016/j.addr.2020.10.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022]
Abstract
Osteoarthritis (OA) and chronic low back pain (CLBP) caused by intervertebral disc (IVD) degeneration are joint diseases that have become major causes for loss of quality of life worldwide. Despite the unmet need, effective treatments other than invasive, and often ineffective, surgery are lacking. Systemic administration of drugs entails suboptimal local drug exposure in the articular joint and IVD. This review provides an overview of the potency of biomaterial-based drug delivery systems as novel treatment modality, with a focus on the biological effects of drug release systems that have reached translation at the level of in vivo models and relevant ex vivo models. These studies have shown encouraging results of biomaterial-based local delivery of several types of drugs, mostly inhibitors of inflammatory cytokines or other degenerative factors. Prevention of inflammation and degeneration and pain relief was achieved, although mainly in small animal models, with interventions applied at an early disease stage. Less convincing data were obtained with the delivery of regenerative factors. Multidisciplinary efforts towards tackling the discord between in vitro and in vivo release, combined with adaptations in the regulatory landscape may be needed to enhance safe and expeditious introduction of more and more effective controlled release-based treatments with the OA and CLBP patients.
Collapse
|
20
|
Kou L, Xiao S, Sun R, Bao S, Yao Q, Chen R. Biomaterial-engineered intra-articular drug delivery systems for osteoarthritis therapy. Drug Deliv 2019; 26:870-885. [PMID: 31524006 PMCID: PMC6758706 DOI: 10.1080/10717544.2019.1660434] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/19/2019] [Accepted: 08/22/2019] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis (OA) is a progressive and degenerative disease, which is no longer confined to the elderly. So far, current treatments are limited to symptom relief, and no valid OA disease-modifying drugs are available. Additionally, OA relative joint is challenging for drug delivery, since the drugs experience rapid clearance in joint, showing a poor bioavailability. Existing therapeutic drugs, like non-steroidal anti-inflammatory drugs (NSAIDs) and corticosteroids, are not conducive for long-term use due to adverse effects. Though supplementations, including chondroitin sulfate and glucosamine, have shown beneficial effects on joint tissues in OA, their therapeutic use is still debatable. New emerging agents, like Kartogenin (KGN) and Interleukin-1 receptor antagonist (IL-1 ra), without a proper formulation, still will not work. Therefore, it is urgent to establish a suitable and efficient drug delivery system for OA therapy. In this review, we pay attention to various types of drug delivery systems and potential therapeutic drugs that may escalate OA treatments.
Collapse
Affiliation(s)
- Longfa Kou
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shuyi Xiao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rui Sun
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shihui Bao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing Yao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ruijie Chen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
21
|
Xi Y, Jiang T, Chaurasiya B, Zhou Y, Yu J, Wen J, Shen Y, Ye X, Webster TJ. Advances in nanomedicine for the treatment of ankylosing spondylitis. Int J Nanomedicine 2019; 14:8521-8542. [PMID: 31806960 PMCID: PMC6831987 DOI: 10.2147/ijn.s216199] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 08/13/2019] [Indexed: 12/17/2022] Open
Abstract
Ankylosing spondylitis (AS) is a complex disease characterized by inflammation and ankylosis primarily at the cartilage-bone interface. The disease is more common in young males and risk factors include both genetic and environmental. While the pathogenesis of AS is not completely understood, it is thought to be an immune-mediated disease involving inflammatory cellular infiltrates, and human leukocyte antigen-B27. Currently, there is no specific diagnostic technique available for this disease; therefore conventional diagnostic approaches such as clinical symptoms, laboratory tests and imaging techniques are used. There are various review papers that have been published on conventional treatment approaches, and in this review work, we focus on the more promising nanomedicine-based treatment modalities to move this field forward.
Collapse
Affiliation(s)
- Yanhai Xi
- Department of Spine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Tingwang Jiang
- Department of Immunology and Microbiology, Institution of Laboratory Medicine of Changshu, Changshu, Jiangsu215500, People’s Republic of China
| | - Birendra Chaurasiya
- Department of Pharmaceutics, Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Yanyan Zhou
- Department of Spine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Jiangmin Yu
- Department of Spine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Jiankun Wen
- Department of Spine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Yan Shen
- Department of Pharmaceutics, Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Xiaojian Ye
- Department of Spine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
22
|
Rudnik‐Jansen I, Tellegen AR, Pouran B, Schrijver K, Meij BP, Emans PJ, de Gendt E, Thomas RE, Kik MJ, de Visser HM, Weinans H, Egas A, van Maarseveen E, Woike N, Mihov G, Thies J, Tryfonidou MA, Creemers LB. Local controlled release of corticosteroids extends surgically induced joint instability by inhibiting tissue healing. Br J Pharmacol 2019; 176:4050-4064. [PMID: 31378925 PMCID: PMC6811746 DOI: 10.1111/bph.14817] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 05/28/2019] [Accepted: 07/11/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Corticosteroids are intra-articularly injected to relieve pain in joints with osteoarthritis (OA) or acute tissue damage such as ligament or tendon tears, despite its unverified contraindication in unstable joints. Biomaterial-based sustained delivery may prolong reduction of inflammatory pain, while avoiding harmful peak drug concentrations. EXPERIMENTAL APPROACH The applicability of prolonged corticosteroid exposure was examined in a rat model of anterior cruciate ligament and medial meniscus transection (ACLT + pMMx) with ensuing degenerative changes. KEY RESULTS Intra-articular injection of a bolus of the corticosteroid triamcinolone acetonide (TAA) resulted in enhanced joint instability in 50% of the joints, but neither instability-induced OA cartilage degeneration, synovitis, nor the OA-related bone phenotype was affected. However, biomaterial microsphere-based extended TAA release enhanced instability in 94% of the animals and induced dystrophic calcification and exacerbation of cartilage degeneration. In healthy joints, injection with TAA releasing microspheres had no effect at all. In vitro, TAA inhibited cell migration out of joint tissue explants, suggesting inhibited tissue healing in vivo as mechanisms for enhanced instability and subsequent cartilage degeneration. CONCLUSIONS AND IMPLICATIONS We conclude that short-term TAA exposure has minor effects on surgically induced unstable joints, but its extended presence is detrimental by extending instability and associated joint degeneration through compromised healing. This supports a contraindication of prolonged corticosteroid exposure in tissue damage-associated joint instability, but not of brief exposure.
Collapse
Affiliation(s)
- Imke Rudnik‐Jansen
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Anna R. Tellegen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Behdad Pouran
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical Maritime and Materials EngineeringDelft University of TechnologyDelftThe Netherlands
| | - Karin Schrijver
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Björn P. Meij
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Pieter J. Emans
- Department of OrthopaedicsMaastricht University Medical Center UtrechtUtrechtThe Netherlands
| | - Erin de Gendt
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Rachel E. Thomas
- Department of Pathobiology, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Marja J.L. Kik
- Department of Pathobiology, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Huub M. de Visser
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Harrie Weinans
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical Maritime and Materials EngineeringDelft University of TechnologyDelftThe Netherlands
| | - Annelies Egas
- Division Laboratory and Pharmacy, Clinical PharmacyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Erik van Maarseveen
- Division Laboratory and Pharmacy, Clinical PharmacyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Nina Woike
- Dept of Science and InnovationDSM BiomedicalGeleenThe Netherlands
| | - George Mihov
- Dept of Science and InnovationDSM BiomedicalGeleenThe Netherlands
| | - Jens Thies
- Dept of Science and InnovationDSM BiomedicalGeleenThe Netherlands
| | - Marianna A. Tryfonidou
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Laura B. Creemers
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
23
|
Nanomedicine for the effective and safe delivery of non-steroidal anti-inflammatory drugs: A review of preclinical research. Eur J Pharm Biopharm 2019; 142:179-194. [DOI: 10.1016/j.ejpb.2019.06.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/22/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023]
|
24
|
Liang X, Chen Y, Wu L, Maharjan A, Regmi B, Zhang J, Gui S. In situ hexagonal liquid crystal for intra-articular delivery of sinomenine hydrochloride. Biomed Pharmacother 2019; 117:108993. [PMID: 31228805 DOI: 10.1016/j.biopha.2019.108993] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/30/2019] [Accepted: 05/13/2019] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to investigate the release behaviors of sinomenine hydrochloride loaded via in situ hexagonal liquid crystal (ISH), and its potential to improve the local bioavailability in knee joints of sinomenine hydrochloride (SMH) after intra-articular administration. The ISH was prepared by a liquid precursor mixture containing phytantriol (PT), Vitamin E acetate (VEA), ethanol (ET), and water. The in vitro release profiles revealed a sustained release of SMH from the optimized ISH formula (PT/VEA/ET/water, 60.8:3.2:16.0:20.0, w/w/w/w), which was selected for the in vivo pharmacokinetics and preliminary pharmacodynamics studies. In both healthy and adjuvant-induced arthritis (AA) rats, the SMH loaded ISH showed significantly smaller SMH AUC0-∞ in plasma (P < 0.01), and higher SMH concentration in synoviums (2˜168 h) than that of SMH solution, indicating that the ISH significantly reduced the leakage of SMH into systemic circulation. The t1/2α of SMH loaded ISH in the knee joints of AA rats, was longer (13.42 h) than that of healthy rats (1.34 h) (P < 0.05), most likely that in vivo drug release behavior of SMH loaded ISH was affected by the physiological environment of the joint. It was found that the SMH loaded ISH could benefit AA-rats by suppressing the level of IL-1β in comparison to SMH solutions. The results of the histopathology of knee joints in AA rats displayed that the SMH loaded ISH might be suitable for the development of treatment strategies for rheumatoid arthritis diseases.
Collapse
Affiliation(s)
- Xiao Liang
- Anhui University of Chinese Medicine, No.1 Qianjiang Street, Xinzhan Distinct, 230012, Hefei, Anhui, China
| | - Yulin Chen
- Anhui University of Chinese Medicine, No.1 Qianjiang Street, Xinzhan Distinct, 230012, Hefei, Anhui, China
| | - Li Wu
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Abi Maharjan
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | | | - Jiwen Zhang
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shuangying Gui
- Anhui University of Chinese Medicine, No.1 Qianjiang Street, Xinzhan Distinct, 230012, Hefei, Anhui, China.; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China; Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province.
| |
Collapse
|
25
|
Sulistio A, Mansfeld FM, Reyes-Ortega F, D’Souza AM, Ng SMY, Birkett S, Blencowe A, Qiao GG, Little CB, Shu CC, Bendele AM, Valade D, Donohue AC, Quinn JF, Whittaker MR, Davis TP, Tait RJ. Intra-articular Treatment of Osteoarthritis with Diclofenac-Conjugated Polymer Reduces Inflammation and Pain. ACS APPLIED BIO MATERIALS 2019; 2:2822-2832. [DOI: 10.1021/acsabm.9b00232] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Adrian Sulistio
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
- PolyActiva Pty Ltd., Level 9, 31 Queen Street, Melbourne, Victoria 3000, Australia
| | - Friederike M. Mansfeld
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
- Children’s Cancer Institute, Australian Centre for NanoMedicine and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Felisa Reyes-Ortega
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
- PolyActiva Pty Ltd., Level 9, 31 Queen Street, Melbourne, Victoria 3000, Australia
| | - Asha M. D’Souza
- PolyActiva Pty Ltd., Level 9, 31 Queen Street, Melbourne, Victoria 3000, Australia
- Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
| | - Sarah M. Y. Ng
- PolyActiva Pty Ltd., Level 9, 31 Queen Street, Melbourne, Victoria 3000, Australia
- Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
| | - Stephen Birkett
- PolyActiva Pty Ltd., Level 9, 31 Queen Street, Melbourne, Victoria 3000, Australia
- Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
| | - Anton Blencowe
- School of Pharmacy and Medical Sciences, University of South Australia, Mawson Lake, South Australia 5095, Australia
| | - Greg G. Qiao
- Department of Chemical and Biomolecular Engineering, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Christopher B. Little
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute, University of Sydney at Royal North Shore Hospital, St Leonards, New South Wales 2065, Australia
| | - Cindy C. Shu
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute, University of Sydney at Royal North Shore Hospital, St Leonards, New South Wales 2065, Australia
| | - Alison M. Bendele
- Bolder BioPATH Inc.,5541 Central Avenue, Suite 160, Boulder, Colorado 80301, United States
| | - David Valade
- PolyActiva Pty Ltd., Level 9, 31 Queen Street, Melbourne, Victoria 3000, Australia
- Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
| | - Andrew C. Donohue
- PolyActiva Pty Ltd., Level 9, 31 Queen Street, Melbourne, Victoria 3000, Australia
- Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
| | - John F. Quinn
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
| | - Michael R. Whittaker
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
| | - Thomas P. Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Russell J. Tait
- PolyActiva Pty Ltd., Level 9, 31 Queen Street, Melbourne, Victoria 3000, Australia
- Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
26
|
Christian R, Thakkar V, Patel T, Gohel M, Baldaniya L, Shah P, Pandya T, Gandhi T. Development of Biodegradable Injectable In situ Forming Implants for Sustained Release of Lornoxicam. Curr Drug Deliv 2019; 16:66-78. [PMID: 30264681 DOI: 10.2174/1567201815666180927155710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 08/03/2018] [Accepted: 09/14/2018] [Indexed: 01/17/2023]
Abstract
OBJECTIVE The focus of this study was to develop in situ injectable implants of Lornoxicam which could provide sustained drug release. METHODS Biodegradable in situ injectable implants were prepared by polymer precipitation method using polylactide-co-glycolide (PLGA). An optimized formulation was obtained on the basis of drug entrapment efficiency, gelling behavior and in vitro drug release. The compatibility of the formulation ingredients were tested by Fourier transform infrared (FT-IR) spectroscopy, and differential scanning colorimetry (DSC). SEM study was performed to characterize in vivo behavior of in situ implant. Pharmacokinetic study and in vivo gelling study of the optimized formulation were performed on Sprague-Dawley rats. Stability testing of optimized formulation was also performed. RESULTS The drug entrapment efficiency increased and burst release decreased with an increase in the polymer concentration. Sustained drug release was obtained up to five days. SEM photomicrographs indicated uniform gel formation. Chemical interaction between the components of the formulation was not observed by FT-IR and DSC study. Pharmacokinetic studies of the optimized formulation revealed that the maximum plasma concentration (Cmax), time to achieve Cmax (Tmax) and area under plasma concentration curve (AUC) were significantly higher than the marketed intramuscular injection of lornoxicam. Stability study of optimized batch showed no change in physical and chemical characteristics. CONCLUSION Lornoxicam can be successfully formulated as in situ injectable implant that provides long-term management of inflammatory disorders with improved patient compliance.
Collapse
Affiliation(s)
- Ruby Christian
- Department of Pharmaceutics, Anand Pharmacy College, Anand- 388 001, Gujarat, India
| | - Vaishali Thakkar
- Department of Pharmaceutics, Anand Pharmacy College, Anand- 388 001, Gujarat, India
| | - Tushar Patel
- Department of Pharmaceutics, Anand Pharmacy College, Anand- 388 001, Gujarat, India
| | - Mukesh Gohel
- Department of Pharmaceutics, Anand Pharmacy College, Anand- 388 001, Gujarat, India
| | - Lalji Baldaniya
- Department of Pharmaceutics, Anand Pharmacy College, Anand- 388 001, Gujarat, India
| | - Purvi Shah
- Department of Pharmaceutics, Anand Pharmacy College, Anand- 388 001, Gujarat, India
| | - Tosha Pandya
- Department of Pharmaceutics, Anand Pharmacy College, Anand- 388 001, Gujarat, India
| | - Tejal Gandhi
- Department of Pharmaceutics, Anand Pharmacy College, Anand- 388 001, Gujarat, India
| |
Collapse
|
27
|
Pawar VA, Manjappa AS, Murumkar PR, Gajaria TK, Devkar RV, Mishra AK, Yadav MR. Drug-fortified liposomes as carriers for sustained release of NSAIDs: The concept and its validation in the animal model for the treatment of arthritis. Eur J Pharm Sci 2018; 125:11-22. [PMID: 30219410 DOI: 10.1016/j.ejps.2018.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/18/2018] [Accepted: 09/11/2018] [Indexed: 02/06/2023]
Abstract
Drug-fortified cationic liposomes of 6‑methoxy‑2‑naphthylacetic acid (6‑MNA) were prepared and characterized by various techniques. The residence time of drug-fortified liposomes in joint cavity was evaluated by intra-articular (IA) administration of the radio-labeled (99mTc) liposomal formulation in the inflamed joints in rats. The cationic liposomal formulation composed of 6‑MNA (3) as an active agent, its double salt (4) with the lipid 1,2‑distearoyl‑sn‑glycero‑3‑phosphoethanolamine (DSPE), and pharmaceutically acceptable excipients such as hydrogenated soyabean phospatidylcholine (HSPC) and 1,2‑dioleyloxy‑3‑trimethylammoniumpropane chloride (DOTAP) were developed using thin film hydration technique. The cryo-TEM analysis confirmed that the prepared optimized liposomal formulation (DFL-2) was a mixture of small unilamellar vesicles (SUVs), large unilamellar vesicles (LUVs) and multilamellar vesicles (MLVs). In addition, the TEM analysis confirmed that the prepared liposomes were of spherical in shape having liposome size in the range of 500-900 nm and zeta potential of about +30 mV. The developed cationic liposomes exhibited sustained release profile of payload of 6‑MNA for over >12 h and about five times higher retention in the inflamed animal joints after 24 h (by scintigraphy of the joints) as compared to the plain 6‑MNA solution when administered by IA route. The anti-inflammatory activity of prepared liposomal composition is evaluated by Freund's adjuvant induced arthritic model in rats. The liposomal formulation was well tolerated by all animals indicating good biocompatibility. Further, the cationic liposomal formulation treated group showed decreased erythrocyte sedimentation rate (ESR), and C-reactive protein (CRP) level in comparison to the control and the standard groups in the in vivo study. The improved efficacy of the drug-fortified liposomal formulation was due to the coupled effect of longer retention and sustained release of the active drug 6‑MNA in the joints. From the obtained results it could be concluded that the combined effect of the cationic charge on the drug-fortified liposomes and the inherent affinity of the active agent towards the synovial joint tissues, coupled with slow release of the active drug due to double salt approach at the site of administration could potentially decrease the frequency of IA drug administration. Hence such a formulation could prove to be a therapeutic boon for the management of late stage arthritis.
Collapse
Affiliation(s)
- Vijay A Pawar
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, India
| | - Arehalli S Manjappa
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, India
| | - Prashant R Murumkar
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, India
| | - Tejal K Gajaria
- Division of Phytotherapeutics and Metabolic Endocrinology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390001, India
| | - Ranjisinh V Devkar
- Division of Phytotherapeutics and Metabolic Endocrinology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara 390001, India
| | - Anil K Mishra
- Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, New Delhi 110054, India
| | - Mange Ram Yadav
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara 390001, India.
| |
Collapse
|
28
|
Zhang X, Shi Y, Zhang Z, Yang Z, Huang G. Intra-articular delivery of tetramethylpyrazine microspheres with enhanced articular cavity retention for treating osteoarthritis. Asian J Pharm Sci 2018; 13:229-238. [PMID: 32104396 PMCID: PMC7032152 DOI: 10.1016/j.ajps.2017.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/25/2017] [Indexed: 01/19/2023] Open
Abstract
Tetramethylpyrazine (TMP) is a traditional Chinese herbal medicine with strong anti-inflammatory and cartilage protection activities, and thus a promising candidate for treating osteoarthritis. However, TMP is rapidly cleared from the joint cavity after intra-articular injection and requires multiple injections to maintain efficacy. The aim of this study was to encapsulate TMP into poly (lactic-co-glycolic acid) (PLGA) microspheres to enhance the TMP retention in the joint, reducing injection frequencies and decreasing dosage. TMP microspheres were prepared by emulsion/solvent evaporation method. The intra-articular retention of the drug was assessed by detecting the drug concentration distributed in the joint tissue at different time points. The therapeutic effect of TMP microspheres was evaluated by the swelling of knee joints and histologic analysis in papain-induced OA rat model. The prepared freeze-dried microspheres with a particle size of about 10 µm can effectively prolong the retention time of the drug in the articular cavity to 30 d, which is 4.7 times that of the TMP solution. Intra-articular injection of TMP microspheres efficiently relieved inflammatory symptoms, improved joint lesions and decreased the depletion of proteoglycan. In conclusion, intra-articular injection of TMP loaded microspheres was a promising therapeutic method in the treatment of OA.
Collapse
Affiliation(s)
| | | | | | | | - Guihua Huang
- Corresponding author. The School of Pharmaceutical Science, Shandong University, 44 Wenhua Xi Road, Ji'nan 250012, Shandong Province, China. Tel.: +86 531 88382015..
| |
Collapse
|
29
|
Janssen M, Timur UT, Woike N, Welting TJM, Draaisma G, Gijbels M, van Rhijn LW, Mihov G, Thies J, Emans PJ. Celecoxib-loaded PEA microspheres as an auto regulatory drug-delivery system after intra-articular injection. J Control Release 2016; 244:30-40. [PMID: 27836707 DOI: 10.1016/j.jconrel.2016.11.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/01/2016] [Accepted: 11/06/2016] [Indexed: 12/20/2022]
Abstract
In this study, we investigated the potential of celecoxib-loaded polyester amide (PEA) microspheres as an auto-regulating drug delivery system for the treatment of pain associated with knee osteoarthritis (OA). Celecoxib release from PEA microspheres and inflammation responsive release of a small molecule from PEA was investigated in vitro. Inflammation responsive release of a small molecule from PEA was observed when PEA was exposed to cell lysates obtained from a neutrophil-like Hl-60 cell line. Following a short initial burst release of ~15% of the total drug load in the first days, celecoxib was slowly released throughout a period of >80days. To investigate biocompatibility and degradation behavior in vivo, celecoxib-loaded PEA microspheres were injected in OA-induced (ACLT+pMMx) or contralateral healthy knee joints of male Lewis rats. Bioactivity of celecoxib from loaded PEA microspheres was confirmed by PGE2 measurements in total rat knee homogenates. Intra-articular biocompatibility was demonstrated histologically, where no cartilage damage or synovial thickening and necrosis were observed after intra-articular injections with PEA microspheres. Degradation of PEA microspheres was significantly higher in OA induced knees compared to contralateral healthy knee joints, while loading the PEA microspheres with celecoxib significantly inhibited degradation, indicating a drug delivery system with auto regulatory behavior. In conclusion, this study suggests the potential of celecoxib-loaded PEA microspheres to be used as a safe drug delivery system with auto regulatory behavior for treatment of pain associated with OA of the knee.
Collapse
Affiliation(s)
- Maarten Janssen
- Department of Orthopaedic Surgery, Research School CAPHRI, Maastricht University Medical Centre, P. Debyelaan, 25, 6229 HX Maastricht, The Netherlands
| | - Ufuk Tan Timur
- Department of Orthopaedic Surgery, Research School CAPHRI, Maastricht University Medical Centre, P. Debyelaan, 25, 6229 HX Maastricht, The Netherlands.
| | - Nina Woike
- DSM Biomedical, Koestraat 1, 6167 RA Geleen, The Netherlands
| | - Tim J M Welting
- Department of Orthopaedic Surgery, Research School CAPHRI, Maastricht University Medical Centre, P. Debyelaan, 25, 6229 HX Maastricht, The Netherlands
| | - Guy Draaisma
- DSM Biomedical, Koestraat 1, 6167 RA Geleen, The Netherlands
| | - Marion Gijbels
- Department of Pathology, Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, The Netherlands; Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Lodewijk W van Rhijn
- Department of Orthopaedic Surgery, Research School CAPHRI, Maastricht University Medical Centre, P. Debyelaan, 25, 6229 HX Maastricht, The Netherlands
| | - George Mihov
- DSM Biomedical, Koestraat 1, 6167 RA Geleen, The Netherlands
| | - Jens Thies
- DSM Biomedical, Koestraat 1, 6167 RA Geleen, The Netherlands
| | - Pieter J Emans
- Department of Orthopaedic Surgery, Research School CAPHRI, Maastricht University Medical Centre, P. Debyelaan, 25, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
30
|
Abd-Allah H, Kamel AO, Sammour OA. Injectable long acting chitosan/tripolyphosphate microspheres for the intra-articular delivery of lornoxicam: Optimization and in vivo evaluation. Carbohydr Polym 2016; 149:263-73. [DOI: 10.1016/j.carbpol.2016.04.096] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/20/2016] [Accepted: 04/22/2016] [Indexed: 12/20/2022]
|
31
|
Kavanaugh TE, Werfel TA, Cho H, Hasty KA, Duvall CL. Particle-based technologies for osteoarthritis detection and therapy. Drug Deliv Transl Res 2016; 6:132-47. [PMID: 25990835 PMCID: PMC4654703 DOI: 10.1007/s13346-015-0234-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Osteoarthritis (OA) is a disease characterized by degradation of joints with the development of painful osteophytes in the surrounding tissues. Currently, there are a limited number of treatments for this disease, and many of these only provide temporary, palliative relief. In this review, we discuss particle-based drug delivery systems that can provide targeted and sustained delivery of imaging and therapeutic agents to OA-affected sites. We focus on technologies such as polymeric micelles and nano-/microparticles, liposomes, and dendrimers for their potential treatment and/or diagnosis of OA. Several promising studies are highlighted, motivating the continued development of delivery technologies to improve treatments for OA.
Collapse
Affiliation(s)
- Taylor E Kavanaugh
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Thomas A Werfel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Hongsik Cho
- University of Tennessee Health Science Center, Memphis, TN, USA
| | - Karen A Hasty
- University of Tennessee Health Science Center, Memphis, TN, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
32
|
Kim C, Jeon OH, Kim DH, Chae JJ, Shores L, Bernstein N, Bhattacharya R, Coburn JM, Yarema KJ, Elisseeff JH. Local delivery of a carbohydrate analog for reducing arthritic inflammation and rebuilding cartilage. Biomaterials 2015; 83:93-101. [PMID: 26773662 DOI: 10.1016/j.biomaterials.2015.12.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/15/2015] [Accepted: 12/28/2015] [Indexed: 11/28/2022]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by articular cartilage degradation. Because OA has a multifactorial nature and complex interrelationship of the individual elements of a whole joint, there is a need for comprehensive therapeutic approaches for cartilage tissue engineering, which simultaneously address multiple aspects of disease etiology. In this work, we investigated a multifunctional carbohydrate-based drug candidate, tri-butanoylated N-acetyl-D-galactosamine analog (3,4,6-O-Bu3GalNAc) that induced cartilage tissue production by human mesenchymal stem cells (hMSCs) and human OA chondrocytes by modulating Wnt/β-catenin signaling activity. The dual effects promoted chondrogenesis of human MSC and reduced inflammation of human OA chondrocytes in in vitro cultures. Translating these findings in vivo, we evaluated therapeutic effect of 3,4,6-O-Bu3GalNAc on the rat model of posttraumatic OA when delivered via local intra-articular sustained-release delivery using microparticles and found this method to be efficacious in preventing OA progression. These results show that 3,4,6-O-Bu3GalNAc, a disease modifying OA drug candidate, has promising therapeutic potential for articular cartilage repair.
Collapse
Affiliation(s)
- Chaekyu Kim
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Ok Hee Jeon
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Do Hun Kim
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - J Jeremy Chae
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Lucas Shores
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Nicholas Bernstein
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Rahul Bhattacharya
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Jeannine M Coburn
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Kevin J Yarema
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Jennifer H Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and the Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA.
| |
Collapse
|
33
|
Singh A, Agarwal R, Diaz-Ruiz CA, Willett NJ, Wang P, Lee LA, Wang Q, Guldberg RE, García AJ. Nanoengineered particles for enhanced intra-articular retention and delivery of proteins. Adv Healthc Mater 2014; 3:1562-7, 1525. [PMID: 24687997 DOI: 10.1002/adhm.201400051] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/02/2014] [Indexed: 12/26/2022]
Abstract
Localized intra-articular delivery of anti-inflammatory proteins can reduce inflammation in osteoarthritis but poses a challenge because of raid clearance within few hours of injection. A new class of polymer is developed that forms self-assembled nanoparticles ranging from 300 to 900 nm and demonstrates particle size dependent prolonged retention in intra-articular joint spaces compared to bolus protein over a period of 14 d.
Collapse
Affiliation(s)
- Ankur Singh
- Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA 30332 USA
- Sibley School of Mechanical and Aerospace Engineering; Cornell University; Ithaca NY 14853 USA
| | - Rachit Agarwal
- Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA 30332 USA
| | - Carlos A. Diaz-Ruiz
- Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA 30332 USA
| | - Nick J. Willett
- Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA 30332 USA
| | - Peiyi Wang
- Department of Chemistry and Biochemistry; University of South Carolina; Columbia SC 29205 USA
| | - L. Andrew Lee
- Department of Chemistry and Biochemistry; University of South Carolina; Columbia SC 29205 USA
- A&Q NanoDesigns, LLC; Columbia SC29201 USA
| | - Qian Wang
- Department of Chemistry and Biochemistry; University of South Carolina; Columbia SC 29205 USA
| | - Robert E. Guldberg
- Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA 30332 USA
- Petit Institute for Bioengineering and Bioscience; Georgia Institute of Technology; Atlanta GA 30332 USA
| | - Andrés J. García
- Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA 30332 USA
- Petit Institute for Bioengineering and Bioscience; Georgia Institute of Technology; Atlanta GA 30332 USA
| |
Collapse
|
34
|
Kang ML, Ko JY, Kim JE, Im GI. Intra-articular delivery of kartogenin-conjugated chitosan nano/microparticles for cartilage regeneration. Biomaterials 2014; 35:9984-9994. [PMID: 25241157 DOI: 10.1016/j.biomaterials.2014.08.042] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 08/29/2014] [Indexed: 12/18/2022]
Abstract
We developed an intra-articular (IA) drug delivery system to treat osteoarthritis (OA) that consisted of kartogenin conjugated chitosan (CHI-KGN). Kartogenin, which promotes the selective differentiation of mesenchymal stem cells (MSCs) into chondrocytes, was conjugated with low-molecular-weight chitosan (LMWCS) and medium-molecular-weight chitosan (MMWCS) by covalent coupling of kartogenin to each chitosan using an ethyl(dimethylaminopropyl) carbodiimide (EDC)/N-hydroxysuccinimide (NHS) catalyst. Nanoparticles (NPs, 150 ± 39 nm) or microparticles (MPs, 1.8 ± 0.54 μm) were fabricated from kartogenin conjugated-LMWCS and -MMWCS, respectively, by an ionic gelation using tripolyphosphate (TPP). The in vitro release profiles of kartogenin from the particles showed sustained release for 7 weeks. When the effects of the CHI-KGN NPs or CHI-KGN MPs were evaluated on the in vitro chondrogenic differentiation of human bone marrow MSCs (hBMMSCs), the CHI-KGN NPs and CHI-KGN MPs induced higher expression of chondrogenic markers from cultured hBMMSCs than unconjugated kartogenin. In particular, hBMMSCs treated with CHI-KGN NPs exhibited more distinct chondrogenic properties in the long-term pellet cultures than those treated with CHI-KGN MPs. The in vivo therapeutic effects of CHI-KGN NPs or CHI-KGN MPs were investigated using a surgically-induced OA model in rats. The CHI-KGN MPs showed longer retention time in the knee joint than the CHI-KGN NPs after IA injection in OA rats. The rats treated with CHI-KGN NPs or CHI-KGN MPs by IA injection showed much less degenerative changes than untreated control or rats treated with unconjugated kartogenin. In conclusion, CHI-KGN NPs or CHI-KGN MPs can be useful polymer-drug conjugates as an IA drug delivery system to treat OA.
Collapse
Affiliation(s)
- Mi Lan Kang
- Department of Orthopedics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| | - Ji-Yun Ko
- Department of Orthopedics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| | - Ji Eun Kim
- Department of Orthopedics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| | - Gun-Il Im
- Department of Orthopedics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea.
| |
Collapse
|
35
|
Drugs and Polymers for Delivery Systems in OA Joints: Clinical Needs and Opportunities. Polymers (Basel) 2014. [DOI: 10.3390/polym6030799] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
36
|
Kang ML, Im GI. Drug delivery systems for intra-articular treatment of osteoarthritis. Expert Opin Drug Deliv 2013; 11:269-82. [PMID: 24308404 DOI: 10.1517/17425247.2014.867325] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Intra-articular (IA) drug delivery is very useful in the treatment of osteoarthritis (OA), the most common chronic joint affliction. However, the therapeutic effect of IA administration depends mostly on the efficacy of drug delivery. AREAS COVERED The present article reviews the current status of IA therapy for OA treatment as well as its rationale. Outlines of drug delivery parameters such as release profile, retention time, distribution, size and transport that influence the drug's biological performance in the joints are summarized. New delivery systems, currently under investigation, including liposome, nanoparticle, microparticle and hydrogel formulations are introduced. Functionalized drug delivery systems by targeting and thermoresponsiveness that are being investigated for OA treatment via IA therapy are also addressed. EXPERT OPINION Several delivery systems, including liposome, microparticles, nanoparticles and hydrogels, have been investigated for the sustained drug delivery to the joints. These can be advanced by the use of functionalized drug delivery systems that can lead targeting to specific regions and thermoresponsiveness for prolonged drug release in the joints. Further advances will bring forth new biocompatible and biodegradable materials as a drug carrier or new combination regimens. Future innovations in this field should be directed toward the development of adapted delivery systems that can induce tissue regeneration in OA patients.
Collapse
Affiliation(s)
- Mi Lan Kang
- Dongguk University Ilsan Hospital, Department of Orthopedics , Goyang 410-773 , Korea +82 31 961 7315 ; +82 31 961 7314 ;
| | | |
Collapse
|
37
|
Feng R, Zhang Z, Li Z, Huang G. Preparation andin vitroevaluation of etoposide-loaded PLGA microspheres for pulmonary drug delivery. Drug Deliv 2013; 21:185-92. [DOI: 10.3109/10717544.2013.840813] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
38
|
Walk RM, Elliott ST, Blanco FC, Snyder JA, Jacobi AM, Rose SD, Behlke MA, Salem AK, Vukmanovic S, Sandler AD. T-cell activation is enhanced by targeting IL-10 cytokine production in toll-like receptor-stimulated macrophages. Immunotargets Ther 2012; 1:13-23. [PMID: 27471682 PMCID: PMC4934151 DOI: 10.2147/itt.s32615] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Toll-like receptor (TLR) agonists represent potentially useful cancer vaccine adjuvants in their ability to stimulate antigen-presenting cells (APCs) and subsequently amplify the cytotoxic T-cell response. The purpose of this study was to characterize APC responses to TLR activation and to determine the subsequent effect on lymphocyte activation. We exposed murine primary bone marrow-derived macrophages to increasing concentrations of agonists to TLRs 2, 3, 4, and 9. This resulted in a dose-dependent increase in production of not only tumor necrosis factor–alpha (TNF-α), a surrogate marker of the proinflammatory response, but also interleukin 10 (IL-10), a well-described inhibitory cytokine. Importantly, IL-10 secretion was not induced by low concentrations of TLR agonists that readily produced TNF-α. We subsequently stimulated lymphocytes with anti-CD3 antibody in the presence of media from macrophages activated with higher doses of TLR agonists and observed suppression of interferon gamma release. Use of both IL-10 knockout macrophages and IL-10 small-interfering RNA (siRNA) ablated this suppressive effect. Finally, IL-10 siRNA was successfully used to suppress CpG-induced IL-10 production in vivo. We conclude that TLR-mediated APC stimulation can induce a paradoxical inhibitory effect on T-cell activation mediated by IL-10.
Collapse
Affiliation(s)
- Ryan M Walk
- Department of Surgery, Walter Reed Army Medical Center, Washington, DC, USA; Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Medical Center, Washington, DC, USA
| | - Steven T Elliott
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Medical Center, Washington, DC, USA
| | - Felix C Blanco
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Medical Center, Washington, DC, USA
| | - Jason A Snyder
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Medical Center, Washington, DC, USA
| | | | - Scott D Rose
- Integrated DNA Technologies, Coralville, IA, USA
| | | | - Aliasger K Salem
- Division of Pharmaceutics, University of Iowa, Iowa City, IA, USA
| | - Stanislav Vukmanovic
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Medical Center, Washington, DC, USA
| | - Anthony D Sandler
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Medical Center, Washington, DC, USA
| |
Collapse
|