1
|
Li J, Jiang H, Tan G, Lv Z, Liu Z, Guo H, Sun Z, Xu X, Shi D. Fibrocartilage hyalinization: A potential therapeutic strategy for articular fibrocartilage. J Orthop Translat 2025; 52:313-324. [PMID: 40421144 PMCID: PMC12104164 DOI: 10.1016/j.jot.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 04/20/2025] [Accepted: 04/23/2025] [Indexed: 05/28/2025] Open
Abstract
Articular fibrocartilage is commonly observed on the joint surface in osteoarthritis (OA) or cartilage injury, often seen as a result of cartilage degeneration. Compared to hyaline cartilage, fibrocartilage exhibits inferior mechanical properties and biological functions, which contribute to further cartilage degeneration and the progression of OA. Despite this, research on cartilage regeneration has not sufficiently addressed the specific challenges and strategies related to fibrocartilage. Although fibrocartilage formation is an unavoidable outcome during cartilage repair, it offers several benefits in the regeneration process, such as providing a natural cell source and establishing a strong integration with surrounding tissues. Recently, a therapeutic approach focused on the in-situ modification of fibrocartilage to promote hyaline cartilage regeneration, referred to as "fibrocartilage hyalinization", has been proposed. Our recent work has demonstrated the feasibility of converting existing fibrocartilage into hyaline cartilage in vivo within the injured area. Key elements of this strategy include modifying the extracellular matrix (ECM), targeting fibrotic chondrocytes, and altering the local microenvironment. This review summarizes the current understanding of articular fibrocartilage's characteristics and mechanisms, while also discussing potential approaches and the feasibility of fibrocartilage hyalinization for cartilage regeneration.
Collapse
Affiliation(s)
- Jiawei Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Department of Orthopedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325200, Zhejiang, PR China
| | - Huiming Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
| | - Guihua Tan
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
| | - Zhongyang Lv
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
| | - Zizheng Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
| | - Hu Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
| | - Ziying Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
| | - Xingquan Xu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Dongquan Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| |
Collapse
|
2
|
Yang J, des Rieux A, Malfanti A. Stimuli-Responsive Nanomedicines for the Treatment of Non-cancer Related Inflammatory Diseases. ACS NANO 2025; 19:15189-15219. [PMID: 40249331 PMCID: PMC12045021 DOI: 10.1021/acsnano.5c00700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Nanomedicines offer a means to overcome the limitations associated with traditional drug dosage formulations by affording drug protection, enhanced drug bioavailability, and targeted drug delivery to affected sites. Inflamed tissues possess unique microenvironmental characteristics (including excessive reactive oxygen species, low pH levels, and hypoxia) that stimuli-responsive nanoparticles can employ as triggers to support on-demand delivery, enhanced accumulation, controlled release, and activation of anti-inflammatory drugs. Stimuli-responsive nanomedicines respond to physicochemical and pathological factors associated with diseased tissues to improve the specificity of drug delivery, overcome multidrug resistance, ensure accurate diagnosis and precision therapy, and control drug release to improve efficacy and safety. Current stimuli-responsive nanoparticles react to intracellular/microenvironmental stimuli such as pH, redox, hypoxia, or specific enzymes and exogenous stimuli such as temperature, magnetic fields, light, and ultrasound via bioresponsive moieties. This review summarizes the general strategies employed to produce stimuli-responsive nanoparticles tailored for inflammatory diseases and all recent advances, reports their applications in drug delivery, and illustrates the progress made toward clinical translation.
Collapse
Affiliation(s)
- Jingjing Yang
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Anne des Rieux
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
- Department
of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
3
|
Joshi N, Yan J, Dang M, Slaughter K, Wang Y, Wu D, Ung T, Bhingaradiya N, Pandya V, Chen MX, Kaur S, Bhagchandani S, Alfassam HA, Joseph J, Gao J, Dewani M, Chu RWC, Yip RCS, Weldon E, Shah P, Pisal ND, Shukla C, Sherman NE, Luo JN, Conway T, Eickhoff JP, Botelho L, Alhasan AH, Karp JM, Ermann J. A mechanically resilient soft hydrogel improves drug delivery for treating post-traumatic osteoarthritis in physically active joints. Proc Natl Acad Sci U S A 2025; 122:e2409729122. [PMID: 40163719 PMCID: PMC12002200 DOI: 10.1073/pnas.2409729122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/12/2025] [Indexed: 04/02/2025] Open
Abstract
Intra-articular delivery of disease-modifying osteoarthritis drugs (DMOADs) is likely to be most effective in the early stages of post-traumatic osteoarthritis (PTOA), when symptoms are minimal, and patients remain physically active. To ensure effective therapy, DMOAD delivery systems therefore must withstand repeated mechanical loading without altering the kinetics of drug release. While soft materials are typically preferred for DMOAD delivery, mechanical loading can compromise their structural integrity and disrupt controlled drug release. In this study, we present a mechanically resilient soft hydrogel that rapidly self-heals under conditions simulating human running while maintaining sustained release of the cathepsin-K inhibitor L-006235, used as a proof-of-concept DMOAD. This hydrogel demonstrated superior performance compared to a previously reported hydrogel designed for intra-articular drug delivery, which, in our study, neither recovered its structure nor maintained drug release under mechanical loading. When injected into mouse knee joints, the hydrogel provided consistent release kinetics of the encapsulated drug in both treadmill-running and nonrunning mice. In a mouse model of severe PTOA exacerbated by treadmill-running, the L-006235 hydrogel significantly reduced cartilage degeneration, whereas the free drug did not. Overall, our data underscore the hydrogel's potential for treating PTOA in physically active patients.
Collapse
Affiliation(s)
- Nitin Joshi
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Jing Yan
- Harvard Medical School, Boston, MA02115
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Mickael Dang
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Kai Slaughter
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Yufeng Wang
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Dana Wu
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Trevor Ung
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Nutan Bhingaradiya
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Virja Pandya
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Mu Xian Chen
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Shahdeep Kaur
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Sachin Bhagchandani
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Haya A. Alfassam
- National Center for Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh11442, Saudi Arabia
- King Abdulaziz City for Science and Technology Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, Riyadh11442, Saudi Arabia
| | - John Joseph
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Jingjing Gao
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Mahima Dewani
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Rachel Wai Chun Chu
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Ryan Chak Sang Yip
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Eli Weldon
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Purna Shah
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Nishkal Dhiraj Pisal
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Chetan Shukla
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Nicholas E. Sherman
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - James N. Luo
- Harvard Medical School, Boston, MA02115
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA02115
| | - Thomas Conway
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | | | | | - Ali H. Alhasan
- National Center for Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh11442, Saudi Arabia
- King Abdulaziz City for Science and Technology Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, Riyadh11442, Saudi Arabia
| | - Jeffrey M. Karp
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
- Broad Institute, Cambridge, MA02142
- Harvard Stem Cell Institute, Cambridge, MA02138
| | - Joerg Ermann
- Harvard Medical School, Boston, MA02115
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| |
Collapse
|
4
|
Kim TT, Malu D, He D, Hu Y, Kim J. Development of Bioorthogonally Degradable Tough Hydrogels Using Enamine N-Oxide Based Crosslinkers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414692. [PMID: 40018818 PMCID: PMC11962699 DOI: 10.1002/adma.202414692] [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] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/20/2025] [Indexed: 03/01/2025]
Abstract
Inducibly degradable polymers present new opportunities to integrate tough hydrogels into a wide range of biomaterials. Rapid and inducible degradation enables fast transition in material properties without sacrificing material integrity prior to removal. In pursuit of bioorthogonal chemical modalities that will enable inducible polymer degradation in biologically relevant environments, enamine N-oxide crosslinkers are developed for double network acrylamide-based polymer/alginate hydrogels. Bioorthogonal dissociation initiated by the application of aqueous diboron solution through several delivery mechanisms effectively lead to polymer degradation. Their degradation by aqueous B2(OH)4 solution results in a fracture energy half-life of <10 min. The biocompatibility of the degradable hydrogels and B2(OH)4 reagent is assessed, and the removability of strongly adhered tough hydrogels on mice skin is evaluated. Thermoresponsive PNiPAAm/Alg hydrogels are fabricated and application of the hydrogels as a chemically inducible degradable intraoral wound dressing is demonstrated. It is demonstrated through in vivo maximum tolerated dose studies that diboron solution administered to mice by oral gavage is well tolerated. Successful integration of enamine N-oxides within the tough double network hydrogels as chemically degradable motifs demonstrates the applicability of enamine N-oxides in the realm of polymer chemistry and highlights the importance of chemically induced bioorthogonal dissociation reactions for materials science.
Collapse
Affiliation(s)
- Thomas T. Kim
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGA30332USA
- Department of Cancer BiologyDana‐Farber Cancer InstituteBostonMA02215USA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMA02115USA
| | - Deep Malu
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Dongjing He
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Yuhang Hu
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Justin Kim
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGA30332USA
- Department of Cancer BiologyDana‐Farber Cancer InstituteBostonMA02215USA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMA02115USA
| |
Collapse
|
5
|
Kuddushi M, Vithalani H, Singh H, Dave H, Jain A, Pal A, Kumar S, Bhatia Z, Seshadri S, Dhanka M. Easily Injectable, Organic Solvent-Free Self-Assembled Hydrogel Platform for Endoscope Mediated Gastrointestinal Polypectomy. Adv Healthc Mater 2025; 14:e2403915. [PMID: 39988843 DOI: 10.1002/adhm.202403915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/31/2025] [Indexed: 02/25/2025]
Abstract
Endoscopic submucosal dissection (ESD) and endoscopic mucosal resection (ESR) are used to eliminate tiny, flat lesions in the gastrointestinal tract (GIT). A substantial submucosal cushion is required for effective dissection. Commonly used saline and hypertonic dextrose injections disperse quickly and do not offer significant elevation, whereas polymers such as gelatin and alginate are challenging to inject. In this study, a novel amphiphilic polyglycerol stearate-based hydrogel (PGSH) platform is demonstrated which could be administered via an endoscopic catheter to help create a stable submucosal elevation. PGSH is easy to inject across different needle gauges, shear-thinning, and forms a long-lasting submucosal cushion during ESD. This hydrogel can encapsulate hydrophilic drugs such as streptomycin, allowing controlled enzymatic and nonenzymatic release. Ex-vivo experiments on goat's GIT demonstrate that PGSH is smoothly injectable without clogging the catheter's needle, achieving the necessary submucosal elevation. Furthermore, ex-vivo blood studies demonstrate immediate clotting behavior while maintaining hemocompatibility. In-vivo, investigations in mice show that the hydrogel forms a biocompatible cushion of suitable height with a nontoxic organ profile that does not overexpress inflammatory cytokines. ESD studies in the porcine model suggest that PGSH has the potential to significantly improve treatment outcomes in the early endoscopic removal of gastrointestinal polyps.
Collapse
Affiliation(s)
- Muzammil Kuddushi
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, 382355, India
| | - Hitasha Vithalani
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, 382355, India
| | - Hemant Singh
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, 382355, India
| | - Harshil Dave
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, 382355, India
| | | | - Ankit Pal
- Muljibhai Patel Urological Hospital, Nadiad, Gujarat, India
| | - Sunny Kumar
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Zeel Bhatia
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Sriram Seshadri
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Mukesh Dhanka
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, 382355, India
| |
Collapse
|
6
|
Kumar A, Rahul, Kanika, Kumar J, Mahajan S, Ali A, Ali N, Bishnoi M, Son YO, Khan R. Multifunctional chrysin-loaded gallic acid-glycerol monostearate conjugate-based injectable hydrogel for targeted inhibition of hypoxia-induced NLRP3 inflammasome in ulcerative colitis. Biomater Sci 2025; 13:1801-1817. [PMID: 39998176 DOI: 10.1039/d4bm01700e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory condition affecting the colon part of the large intestine. Since there is no cure for this disease, conventional therapies only provide symptomatic relief. Recently, phytomolecules have shown promising treatment results in various diseases. However, short half-life, hydrophobicity, and poor bioavailability limit their therapeutic potential. To overcome all these challenges, we have earlier conjugated a phytomolecule (gallic acid) (GA) with the FDA-approved generally recognized as safe (GRAS) material that is glycerol monostearate (GMS). This GA-GMS conjugate self-assembles as a hydrogel via the heating-cooling method and acts as a pro-drug of GA. The in vivo imaging results suggest that the GA-GMS hydrogel more efficiently adheres to the inflamed colon than a therapeutic enema. Additionally, it is known that the gut microbiota exaggerates UC by creating a hypoxic environment in the colon. This hypoxia is linked with NLRP3 inflammasome activation that triggers the release of IL-1β and IL-18 that downregulates MUC2 protein expression in the colon, responsible for mucin secretion in the colon. Therefore, chrysin (CR) (HIF-1α inhibitor) is encapsulated into the GA-GMS hydrogel to target hypoxia. The CR@GA-GMS hydrogel follows the enzyme-responsive release of the CR and restores DSS-induced damage to colonic tissue. Furthermore, the CR@GA-GMS hydrogel downregulates HIF-1α mediated NLRP3 inflammasome signalling while upregulating MUC2 production.
Collapse
Affiliation(s)
- Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| | - Rahul
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| | - Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| | - Jattin Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| | - Shubham Mahajan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| | - Aneesh Ali
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| | - Nemat Ali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute, Mohali, Punjab 140306, India
| | - Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, 63243, Republic of Korea
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar) 140306, Punjab, India.
| |
Collapse
|
7
|
Kumar A, Rahul, Kanika, Kumar J, Ahmad A, Ali A, Kumar B, Mahajan S, Ali N, Khan R. Engineered Drug-Amphiphile Conjugate Nanoparticles for Targeted Inhibition of AQP4-Mediated NLRP3 Inflammasome Signaling in Collagen-Induced Rheumatoid Arthritis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16590-16601. [PMID: 40038599 DOI: 10.1021/acsami.4c20973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Aquaporins (AQPs) are transmembrane proteins that transport water, small solutes, and molecules across cell membranes. Studies have reported the role of AQPs in the activation, migration, and proliferation of immune cells, thus modulating the pathogenesis of autoimmune disease. In joints, the enhanced AQP4 expression exaggerates pathological changes like hydrarthrosis, acidosis, and hyperosmotic stress-inducing dysfunction of the articular chondrocytes, leading to articular cartilage destruction in collagen-induced arthritis (CIA). Acetazolamide (AZM), a sulfonamide carbonic anhydrase inhibitor of AQP4, reversibly decreases water permeability through AQP4 and is a potential molecule for targeting AQP4 in the CIA. However, its low solubility and low bioavailability limit its therapeutic effectiveness. Therefore, in this study, we have synthesized a polyphenol drug (gallic acid) (GA) and an amphiphile (glycerol monostearate) (GMS) conjugate to self-assemble into nanoparticles and encapsulated with AZM. Apart from AZM, GA is known for its antioxidant and anti-inflammatory properties. Therefore, intra-articular injection of AZM@GA-GMS NPs efficiently downregulates the expression of AQP4 and associated NLRP3 inflammasome activation. Moreover, the NPs are cytocompatible and showed enzyme-responsive drug release and thus offer a promising therapeutic strategy for RA by inhibiting AQP4-mediated inflammatory pathways. This opens up an avenue for treatment for RA.
Collapse
MESH Headings
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
- Animals
- Nanoparticles/chemistry
- Inflammasomes/metabolism
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/chemically induced
- Arthritis, Rheumatoid/pathology
- Aquaporin 4/metabolism
- Aquaporin 4/antagonists & inhibitors
- Mice
- Acetazolamide/chemistry
- Acetazolamide/pharmacology
- Acetazolamide/therapeutic use
- Signal Transduction/drug effects
- Humans
- Collagen
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/pathology
- Male
Collapse
Affiliation(s)
- Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar), Punjab 140306, India
| | - Rahul
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar), Punjab 140306, India
| | - Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar), Punjab 140306, India
| | - Jattin Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar), Punjab 140306, India
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Aneesh Ali
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar), Punjab 140306, India
| | - Bhuvnesh Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar), Punjab 140306, India
| | - Shubham Mahajan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar), Punjab 140306, India
| | - Nemat Ali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali (SAS Nagar), Punjab 140306, India
| |
Collapse
|
8
|
Lin S, Cui T, Jiang Y, Xie J, Zhong D, Jiang J, Deng D, Zhao M, Xue C, Gan S, Qiu J, Wang X. Microenvironment-responsive NIR-IIb multifunctional nanozyme platform for bacterial imaging and specialized anti-anaerobic bacteria periodontal therapy. J Nanobiotechnology 2025; 23:189. [PMID: 40055819 PMCID: PMC11889803 DOI: 10.1186/s12951-025-03270-9] [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: 12/30/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025] Open
Abstract
Periodontitis is a chronic inflammatory disease caused by plaque. In order to remove pathogens and promote tissue repair, the following steps need to be taken simultaneously: localizing the diseased area, improving the anaerobic microenvironment, as well as addressing the anti-inflammatory and osteogenic needs. This study aims to address these issues by developing a responsive near-infrared-IIb nanozyme system (DMUP), assembled from lanthanide-doped down-converted nanoparticles and multi-enzymatically active nanozyme. DMUP binds to bacterial membranes via the bacterial targeting peptide ubiquicidin29-41 (UBI29-41). Upon responding to the inflammatory microenvironment, it releases manganese (Mn) nanozyme and paeonol (Pae), and localized infected areas by fluorescent bacterial imaging in the near-infrared IIb (NIR-IIb) region. In particular, the released Mn nanozyme reacts with hydrogen peroxide in the inflammatory microenvironment to generate oxygen (O2) in situ, thereby improving the anoxic environment to inhibit anaerobic bacteria. On the other hand, as a metal oxide nanozyme, Mn nanozyme scavenges reactive oxygen species (ROS) by mimicking the cascade process of superoxide dismutase and catalase. The phenolic antioxidant Pae shifts macrophages from pro-inflammatory (M1-type) to anti-inflammatory (M2-type) through the Akt/mTOR pathway. It can synergize with Mn nanozyme to regulate the inflammatory microenvironment, thereby reducing inflammation, promoting osteogenic genes expression, and accelerating periodontal tissues regeneration.
Collapse
Affiliation(s)
- Suai Lin
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
- Jiangxi Provincial Key Laboratory of Oral Diseases, Department of Stomatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Tiehan Cui
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
- Jiangxi Provincial Key Laboratory of Oral Diseases, Department of Stomatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Yuxin Jiang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China
| | - Jialiang Xie
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China
| | - Da Zhong
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China
| | - Junkai Jiang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China
| | - Dan Deng
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China
| | - Mengzhen Zhao
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China
| | - Chengzhou Xue
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China
| | - Shiyu Gan
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China
| | - Jiaxuan Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China.
- Jiangxi Provincial Key Laboratory of Oral Diseases, Department of Stomatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China.
| | - Xiaolei Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China.
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi, 330088, P. R. China.
| |
Collapse
|
9
|
Wei Z, Zhu M, Morin N, Wollsten D, Hirvonen J, Yang X, Santos HA, Li W. Polymeric Microspheres with High Mass Fraction of Therapeutics Enabled by the Manipulation of Kinetics Factor During Emulsion Droplet Solidification. ADVANCED FUNCTIONAL MATERIALS 2025; 35. [DOI: 10.1002/adfm.202417307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Indexed: 03/01/2025]
Abstract
AbstractHigh drug‐loaded polymeric microspheres hold promise in biomedical fields due to reduced excipient administration, minimized side effects, and enhanced therapeutical efficacy. Although thermodynamic factors like drug‐carrier material compatibility are well‐known to influence the drug loading capacity of microspheres, they fail to explain the huge difference in drug loading degree observed for polymers and drugs with similar interactions. Here, based on the droplet microfluidic platform, the single droplet solidification process is investigated. The results indicated that amorphous polymers can hinder drug diffusion during droplet solidification compared to crystalline polymers, resulting in a higher drug loading degree. Next, this principle is applied to improve the drug loading capability of crystalline polymers (polycaprolactone (PCL) and poly(L‐lactide) (PLLA)) by random co‐polymerization (poly(caprolactone‐co‐L‐lactide) (PCL‐PLLA)), achieving 6.2–22.2 times increased drug loading degree. Moreover, PCL‐PLLA microspheres with a high content of indomethacin exhibited superior therapeutical efficacy in the treatment of gout arthritis. Overall, these results offer insights into the impact of polymer crystallization on droplet solidification kinetics, which consequently affects the drug loading capacity. These findings provide guidelines for the development of polymers for efficient drug encapsulation.
Collapse
Affiliation(s)
- Zhenyang Wei
- National Engineering Research Center for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology Wuhan 430074 China
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki 00014 Finland
| | - Mingyu Zhu
- National Engineering Research Center for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology Wuhan 430074 China
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki 00014 Finland
| | - Nicolas Morin
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki 00014 Finland
| | - Daniela Wollsten
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki 00014 Finland
| | - Jouni Hirvonen
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki 00014 Finland
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology Wuhan 430074 China
| | - Hélder A. Santos
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki 00014 Finland
- Department of Biomaterials and Biomedical Technology The Personalized Medicine Research Institute (PRECISION) University Medical Center Groningen (UMCG) University of Groningen Ant. Deusinglaan 1 Groningen 9713 AV The Netherlands
| | - Wei Li
- National Engineering Research Center for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology Wuhan 430074 China
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki 00014 Finland
| |
Collapse
|
10
|
Dave H, Vithalani H, Singh H, Yadav I, Jain A, Pal A, Patidar N, Navale A, Dhanka M. Amphiphilic Gelator-Based Shear-Thinning Hydrogel for Minimally Invasive Delivery via Endoscopy Catheter to Remove Gastrointestinal Polyps. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405508. [PMID: 39506390 DOI: 10.1002/smll.202405508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/04/2024] [Indexed: 11/08/2024]
Abstract
Injectable polymeric hydrogels delivered via endoscopic catheter have emerged as promising submucosal agents, offering durable, long-lasting cushions to enhance the efficacy of endoscopic submucosal dissection (ESD) for the removal of small, flat polyps from the gastrointestinal tract (GIT). However, polymer-based injections do not meet the easy-injectability criteria via catheter because their high viscosity tends to clog the catheter needle. To the best of knowledge, for the first time, report the fabrication of an amphiphile-based small molecule hydrogel of diglycerol monostearate (DGMS) that self-assembles to form hydrogel (DGMSH) for delivery via an endoscopic catheter. Physicochemical characterization of the hydrogel reveals its fibrous morphology, shear-thinning behaviour, and easy injectability, along with its scalability and long shelf-life (6 months). Ex vivo studies on the goat's stomach and intestine demonstrate the ease of injectability through the catheters and the development of visible submucosal cushion depots with the desired height. Moreover, the hydrogel can encapsulate both hydrophobic and hydrophilic drugs/dyes. In vivo studies in small animals have found that the hydrogel depot is durable, biocompatible, non-immunogenic, and has a hemostatic effect. Endoscopic studies in the porcine model demonstrate a safe injection and endoscopic excision of GI polyps acting as a suitable agent for ESD.
Collapse
Affiliation(s)
- Harshil Dave
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gandhinagar, Gujarat, 382355, India
| | - Hitasha Vithalani
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gandhinagar, Gujarat, 382355, India
| | - Hemant Singh
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gandhinagar, Gujarat, 382355, India
| | - Indu Yadav
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gandhinagar, Gujarat, 382355, India
| | - Abhinav Jain
- Gastro1 Hospital, Ahmedabad, Gujarat, 380060, India
| | - Ankit Pal
- Muljibhai Patel Urological Hospital, Nadiad, Gujarat, 38700, India
| | - Nishant Patidar
- Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, 391760, India
| | - Archana Navale
- Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, 391760, India
| | - Mukesh Dhanka
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gandhinagar, Gujarat, 382355, India
| |
Collapse
|
11
|
Chetty K, Peters XQ, Omolo CA, Ismail EA, Gafar MA, Elhassan E, Kassam SZF, Govender J, Dlamini S, Govender T. Multifunctional Dual Enzyme-Responsive Nanostructured Lipid Carriers for Targeting and Enhancing the Treatment of Bacterial Infections. ACS APPLIED BIO MATERIALS 2025; 8:548-569. [PMID: 39714140 DOI: 10.1021/acsabm.4c01436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Bacterial infections pose an increasingly worrisome threat to the health of humankind, with antibiotic resistance contributing significantly to this burden. With current conventional antibiotics perpetuating the problem, and a paucity in developing antibiotics, drug delivery systems incorporating nanotechnology appear promising. As such, a dual enzyme-responsive multifunctional nanostructured lipid carrier (NLC) incorporating farnesol (FAN) and triglycerol monostearate (TGMS), was conceptualized for the codelivery of vancomycin (VCM) and antimicrobial peptide (AMP) to enhance the antibacterial activity of VCM. In silico studies and Microscale Thermophoresis demonstrated the strong binding relationships between the NLC constituents and two enzymes that exist in higher concentrations during host infection, namely lipase and a matrix metalloproteinase (MMP). The formulated nanosystem, VCM-AMP-TF-NLCs, had a particle size, polydispersity index, zeta potential, and entrapment efficiency of 149.00 ± 2.97 nm, 0.07 ± 0.01, -5.51 ± 1.21 mV, and 86.20% ± 1.47%, respectively. The NLCs, which showed stability, and biocompatibility, also demonstrated lipase- and MMP-responsiveness. The in vitro antibacterial studies revealed 2-fold and 8-fold reductions in the minimum inhibitory concentration for the NLCs compared to bare VCM, against methicillin-resistant Staphylococcal aureus (MRSA) and Escherichia coli, respectively. Furthermore, in vivo studies revealed that tissues treated with the VCM-AMP-TF-NLCs displayed significantly reduced bacterial burdens (up to 8.73-fold) and less histopathological cellular injury, edema, and necrosis compared to the tissues treated with bare VCM alone. The results support the superiority of the VCM-AMP-TF-NLCs as a multifunctional dual enzyme-responsive NLC compared to bare VCM.
Collapse
Affiliation(s)
- Kerisha Chetty
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4300, South Africa
| | - Xylia Q Peters
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4300, South Africa
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4300, South Africa
- Department of Pharmaceutics, School of Pharmacy and Health Sciences, United States International University-Africa, P.O. Box 14634, Nairobi 00800, Kenya
| | - Eman A Ismail
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4300, South Africa
| | - Mohammed A Gafar
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4300, South Africa
| | - Eman Elhassan
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4300, South Africa
| | - Sania Z F Kassam
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4300, South Africa
| | - Jasoda Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4300, South Africa
| | - Sbongumusa Dlamini
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4300, South Africa
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4300, South Africa
| |
Collapse
|
12
|
Thangadurai M, Sethuraman S, Subramanian A. Drug Delivery Approaches for Rheumatoid Arthritis: Recent Advances and Clinical Translation Aspects. Crit Rev Ther Drug Carrier Syst 2025; 42:1-54. [PMID: 40084516 DOI: 10.1615/critrevtherdrugcarriersyst.v42.i3.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Rheumatoid arthritis (RA) is a multifactorial autoimmune disease characterized with symmetrical progression of joint deformity that is often diagnosed at a chronic condition with other associated pathological conditions such as pericarditis, keratitis, pulmonary granuloma. Despite the understanding of RA pathophysiology in disease progression, current clinical treatment options such as disease-modifying anti-rheumatic drugs (DMARDs), biologics, steroids, and non-steroidal anti-inflammatory drugs (NSAIDs) provide only palliative therapy while causing adverse side effects such as off-target multi-organ toxicity and risk of infections. Further, available drug delivery strategies to treat RA pathogenicity does not successfully reach the site of action due to various barriers such as phagocytosis and first pass effect in addition to the disease complexity and unknown etiology, thereby leading to the development of irreversible joint dysfunction. Therefore, novel and effective strategies remain an unmet need to control the disease progression and to maintain the balance between pro- and anti-inflammatory cytokines. This review provides a comprehensive outlook on the RA pathophysiology and its corresponding disease progression. Contributions of synoviocytes such as macrophages, fibroblast-like cells in increasing invasiveness to exacerbate joint damage is also outlined in this review, which could be a potential future therapeutic target to complement the existing treatment regimens in controlling RA pathogenesis. Further, various smart drug delivery approaches under research to achieve maximum therapeutic efficacy with minimal adverse side effects have been discussed, which in turn emphasize the unmet challenges and future perspectives in addressing RA complications.
Collapse
Affiliation(s)
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Laboratory, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| | - Anuradha Subramanian
- Tissue Engineering & Additive Manufacturing (TEAM) Laboratory, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| |
Collapse
|
13
|
Wang Q, Ren J, Lin X, Zhang B, Li J, Weng Y. Inflammatory stimulus-responsive polymersomes reprogramming glucose metabolism mitigates rheumatoid arthritis. Biomaterials 2025; 312:122760. [PMID: 39163825 DOI: 10.1016/j.biomaterials.2024.122760] [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: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/22/2024]
Abstract
Inflammation-resident cells within arthritic sites undergo a metabolic shift towards glycolysis, which greatly aggravates rheumatoid arthritis (RA). Reprogramming glucose metabolism can suppress abnormal proliferation and activation of inflammation-related cells without affecting normal cells, holding potential for RA therapy. Single 2-deoxy-d-glucose (2-DG, glycolysis inhibitor) treatment often cause elevated ROS, which is detrimental to RA remission. The rational combination of glycolysis inhibition with anti-inflammatory intervention might cooperatively achieve favorable RA therapy. To improve drug bioavailability and exert synergetic effect, stable co-encapsulation of drugs in long circulation and timely drug release in inflamed milieu is highly desirable. Herein, we designed a stimulus-responsive hyaluronic acid-triglycerol monostearate polymersomes (HTDD) co-delivering 2-DG and dexamethasone (Dex) to arthritic sites. After intravenous injection, HTDD polymersomes facilitated prolonged circulation and preferential distribution in inflamed sites, where overexpressed matrix metalloproteinases and acidic pH triggered drug release. Results indicated 2-DG can inhibit the excessive cell proliferation and activation, and improve Dex bioavailability by reducing Dex efflux. Dex can suppress inflammatory signaling and prevent 2-DG-induced oxidative stress. Thus, the combinational strategy ultimately mitigated RA by inhibiting glycolysis and hindering inflammatory signaling. Our study demonstrated the great potential in RA therapy by reprogramming glucose metabolism in arthritic sites.
Collapse
Affiliation(s)
- Qin Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Jianheng Ren
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xin Lin
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Bin Zhang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jiao Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yajun Weng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
14
|
Nosrati Z, Chen YA, Bergamo M, Rodríguez‐Rodríguez C, Chan J, Shojania K, Kherani RB, Chin C, Kelsall JT, Dehghan N, Colwill AM, Collins D, Saatchi K, Häfeli UO. Prodrug Nanomedicine for Synovium Targeted Therapy of Inflammatory Arthritis: Insights from Animal Model and Human Synovial Joint Fluid. Adv Healthc Mater 2024; 13:e2401936. [PMID: 39380387 PMCID: PMC11616258 DOI: 10.1002/adhm.202401936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/29/2024] [Indexed: 10/10/2024]
Abstract
Many patients cannot tolerate low-dose weekly methotrexate (MTX) therapy for inflammatory arthritis treatment due to life-threatening toxicity. Although biologics offer a target-specific therapy, it raises the risk of serious infections and even cancer due to immune system suppression. We introduce an anti-inflammatory arthritis MTX ester prodrug using a long-circulating biocompatible polymeric macromolecule: folic acid (FA) functionalized hyperbranched polyglycerol (HPG). In vitro the drug MTX is incrementally released through pH and enzymatic degradation over 2 weeks. The role of matrix metalloproteinases (MMPs) in site-specific prodrug activation was verified using synovial fluid (SF) of 26 rheumatology patients and 4 healthy controls. Elevated levels of specific MMPs-markers of joint inflammation-positively correlated with enhanced prodrug release explained by acid-catalyzed hydrolysis of esters by proteases. Intravenously administered 111In-radiolabeled prodrug confirmed by SPECT/CT imaging that it accumulated preferentially in inflamed joints while reducing off-target side-effects in a mouse model of rheumatoid arthritis (RA). Added FA as a targeting vector prolonged prodrug action; prodrug with 4x less MTX applied every 2 weeks was as effective as weekly MTX therapy. The preclinical results suggest a prodrug-based strategy for the treatment of inflammatory joint diseases, with potential for other chronic inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Zeynab Nosrati
- Faculty of Pharmaceutical SciencesUniversity of British Columbia2405 Wesbrook MallVancouverBCV5Z 3P2Canada
| | - Yun An Chen
- Faculty of Pharmaceutical SciencesUniversity of British Columbia2405 Wesbrook MallVancouverBCV5Z 3P2Canada
| | - Marta Bergamo
- Faculty of Pharmaceutical SciencesUniversity of British Columbia2405 Wesbrook MallVancouverBCV5Z 3P2Canada
| | | | - Jonathan Chan
- Department of Medicine – RheumatologyUniversity of British Columbia2775 Laurel StVancouverBCV5Z 1M9Canada
| | - Kam Shojania
- Department of Medicine – RheumatologyUniversity of British Columbia2775 Laurel StVancouverBCV5Z 1M9Canada
| | - Raheem B. Kherani
- Department of Medicine – RheumatologyUniversity of British Columbia2775 Laurel StVancouverBCV5Z 1M9Canada
| | - Carson Chin
- Burnaby Medical and Surgical SpecialistsBurnabyBCV3J 1M2Canada
| | - John T. Kelsall
- Department of Medicine – RheumatologyUniversity of British Columbia2775 Laurel StVancouverBCV5Z 1M9Canada
| | | | | | - David Collins
- Department of Medicine – RheumatologyUniversity of British Columbia2775 Laurel StVancouverBCV5Z 1M9Canada
| | - Katayoun Saatchi
- Faculty of Pharmaceutical SciencesUniversity of British Columbia2405 Wesbrook MallVancouverBCV5Z 3P2Canada
| | - Urs O. Häfeli
- Faculty of Pharmaceutical SciencesUniversity of British Columbia2405 Wesbrook MallVancouverBCV5Z 3P2Canada
- Department of PharmacyFaculty of Health and Medical SciencesUniversity of CopenhagenUniversitetsparken 2CopenhagenDenmark2100
| |
Collapse
|
15
|
Ye Q, Zhang M, Li S, Liu W, Xu C, Li Y, Xie R. Controlled Stimulus-Responsive Delivery Systems for Osteoarthritis Treatment. Int J Mol Sci 2024; 25:11799. [PMID: 39519350 PMCID: PMC11545989 DOI: 10.3390/ijms252111799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Osteoarthritis (OA), a common and disabling degenerative joint disease, affects millions of people worldwide and imposes a considerable burden on patients and society due to its high prevalence and economic costs. The pathogenesis of OA is closely related to the progressive degradation of articular cartilage and the accompany inflammation; however, articular cartilage itself cannot heal and modulate the inflammation due to the lack of nerves, blood vessels, and lymph-vessels. Therefore, reliable and effective methods to treat OA remain highly desired. Local administration of drugs or bioactive materials by intra-articular injection of the delivery system represents a promising approach to treat OA, especially considering the prolonged joint retention, cartilage or chondrocytes targeting, and stimuli-responsive release to achieve precision OA therapy. This article summarizes and discusses the advances in the currently used delivery systems (nanoparticle, hydrogel, liposome, and microsphere) and then focuses on their applications in OA treatment from the perspective of endogenous stimulus (redox reactions, pH, enzymes, and temperature) and exogenous stimulus (near-infrared, magnetic, and ultrasound)-responsive release. Finally, the challenges and potential future directions for the development of nano-delivery systems are summarized.
Collapse
Affiliation(s)
- Qianwen Ye
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Mingshuo Zhang
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Shuyue Li
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Wenyue Liu
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Chunming Xu
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yumei Li
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Renjian Xie
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
16
|
Xu M, Fu T, Zhang C, An Z, Yan J, Lu Z, Wu H, Liu J, Qiu L, Shi L, Lin J, Cao Y, Pei R. Prolonged, staged, and self-regulated methotrexate release coupled with ROS scavenging in an injectable hydrogel for rheumatoid arthritis therapy. J Control Release 2024; 375:60-73. [PMID: 39216600 DOI: 10.1016/j.jconrel.2024.08.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Rheumatoid arthritis (RA) remains a formidable healthcare challenge due to its chronic nature and potential for irreversible joint damage. Methotrexate (MTX) is a cornerstone treatment for RA but carries significant risks of adverse effects with repeated administration, necessitating the exploration of alternative delivery methods. Injectable hydrogels loaded with MTX for intra-articular injection present a promising solution, allowing sustained drug release directly into affected joints. However, current hydrogel systems often lack extended therapeutic periods and the ability to self-regulate drug release according to disease state. Furthermore, RA is associated with excessive production of reactive oxygen species (ROS), which exacerbates inflammation and joint damage. Herein, we developed an advanced injectable hydrogel (MPDANPs/MTX HA-PEG gel) based on "bio-orthogonal chemistry", combining hyaluronic acid and polyethylene glycol (PEG) matrices co-loaded with mesoporous polydopamine nanoparticles (MPDANPs) and MTX. MPDANPs/MTX HA-PEG gel achieved prolonged, staged, and self-regulated MTX release, coupled with ROS scavenging capabilities for enhanced therapeutic efficacy. Due to its optimized MTX release behavior and significant ROS scavenging function, MPDANPs/MTX HA-PEG gel exhibited potent anti-inflammatory effects in collagen-induced arthritis (CIA) rats following a single intra-articular injection. Our findings highlight the potential of MPDANPs/MTX HA-PEG gel as a highly effective treatment strategy for RA, offering a promising avenue for improving patient outcomes.
Collapse
Affiliation(s)
- Mingsheng Xu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Tingting Fu
- Department of Orthopaedics, The Fourth Affiliated of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215001, China
| | - Chenhui Zhang
- Department of Orthopaedics, The Fourth Affiliated of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215001, China
| | - Zhen An
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jincong Yan
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zhongzhong Lu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Hanfei Wu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jihuan Liu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Lei Qiu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Lei Shi
- Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan 215300, China
| | - Jun Lin
- Department of Orthopaedics, The Fourth Affiliated of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215001, China.
| | - Yi Cao
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; Jiangxi Institute of Nanotechnology, Nanchang 330200, China.
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| |
Collapse
|
17
|
Zhang XL, Yue YX, Yang Y, Ying AK, Ma R, Chen J, Chen FY, Hou XY, Pan YC, Ren DZ, Yang T, Li ZQ, Guo DS. A single molecule carrier for ocular posterior segment diseases. J Control Release 2024:S0168-3659(24)00725-9. [PMID: 39490420 DOI: 10.1016/j.jconrel.2024.10.052] [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: 06/24/2024] [Revised: 10/01/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Eye drops are envisaged as the most promising non-invasive formulation for the treatment of the ocular posterior segment diseases, while it is hindered by a series of complex ocular barriers, both static and dynamic in nature. In this context, we propose a single molecule nanomedicine based on host-guest chemistry to achieve highly efficient drug delivery targeted to ocular posterior segment. Sulfonated azocalix[4]arene (SAC4A) serves as the single molecule carrier, owing the multiple features of small size (24.0 Å in length, 21.2 Å in width, 14.8 Å in height with a Van der Waals volume of 930 Å3), negative charge, hydrophilicity, loading universality and hypoxia-triggered release. As a proof-of-concept, an eye drop formed by the complexation of SAC4A with sunitinib (SUN) is prepared to treat wet age-related macular degeneration (wAMD). SAC4A successfully transports SUN to the ocular posterior segment (the amount of SUN reaching the retinal-choroid tissue in the SUN@SAC4A group was 2.47 times larger than that in the SUN group at 30 min), significantly enhancing its anti-choroidal neoangiogenesis effect of SUN to wAMD, which played a key role in the treatment. We believe that the single molecule nanomedicine paradigm is highly amenable for treating various ocular posterior segment diseases in the future.
Collapse
Affiliation(s)
- Xiao-Ling Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China
| | - Yu-Xin Yue
- Tianjin Eye Hospital, College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Yang Yang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China
| | - An-Kang Ying
- Tianjin Eye Hospital, College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Rong Ma
- Tianjin Eye Hospital, College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Jie Chen
- Tianjin Eye Hospital, College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Fang-Yuan Chen
- Tianjin Eye Hospital, College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Xiao-Yun Hou
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China
| | - Yu-Chen Pan
- Tianjin Eye Hospital, College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China
| | - Da-Zhuang Ren
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China
| | - Tao Yang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China
| | - Zhi-Qing Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300392, China.
| | - Dong-Sheng Guo
- Tianjin Eye Hospital, College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China.
| |
Collapse
|
18
|
Han J, Sheng T, Zhang Y, Cheng H, Gao J, Yu J, Gu Z. Bioresponsive Immunotherapeutic Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2209778. [PMID: 36639983 DOI: 10.1002/adma.202209778] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/31/2022] [Indexed: 06/17/2023]
Abstract
The human immune system is an interaction network of biological processes, and its dysfunction is closely associated with a wide array of diseases, such as cancer, infectious diseases, tissue damage, and autoimmune diseases. Manipulation of the immune response network in a desired and controlled fashion has been regarded as a promising strategy for maximizing immunotherapeutic efficacy and minimizing side effects. Integration of "smart" bioresponsive materials with immunoactive agents including small molecules, biomacromolecules, and cells can achieve on-demand release of agents at targeted sites to reduce overdose-related toxicity and alleviate off-target effects. This review highlights the design principles of bioresponsive immunotherapeutic materials and discusses the critical roles of controlled release of immunoactive agents from bioresponsive materials in recruiting, housing, and manipulating immune cells for evoking desired immune responses. Challenges and future directions from the perspective of clinical translation are also discussed.
Collapse
Affiliation(s)
- Jinpeng Han
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Tao Sheng
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuqi Zhang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hao Cheng
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Jianqing Gao
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Jicheng Yu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Zhen Gu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
19
|
Fei Y, Li X, Lv Z, Liu Z, Xie Y, Chen J, Li W, Liu X, Guo H, Liu H, Zhang Z, Wang X, Fan J, Hu C, Jin X, Jiang R, Xu N, Xia J, Li Y, Shi D. Promoting chondrogenesis by targeted delivery to the degenerating cartilage in early treatment of osteoarthritis. Bioact Mater 2024; 40:624-633. [PMID: 39247402 PMCID: PMC11377143 DOI: 10.1016/j.bioactmat.2024.08.004] [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: 04/08/2024] [Revised: 07/21/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Osteoarthritis (OA) is a highly incident total joint degenerative disease with cartilage degeneration as the primary pathogenesis. The cartilage matrix is mainly composed of collagen, a matrix protein with a hallmark triple-helix structure, which unfolds with collagen degradation on the cartilage surface. A collagen hybridizing peptide (CHP) is a synthetic peptide that binds the denatured collagen triple helix, conferring a potential disease-targeting possibility for early-stage OA. Here, we constructed an albumin nanoparticle (An) conjugated with CHP, loaded with a chondrogenesis-promoting small molecule drug, kartogenin (KGN). The CHP-KGN-An particle exhibited sustained release of KGN in vitro and prolonged in vivo retention selectively within the degenerated cartilage in the knee joints of model mice with early-stage OA. Compared to treatment with KGN alone, CHP-KGN-An robustly attenuated cartilage degradation, synovitis, osteophyte formation, and subchondral bone sclerosis in OA model mice and exhibited a more prominent effect on physical activity improvement and pain alleviation. Our study showcases that targeting the degenerated cartilage by collagen hybridization can remarkably promote the efficacy of small molecule drugs and may provide a novel delivery strategy for early-stage OA therapeutics.
Collapse
Affiliation(s)
- Yuxiang Fei
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xiaojing Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, PR China
| | - Zhongyang Lv
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China
| | - Zizheng Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Ya Xie
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Jiaqi Chen
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Weitong Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xiyu Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Hu Guo
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Huan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Zhaofeng Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xunhao Wang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Jingjing Fan
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Chunqing Hu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xiaoyu Jin
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Ruiyang Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Nuo Xu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR China
| | - Yang Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, PR China
| | - Dongquan Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| |
Collapse
|
20
|
Ali A, Rahul, Jori C, Kumar J, Kumar A, Kanika, Ansari MM, Ahmad A, Ali N, Yadav P, Parvez S, Navik U, Son YO, Khan R. Sinapic acid-pullulan based inflammation responsive nanomicelles for the local treatment of experimental inflammatory arthritis. Int J Biol Macromol 2024; 278:134903. [PMID: 39168211 DOI: 10.1016/j.ijbiomac.2024.134903] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disorder of joints. It is one of the major causes of disability and morbidity worldwide. Administration of conventional drugs through the systemic route restricts the bioavailability of drugs, systemic toxicity, and reduced efficacy. We have introduced Rebamipide (Reb)-loaded Sinapic acid (SA)-Pullulan (PL) nanomicelles (Reb@SA-PL NMs), a nanotechnology based drug delivery system for the treatment of inflammatory arthritis. PL is a polysaccharide obtained from the fungus Aureobasidium pullulans, and SA is a bioactive polyphenol found in various plants. Both are classified by US-FDA Generally Recognised as Safe (GRAS) materials. Reb@SA-PL NMs found to be cytocompatible. Subsequently, intra-articular administration of Reb@SA-PL NMs enhances the anti-arthritic potential compared to free Reb drug in collagen-induced experimental inflammatory arthritis rat model. Reb@SA-PL NMs reduced the expression of RANKL receptor and Nf-κB. Reb@SA-PL NMs reverses the breakdown of type II collagen, MMP-13, and inhibits the pro-inflammatory markers. Reb@SA-PL NMs prevented bone erosion, cartilage degradation, joint oedema, and synovial inflammation. The results of the study demonstrated that Reb@SA-PL NMs, an enzyme-responsive drug delivery system, has excellent potential for alleviating inflammatory arthritis by blocking MMP-13 and RANKL.
Collapse
Affiliation(s)
- Aneesh Ali
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali 140306, Punjab, India
| | - Rahul
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali 140306, Punjab, India
| | - Chandrashekhar Jori
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali 140306, Punjab, India
| | - Jattin Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali 140306, Punjab, India
| | - Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali 140306, Punjab, India
| | - Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali 140306, Punjab, India
| | - Md Meraj Ansari
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju Special Self-Governing Province, 63243, South Korea. Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Republic of Korea; Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110 062, India
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC), Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, Foothills Medical Centre, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Nemat Ali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110 062, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India
| | - Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju Special Self-Governing Province, 63243, South Korea. Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Republic of Korea; Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali 140306, Punjab, India.
| |
Collapse
|
21
|
Gai Y, Zhou H, Yang Y, Chen J, Chi B, Li P, Yin Y, Wang Y, Li J. Injectable body temperature responsive hydrogel for encephalitis treatment via sustained release of nano-anti-inflammatory agents. BIOMATERIALS TRANSLATIONAL 2024; 5:300-313. [PMID: 39734706 PMCID: PMC11681188 DOI: 10.12336/biomatertransl.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/10/2024] [Accepted: 08/30/2024] [Indexed: 12/31/2024]
Abstract
Skull defects are common in the clinical practice of neurosurgery, and they are easily complicated by encephalitis, which seriously threatens the life and health safety of patients. The treatment of encephalitis is not only to save the patient but also to benefit the society. Based on the advantages of injectable hydrogels such as minimally invasive surgery, self-adaptation to irregularly shaped defects, and easy loading and delivery of nanomedicines, an injectable hydrogel that can be crosslinked in situ at the ambient temperature of the brain for the treatment of encephalitis caused by cranial defects is developed. The hydrogel is uniformly loaded with nanodrugs formed by cationic liposomes and small molecule drugs dexmedetomidine hydrochloride (DEX-HCl), which can directly act on the meninges to achieve sustained release delivery of anti-inflammatory nanodrug preparations and achieve the goal of long-term anti-inflammation at cranial defects. This is the first time that DEX-HCl has been applied within this therapeutic system, which is innovative. Furthermore, this study is expected to alleviate the long-term suffering of patients, improve the clinical medication strategies for anti-inflammatory treatment, promote the development of new materials for cranial defect repair, and expedite the translation of research outcomes into clinical practice.
Collapse
Affiliation(s)
- Yuqi Gai
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Huaijuan Zhou
- Advanced Research Institute of Multidisciplinary Sciences, Beijing Institute of Technology, Beijing, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology (BIT), Zhuhai, Guangdong Province, China
| | - Yingting Yang
- Advanced Research Institute of Multidisciplinary Sciences, Beijing Institute of Technology, Beijing, China
| | - Jiatian Chen
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Bowen Chi
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Pei Li
- Center for Advanced Biotechnology & Medicine, Rutgers University, Piscataway, NJ, USA
| | - Yue Yin
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology (BIT), Zhuhai, Guangdong Province, China
| |
Collapse
|
22
|
Shamiya Y, Chakraborty A, Zahid AA, Bainbridge N, Guan J, Feng B, Pjontek D, Chakrabarti S, Paul A. Ascorbyl palmitate nanofiber-reinforced hydrogels for drug delivery in soft issues. COMMUNICATIONS MATERIALS 2024; 5:197. [PMID: 39309138 PMCID: PMC11415299 DOI: 10.1038/s43246-024-00641-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
Nanofiber-based hydrogel delivery systems have recently shown great potential in biomedical applications, specifically due to their high surface-to-volume ratio of ultra-fine nanofibers and their ability to carry low solubility drugs. Herein, we introduce a visible light-triggered in situ-gelling drug vehicle (GAP Gel) composed of ascorbyl palmitate (AP) nanofibers and gelatin methacryloyl polymer. AP nanofibers form self-assembled structures through intermolecular interactions with a hydrophobic drug-loading core. We demonstrate that the hydrophilic periphery of AP nanofibers allows them to interact with other hydrophilic molecules via hydrogen bonds. The presence of AP nanofibers significantly enhances the viscoelasticity of GAP Gel in a concentration-dependent manner. Further, GAP Gel shows in vitro biocompatibility and sustained drug delivery efficacy when loaded with a hydrophobic antibiotic. Likewise, GAP Gel shows excellent in vivo biocompatibility when implanted in immunocompetent mice in various forms. Lastly, GAP Gels maintain cell viability when cultured in a 3D-environment over 7 days, establishing it as a promising and versatile hydrogel platform for the delivery of biotherapeutics.
Collapse
Affiliation(s)
- Yasmeen Shamiya
- Department of Chemistry, The University of Western Ontario, London, ON Canada
| | - Aishik Chakraborty
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON Canada
- Collaborative Specialization in Muscoskeletal Health Research and Bone and Joint Institute, The University of Western Ontario, London, ON Canada
| | - Alap Ali Zahid
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON Canada
| | - Nicholas Bainbridge
- Department of Chemistry, The University of Western Ontario, London, ON Canada
| | - Jingyuan Guan
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON Canada
| | - Biao Feng
- Department of Pathology and Laboratory Medicine, The University of Western Ontario, London, ON Canada
| | - Dominic Pjontek
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON Canada
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, The University of Western Ontario, London, ON Canada
| | - Arghya Paul
- Department of Chemistry, The University of Western Ontario, London, ON Canada
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON Canada
| |
Collapse
|
23
|
Kim KM, D'Elia AM, Rodell CB. Hydrogel-based approaches to target hypersensitivity mechanisms underlying autoimmune disease. Adv Drug Deliv Rev 2024; 212:115395. [PMID: 39004347 DOI: 10.1016/j.addr.2024.115395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/23/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
A robust adaptive immune response is essential for combatting pathogens. In the wrong context such as due to genetic and environmental factors, however, the same mechanisms crucial for self-preservation can lead to a loss of self-tolerance. Resulting autoimmunity manifests in the development of a host of organ-specific or systemic autoimmune diseases, hallmarked by aberrant immune responses and tissue damage. The prevalence of autoimmune diseases is on the rise, medical management of which focuses primarily on pharmacological immunosuppression that places patients at a risk of side effects, including opportunistic infections and tumorigenesis. Biomaterial-based drug delivery systems confer many opportunities to address challenges associated with conventional disease management. Hydrogels, in particular, can protect encapsulated cargo (drug or cell therapeutics) from the host environment, afford their presentation in a controlled manner, and can be tailored to respond to disease conditions or support treatment via multiplexed functionality. Moreover, localized delivery to affected sites by these approaches has the potential to concentrate drug action at the site, reduce off-target exposure, and enhance patient compliance by reducing the need for frequent administration. Despite their many benefits for the management of autoimmune disease, such biomaterial-based approaches focus largely on the downstream effects of hypersensitivity mechanisms and have a limited capacity to eradicate the disease. In contrast, direct targeting of mechanisms of hypersensitivity reactions uniquely enables prophylaxis or the arrest of disease progression by mitigating the basis of autoimmunity. One promising approach is to induce self-antigen-specific tolerance, which specifically subdues damaging autoreactivity while otherwise retaining the normal immune responses. In this review, we will discuss hydrogel-based systems for the treatment of autoimmune disease, with a focus on those that target hypersensitivity mechanisms head-on. As the field continues to advance, it will expand the range of therapeutic choices for people coping with autoimmune diseases, providing fresh prospects for better clinical outcomes and improved quality of life.
Collapse
Affiliation(s)
- Kenneth M Kim
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA.
| | - Arielle M D'Elia
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA.
| | - Christopher B Rodell
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA; School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Ma Y, Liu Y, Guo J, Chen Z, Zhao Z, Zheng J. Topical application of daphnetin hydrogel for traumatic brain injury. Front Neurosci 2024; 18:1450072. [PMID: 39170676 PMCID: PMC11335657 DOI: 10.3389/fnins.2024.1450072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
Background Traumatic brain injury (TBI) causes neuronal cell damage and dysfunction. According to previous studies, daphnetin (Dap) has a protective effect in neurological injury. However, the in vivo bioavailability of daphnetin is not high. The purpose of this study was to determine whether administering daphnetin directly into the site of injury via a hydrogel drug carrier could improve its therapeutic impact. Methods Tripolycerol monostearates / daphnetin (TM/Dap) hydrogels were prepared and characterised using water bath heating, scanning electron microscopy (SEM) and small animal in vivo imaging techniques. The TBI model was established using the Feeney free fall impact method. Using the Morris water maze test, the mNSS neurological deficit rating scale, haematoxylin-eosin staining, and liver and kidney function tests, the therapeutic benefit of TM/Dap and its toxic side effects were assessed. The therapeutic effects of TM/Dap were further investigated using wet and dry gravimetric methods, Evans blue staining, protein immunoblotting, immunofluorescence staining techniques and ELISA. Results The efficacy of the TM/Dap hydrogel in gradually releasing daphnetin in the context of traumatic brain damage was shown by both in vitro and in vivo tests. Behavioral experiments showed that the learning and spatial memory abilities of TM/Dap hydrogel treated mice were significantly improved in the water maze experiment. And TM/Dap hydrogel has high biosafety for organisms. The results of the therapeutic mechanism of action showed that TM/Dap hydrogel showed more significant efficacy in reducing the neuroinflammatory response caused by TNF-α, IL-6 and other factors, as well as promoting the recovery of post-traumatic neurological function. Conclusion The use of hydrogel as a drug carrier for daphnetin showed more significant efficacy in reducing neuroinflammatory response, protecting nerve tissue and promoting post-traumatic neurological recovery compared with traditional drug delivery methods.
Collapse
Affiliation(s)
- Yuanhao Ma
- Department of Neurosurgery, Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, China
- Department of Neurosurgery, Huzhou Central Hospital, Huzhou, China
- Xuzhou Medical University, Xuzhou, China
| | - Yu Liu
- Department of Neurosurgery, Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, China
- Xuzhou Medical University, Xuzhou, China
| | - Jianqiang Guo
- Department of Neurosurgery, Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, China
- Xuzhou Medical University, Xuzhou, China
| | - Zhongjun Chen
- Department of Neurosurgery, Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, China
| | - Zongren Zhao
- Department of Neurosurgery, Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, China
| | - Jinyu Zheng
- Department of Neurosurgery, Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, China
| |
Collapse
|
25
|
Varpe P, Joga R, Aglave G, Vasu P, Yerram S, Bellapu KK, Srivastava S, Kumar S. Esterase responsive release of anti-cancer agents from conjugated lipid nanocarrier and the regulatory considerations. Pharm Pat Anal 2024; 13:31-43. [PMID: 39324857 PMCID: PMC11449025 DOI: 10.1080/20468954.2024.2347796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/20/2024] [Indexed: 09/27/2024]
Abstract
The release of active agents in tumors rather than normal tissues, limits systemic exposure and toxicities. Targeting over-expressed esterase enzyme in the tumor microenvironment can selectively release immune-active agents like Programmed Death-1 (PD-1) and PD-1 ligand inhibitors from ester-sensitive lipid nanocarriers, offering a novel approach compared with conventional therapies. PD-1 and PD-L1 association cause T-cell inactivation, whereas blocking their association improves their cytotoxic mechanism. The patent application US2022/0080051-A1 discloses a novel immune-active agent conjugated with lipid to form a nanocarrier for esterase-sensitive release. These nanocarriers selectively enter leaky vasculature of tumors through enhanced permeability and retention effect, undergo ester cleavage to release agents, and are reported to increase bioavailability by 24 times. Further, with other agents or alone it achieves targeted synergistic cancer therapy. Also, the current patent spotlight delves into the crucial formulation considerations necessary for obtaining successful approval of lipidic nano products from relevant regulatory authorities.
Collapse
Affiliation(s)
- Priya Varpe
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Ramesh Joga
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Gayatri Aglave
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Pavan Vasu
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Sravani Yerram
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Kiran Kumar Bellapu
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana , 500037, India
| | - Sandeep Kumar
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
- Department of Pharmaceutics, Nims Institute of Pharmacy, Nims University Rajasthan, Jaipur, Rajasthan 303121, India
| |
Collapse
|
26
|
Ibrahiem B, Shamma R, Salama A, Refai H. Magnetic targeting of lornoxicam/SPION bilosomes loaded in a thermosensitive in situ hydrogel system for the management of osteoarthritis: Optimization, in vitro, ex vivo, and in vivo studies in rat model via modulation of RANKL/OPG. Drug Deliv Transl Res 2024; 14:1982-2002. [PMID: 38158473 PMCID: PMC11153292 DOI: 10.1007/s13346-023-01503-8] [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] [Accepted: 12/17/2023] [Indexed: 01/03/2024]
Abstract
Osteoarthritis is a bone and joint condition characterized pathologically by articular cartilage degenerative damage and can develop into a devastating and permanently disabling disorder. This investigation aimed to formulate the anti-inflammatory drug lornoxicam (LOR) into bile salt-enriched vesicles loaded in an in situ forming hydrogel as a potential local treatment of osteoarthritis. This was achieved by formulating LOR-loaded bilosomes that are also loaded with superparamagnetic iron oxide nanoparticles (SPIONs) for intra-muscular (IM) administration to improve joint targeting and localization by applying an external magnet to the joint. A 31.22 full factorial design was employed to develop the bilosomal dispersions and the optimized formula including SPION (LSB) was loaded into a thermosensitive hydrogel. Moreover, in vivo evaluation revealed that the IM administration of LSB combined with the application of an external magnet to the joint reversed carrageen-induced suppression in motor activity and osteoprotegerin by significantly reducing the elevations in mitogen-activated protein kinases, extracellular signal-regulated kinase, and receptor activator of nuclear factor kappa beta/osteoprotegerin expressions. In addition, the histopathological evaluation of knee joint tissues showed a remarkable improvement in the injured joint tissues. The results proved that the developed LSB could be a promising IM drug delivery system for osteoarthritis management.
Collapse
Affiliation(s)
- Basma Ibrahiem
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, 12566, Egypt
| | - Rehab Shamma
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, El-Kasr El-Aini Street, Cairo, 11562, Egypt
| | - Abeer Salama
- Department of Pharmacology, National Research Centre (NRC), Giza, 12622, Egypt
| | - Hanan Refai
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, 12566, Egypt.
| |
Collapse
|
27
|
Ali A, Jori C, Kanika, Kumar A, Vyawahare A, Kumar J, Kumar B, Ahmad A, Fareed M, Ali N, Navik U, Khan R. A bioactive and biodegradable vitamin C stearate-based injectable hydrogel alleviates experimental inflammatory arthritis. Biomater Sci 2024; 12:3389-3400. [PMID: 38804911 DOI: 10.1039/d4bm00243a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory joint disorder affecting nearly 1% of the global population. In RA, synovial joints are infiltrated by inflammatory mediators and enzymes, leading to articular cartilage deterioration, joint damage, and bone erosion. Herein, the 9-aminoacridine-6-O-stearoyl-L-ascorbic acid hydrogel (9AA-SAA hydrogel) was formulated by the heat-cool method and further characterized for surface charge, surface morphology, rheology, and cytocompatibility. Furthermore, we evaluated the therapeutic efficacy of the 9AA-SAA hydrogel, an enzyme-responsive drug delivery system with on-and-off switching capabilities based on disease severity against collagen-induced experimental arthritis in Wistar rats. The anti-inflammatory action of the US FDA-approved drug 9-aminoacridine (9AA) was revealed which acted through nuclear receptor subfamily 4 group A member 1 (NR4A1), an anti-inflammatory orphan nuclear receptor that inhibits nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB). Furthermore, we have explored the role of ascorbic acid, an active moiety of 6-O-stearoyl-L-ascorbic acid (SAA), in promoting the production of collagen production through ten-eleven translocation-2 (TET2) upregulation. Targeting through NR4A1 and TET2 could be the probable mechanism for the treatment of experimental arthritis. The combination of 9AA and ascorbic acid demonstrated enhanced therapeutic efficacy in the 9AA-SAA hydrogel, significantly reducing the severity of experimental arthritis. This approach, in contrast to existing treatments with limited effectiveness, presents a promising and more effective strategy for RA treatment by mitigating inflammation in experimental arthritis.
Collapse
Affiliation(s)
- Aneesh Ali
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Chandrashekhar Jori
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Akshay Vyawahare
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Jattin Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Bhuvnesh Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC), Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, Foothills Medical Centre, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Mohammad Fareed
- Environmental Health and Clinical Epidemiology Laboratory, Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, India
| | - Nemat Ali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| |
Collapse
|
28
|
Roy HS, Murugesan P, Kulkarni C, Arora M, Nagar GK, Guha R, Chattopadhyay N, Ghosh D. On-demand release of a selective MMP-13 blocker from an enzyme-responsive injectable hydrogel protects cartilage from degenerative progression in osteoarthritis. J Mater Chem B 2024; 12:5325-5338. [PMID: 38669084 DOI: 10.1039/d3tb02871b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
In osteoarthritis (OA), the degradation of cartilage is primarily driven by matrix metalloprotease-13 (MMP-13). Hence, the inhibition of MMP-13 has emerged as an attractive target for OA treatment. Among the various approaches that are being explored for MMP-13 regulation, blocking of the enzyme with specific binding molecules appears to be a more promising strategy for preventing cartilage degeneration. To enhance effectiveness and ensure patient compliance, it is preferable for the binding molecule to exhibit sustained activity when administered directly into the joint. Herein, we present an enzyme-responsive hydrogel that was designed to exhibit on-demand, the sustained release of BI-4394, a potent and highly selective MMP-13 blocker. The stable and compatible hydrogel was prepared using triglycerol monostearate. The efficacy of the hydrogel to prevent cartilage damage was assessed in a rat model of OA induced by anterior cruciate ligament transection (ACLT). The results revealed that in comparison to the rats administrated weekly with intra-articular BI-4394, the hydrogel implanted rats had reduced levels of inflammation and bone erosion. In comparison to untreated control, the cartilage in animals administered with BI-4394/hydrogel exhibited significant levels of collagen-2 and aggrecan along with reduced MMP-13. Overall, this study confirmed the potential of BI-4394 delivery using an enzyme-responsive hydrogel as a promising treatment option to treat the early stages of OA by preventing further cartilage degradation.
Collapse
Affiliation(s)
- Himadri Shekhar Roy
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Preethi Murugesan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Chirag Kulkarni
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Malika Arora
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Geet Kumar Nagar
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Rajdeep Guha
- Division of Laboratory Animal Facility, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Deepa Ghosh
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| |
Collapse
|
29
|
Haloi P, Choudhary R, Lokesh BS, Konkimalla VB. Dual drug nanoparticle synergistically induced apoptosis, suppressed inflammation, and protected autophagic response in rheumatoid arthritis fibroblast-like synoviocytes. Immunol Lett 2024; 267:106854. [PMID: 38537719 DOI: 10.1016/j.imlet.2024.106854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 03/08/2024] [Accepted: 03/23/2024] [Indexed: 04/05/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic immune-mediated joint inflammatory disorder associated with aberrant activation of fibroblast-like synoviocytes (FLS). Recently, FLS gained importance due to its crucial role in RA pathogenesis, and thus, targeting FLS is suggested as an attractive treatment strategy for RA. FLS-targeted approaches may be combined with disease-modifying antirheumatic drugs (DMARDs) and natural phytochemicals to improve efficacy in RA control and negate immunosuppression. In this study, we assessed the therapeutic effectiveness of DD NP HG in primary RA-FLS cells isolated from the synovial tissue of FCA-induced RA rats. We observed that DD NP HG had good biosafety for healthy FLS cells and, at higher concentrations, a mild inhibitory effect on RA-FLS. The combination therapy (DD NP HG) of MTX NP and PEITC NE in RA-FLS showed a higher rate of apoptosis with significantly reduced LPS-induced expression of pro-inflammatory cytokines (TNF-α, IL-17A, and IL-6) in arthritic FLS. Further, the gene expression studies showed that DD NP HG significantly down-regulated the mRNA expression of IL-1β, RANKL, NFATc1, DKK1, Bcl-xl, Mcl-1, Atg12, and ULK1, and up-regulated the mRNA expression of OPG, PUMA, NOXA and SQSTM1 in LPS-stimulated RA-FLS cells. Collectively, our results demonstrated that DD NP HG significantly inhibited the RA-FLS proliferation via inducing apoptosis, down-regulating pro-inflammatory cytokines, and further enhancing the expression of genes associated with bone destruction in RA pathogenesis. A nanotechnology approach is a promising strategy for the co-delivery of dual drugs to regulate the RA-FLS function and achieve synergistic treatment of RA.
Collapse
Affiliation(s)
- Prakash Haloi
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Jatni, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Rajat Choudhary
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Jatni, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - B Siva Lokesh
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Jatni, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - V Badireenath Konkimalla
- School of Biological Sciences, National Institute of Science Education and Research, HBNI, Jatni, Odisha 752050, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India.
| |
Collapse
|
30
|
Joshi N, Yan J, Dang M, Slaughter K, Wang Y, Wu D, Ung T, Pandya V, Chen MX, Kaur S, Bhagchandani S, Alfassam HA, Joseph J, Gao J, Dewani M, Yip RCS, Weldon E, Shah P, Shukla C, Sherman NE, Luo JN, Conway T, Eickhoff JP, Botelho L, Alhasan AH, Karp JM, Ermann J. A Mechanically Resilient Soft Hydrogel Improves Drug Delivery for Treating Post-Traumatic Osteoarthritis in Physically Active Joints. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.16.594611. [PMID: 38826308 PMCID: PMC11142096 DOI: 10.1101/2024.05.16.594611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Intra-articular delivery of disease-modifying osteoarthritis drugs (DMOADs) is likely to be most effective in early post-traumatic osteoarthritis (PTOA) when symptoms are minimal and patients are physically active. DMOAD delivery systems therefore must withstand repeated mechanical loading without affecting the drug release kinetics. Although soft materials are preferred for DMOAD delivery, mechanical loading can compromise their structural integrity and disrupt drug release. Here, we report a mechanically resilient soft hydrogel that rapidly self-heals under conditions resembling human running while maintaining sustained release of the cathepsin-K inhibitor L-006235 used as a proof-of-concept DMOAD. Notably, this hydrogel outperformed a previously reported hydrogel designed for intra-articular drug delivery, used as a control in our study, which neither recovered nor maintained drug release under mechanical loading. Upon injection into mouse knee joints, the hydrogel showed consistent release kinetics of the encapsulated agent in both treadmill-running and non-running mice. In a mouse model of aggressive PTOA exacerbated by treadmill running, L-006235 hydrogel markedly reduced cartilage degeneration. To our knowledge, this is the first hydrogel proven to withstand human running conditions and enable sustained DMOAD delivery in physically active joints, and the first study demonstrating reduced disease progression in a severe PTOA model under rigorous physical activity, highlighting the hydrogel's potential for PTOA treatment in active patients.
Collapse
|
31
|
Qu W, Tian R, Yang B, Guo T, Wu Z, Li Y, Geng Z, Wang Z. Dual-Channel/Localization Single-Molecule Fluorescence Probe for Monitoring ATP and HOCl in Early Diagnosis and Therapy of Rheumatoid Arthritis. Anal Chem 2024; 96:5428-5436. [PMID: 38551643 DOI: 10.1021/acs.analchem.3c05342] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Rheumatoid arthritis (RA), a common chronic inflammatory illness, is still incurable, reducing the sufferers' quality of life significantly. Adenosine 5'-triphosphate (ATP) and hypochlorous acid (HOCl) are key indicators in RA, but their precise mechanisms in RA pathophysiology are unknown. As a result, in order to detect ATP and HOCl simultaneously, we created two new dual-channel/localization single-molecule fluorescence probes, RhTNMB and RhFNMB. Furthermore, RhFNMB outperformed RhTNMB in terms of detection performance. ATP and HOCl produce independent fluorescence responses in the light red channel (λex = 520 nm, λem = 586 nm) and deep red channel (λex = 620 nm, λem = 688 nm), respectively, without spectral crosstalk. It should be noted that the probe RhFNMB successfully imaged ATP in mitochondria and HOCl in cells. Surprisingly, the probe RhFNMB demonstrated remarkable detection ability in the diagnosis and treatment of Pseudomonas aeruginosa-induced abdominal inflammation in mice. We continued to apply the probe RhFNMB to track ATP and HOCl in RA and discovered that ATP and HOCl concentrations were considerably greater in RA joints than in normal joints. We also confirmed the therapeutic effect of methotrexate on RA. This study is the first to achieve dual-channel imaging of ATP and HOCl, which is of great value for the early diagnosis and therapy of RA.
Collapse
Affiliation(s)
- Wangbo Qu
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210023, P. R. China
| | - Ruowei Tian
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210023, P. R. China
| | - Bin Yang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210023, P. R. China
| | - Taiyu Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210023, P. R. China
| | - Zhou Wu
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210023, P. R. China
| | - Yong Li
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210023, P. R. China
| | - Zhirong Geng
- College of Pharmacy, Jiangsu Joint International Laboratory of Animal-Derived Chinese Medicine and Functional Peptides, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Zhilin Wang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
32
|
Kennedy O, Kitson A, Okpara C, Chow LW, Gonzalez-Fernandez T. Immunomodulatory Strategies for Cartilage Regeneration in Osteoarthritis. Tissue Eng Part A 2024; 30:259-271. [PMID: 38126327 DOI: 10.1089/ten.tea.2023.0255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent musculoskeletal disorder and a leading cause of disability globally. Although many efforts have been made to treat this condition, current tissue engineering (TE) and regenerative medicine strategies fail to address the inflammatory tissue environment that leads to the rapid progression of the disease and prevents cartilage tissue formation. First, this review addresses in detail the current anti-inflammatory therapies for OA with a special emphasis on pharmacological approaches, gene therapy, and mesenchymal stromal cell (MSC) intra-articular administration, and discusses the reasons behind the limited clinical success of these approaches at enabling cartilage regeneration. Then, we analyze the state-of-the-art TE strategies and how they can be improved by incorporating immunomodulatory capabilities such as the optimization of biomaterial composition, porosity and geometry, and the loading of anti-inflammatory molecules within an engineered structure. Finally, the review discusses the future directions for the new generation of TE strategies for OA treatment, specifically focusing on the spatiotemporal modulation of anti-inflammatory agent presentation to allow for tailored patient-specific therapies. Impact statement Osteoarthritis (OA) is a prevalent and debilitating musculoskeletal disorder affecting millions worldwide. Despite significant advancements in regenerative medicine and tissue engineering (TE), mitigating inflammation while simultaneously promoting cartilage tissue regeneration in OA remains elusive. In this review article, we discuss current anti-inflammatory therapies and explore their potential synergy with cutting-edge cartilage TE strategies, with a special focus on novel spatiotemporal and patient-specific anti-inflammatory strategies.
Collapse
Affiliation(s)
- Orlaith Kennedy
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
- Department of Biomedical Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
| | - Andrew Kitson
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Chiebuka Okpara
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Lesley W Chow
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | | |
Collapse
|
33
|
Zhang Y, Kang X, Li J, Song J, Li X, Li W, Qi J. Inflammation-Responsive Nanoagents for Activatable Photoacoustic Molecular Imaging and Tandem Therapies in Rheumatoid Arthritis. ACS NANO 2024; 18:2231-2249. [PMID: 38189230 DOI: 10.1021/acsnano.3c09870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Rheumatoid arthritis (RA) severely lowers the life quality by progressively destructing joint functions and eventually causing permanent disability, representing a pressing public health concern. The pathogenesis of RA includes the excessive production of proinflammatory cytokines and harmful oxygen-derived free radicals, such as nitric oxide (NO), which constitute vital targets for precise diagnosis and effective treatment of RA. In this study, we introduce an advanced nanoagent that integrates the RA microenvironment-activatable photoacoustic (PA) imaging with multitarget synergistic treatment for RA. A highly sensitive organic probe with NO-tunable energy transformation and molecular geometry is developed, which enables strong near-infrared absorption with a turn-on PA signal, and the active intramolecular motion could further boost PA conversion. The probe is coassembled with an inflammation-responsive prodrug to construct the theranostic nanoagent, on which a macrophage-derived cell membrane with natural tropism to the inflammatory sites is camouflaged to improve the targeting ability to inflamed joints. The nanoagent could not only sensitively detect RA and differentiate the severity but also efficiently alleviate RA symptoms and improve joint function. The combination of activatable probe-mediated NO scavenging and on-demand activation of anti-inflammatory prodrug significantly inhibits the proinflammatory factors and promotes macrophage repolarization from M1 to M2 phenotype. This meticulously designed nanoagent ingeniously integrates RA-specific PA molecular imaging with synergistic multitarget therapy, rendering tremendous promise for precise intervention of RA-related diseases.
Collapse
Affiliation(s)
- Yuan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaoying Kang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jia Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jianwen Song
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xueping Li
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Wen Li
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Ji Qi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
34
|
Yu H, Gao R, Liu Y, Fu L, Zhou J, Li L. Stimulus-Responsive Hydrogels as Drug Delivery Systems for Inflammation Targeted Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306152. [PMID: 37985923 PMCID: PMC10767459 DOI: 10.1002/advs.202306152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/19/2023] [Indexed: 11/22/2023]
Abstract
Deregulated inflammations induced by various factors are one of the most common diseases in people's daily life, while severe inflammation can even lead to death. Thus, the efficient treatment of inflammation has always been the hot topic in the research of medicine. In the past decades, as a potential biomaterial, stimuli-responsive hydrogels have been a focus of attention for the inflammation treatment due to their excellent biocompatibility and design flexibility. Recently, thanks to the rapid development of nanotechnology and material science, more and more efforts have been made to develop safer, more personal and more effective hydrogels for the therapy of some frequent but tough inflammations such as sepsis, rheumatoid arthritis, osteoarthritis, periodontitis, and ulcerative colitis. Herein, from recent studies and articles, the conventional and emerging hydrogels in the delivery of anti-inflammatory drugs and the therapy for various inflammations are summarized. And their prospects of clinical translation and future development are also discussed in further detail.
Collapse
Affiliation(s)
- Haoyu Yu
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdong518033P. R. China
| | - Rongyao Gao
- Department of ChemistryRenmin University of ChinaBeijing100872P. R. China
| | - Yuxin Liu
- Department of Biomolecular SystemsMax‐Planck Institute of Colloids and Interfaces14476PotsdamGermany
| | - Limin Fu
- Department of ChemistryRenmin University of ChinaBeijing100872P. R. China
| | - Jing Zhou
- Department of ChemistryCapital Normal UniversityBeijing100048P. R. China
| | - Luoyuan Li
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdong518033P. R. China
| |
Collapse
|
35
|
Xu H, Zhang Y, Li L, Ren Y, Qian F, Wang L, Ma H, Quan A, Liu H, Yu R. The nanoprodrug of polytemozolomide combines with MGMT siRNA to enhance the effect of temozolomide in glioma. Drug Deliv 2023; 30:1-13. [PMID: 36579448 PMCID: PMC9809344 DOI: 10.1080/10717544.2022.2152911] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Temozolomide (TMZ) is a conventional chemotherapeutic drug for glioma, however, its clinical application and efficacy is severely restricted by its drug resistance properties. O6-methylguanine-DNA methyltransferase (MGMT) is a DNA repair enzyme, which can repair the DNA damage caused by TMZ. A large number of clinical data show that reducing the expression of MGMT can enhance the chemotherapeutic efficacy of TMZ. Therefore, in order to improve the resistance of glioma to TMZ, an angiopep-2 (A2) modified nanoprodrug of polytemozolomide (P(TMZ)n) that combines with MGMT siRNA (siMGMT) targeting MGMT was developed (A2/T/D/siMGMT). It not only increased the amount of TMZ within tumor lesion site, but also reduced MGMT expression in glioma. The in vitro experiments indicated that the A2/T/D/siMGMT effectively enhanced the cellular uptake of TMZ and siMGMT, and resulted in a significant cell apoptosis and cytotoxicity in the glioma cells. The in vivo experiments showed that glioma growth was inhibited and the survival time of animals were prolonged remarkably after A2/T/D/siMGMT was injected via tail vein. The results showed that the therapeutic effect of A2/T/D/siMGMT in the treatment of glioma was significantly improved.
Collapse
Affiliation(s)
- Haoyue Xu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yongkang Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Linfeng Li
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yanhong Ren
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Feng Qian
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Lansheng Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Hongwei Ma
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Ankang Quan
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Hongmei Liu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China,Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China,Hongmei Liu Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China,CONTACT Yu Rutong;
| |
Collapse
|
36
|
Yao J, Song S, Zhao H, Yuan Y. Platinum-based drugs and hydrogel: a promising anti-tumor combination. Drug Deliv 2023; 30:2287966. [PMID: 38083803 PMCID: PMC10987050 DOI: 10.1080/10717544.2023.2287966] [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: 06/14/2023] [Accepted: 10/04/2023] [Indexed: 12/18/2023] Open
Abstract
Platinum-based drugs are widely used as first-line anti-tumor chemotherapy agents. However, they also have nonnegligible side effects due to the free drugs in circulation. Therefore, it is necessary to develop efficient and safe delivery systems for better tumor cell targeting. Hydrogel is a promising anti-tumor drug carrier that can form a platinum/hydrogel combination system for drug release, which has shown better anti-tumor effects in some studies. However, there is a lack of systematic summary in this field. This review aims to provide a comprehensive overview of the platinum/hydrogel combination system with the following sections: firstly, an introduction of platinum-based drugs; secondly, an analysis of the platinum/hydrogel combination system; and thirdly, a discussion of the advantages of the hydrogel-based delivery system. We hope this review can offer some insights for the development of the platinum/hydrogel combination system for better cancer therapy.
Collapse
Affiliation(s)
- Jiamin Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Shaojuan Song
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Hang Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yao Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
37
|
Min K, Sahu A, Jeon SH, Tae G. Emerging drug delivery systems with traditional routes - A roadmap to chronic inflammatory diseases. Adv Drug Deliv Rev 2023; 203:115119. [PMID: 37898338 DOI: 10.1016/j.addr.2023.115119] [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: 09/15/2022] [Revised: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 10/30/2023]
Abstract
Inflammation is prevalent and inevitable in daily life but can generally be accommodated by the immune systems. However, incapable self-healing and persistent inflammation can progress to chronic inflammation, leading to prevalent or fatal chronic diseases. This review comprehensively covers the topic of emerging drug delivery systems (DDSs) for the treatment of chronic inflammatory diseases (CIDs). First, we introduce the basic biology of the chronic inflammatory process and provide an overview of the main CIDs of the major organs. Next, up-to-date information on various DDSs and the associated strategies for ensuring targeted delivery and stimuli-responsiveness applied to CIDs are discussed extensively. The implementation of traditional routes of drug administration to maximize their therapeutic effects against CIDs is then summarized. Finally, perspectives on future DDSs against CIDs are presented.
Collapse
Affiliation(s)
- Kiyoon Min
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Abhishek Sahu
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Hajipur, 844102, India
| | - Sae Hyun Jeon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea.
| |
Collapse
|
38
|
Schiffmann N, Liang Y, Nemcovsky CE, Almogy M, Halperin-Sternfeld M, Gianneschi NC, Adler-Abramovich L, Rosen E. Enzyme-Responsive Nanoparticles for Dexamethasone Targeted Delivery to Treat Inflammation in Diabetes. Adv Healthc Mater 2023; 12:e2301053. [PMID: 37498238 DOI: 10.1002/adhm.202301053] [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: 04/03/2023] [Revised: 07/25/2023] [Indexed: 07/28/2023]
Abstract
Diabetes is a global epidemic accompanied by impaired wound healing and increased risk of persistent infections and resistance to standard treatments. Therefore, there is an immense need to develop novel methods to specifically target therapeutics to affected tissues and improve treatment efficacy. This study aims to use enzyme-responsive nanoparticles for the targeted delivery of an anti-inflammatory drug, dexamethasone, to treat inflammation in diabetes. These nanoparticles are assembled from fluorescently-labeled, dexamethasone-loaded peptide-polymer amphiphiles. The nanoparticles are injected in vivo, adjacent to labeled collagen membranes sub-periosteally implanted on the calvaria of diabetic rats. Following their implantation, collagen membrane resorption is linked to inflammation, especially in hyperglycemic individuals. The nanoparticles show strong and prolonged accumulation in inflamed tissue after undergoing a morphological switch into microscale aggregates. Significantly higher remaining collagen membrane area and less inflammatory cell infiltration are observed in responsive nanoparticles-treated rats, compared to control groups injected with free dexamethasone and non-responsive nanoparticles. These factors indicate improved therapeutic efficacy in inflammation reduction. These results demonstrate the potential use of enzyme-responsive nanoparticles as targeted delivery vehicles for the treatment of diabetic and other inflammatory wounds.
Collapse
Affiliation(s)
- Nathan Schiffmann
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Faculty of Medicine, and The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Yifei Liang
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Carlos E Nemcovsky
- Department of Periodontology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Michal Almogy
- Department of Periodontology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Michal Halperin-Sternfeld
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Faculty of Medicine, and The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Nathan C Gianneschi
- Department of Chemistry, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science & Engineering, Department of Biomedical Engineering, Department of Pharmacology, Northwestern University, Evanston, IL, 60208, USA
| | - Lihi Adler-Abramovich
- Department of Oral Biology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Faculty of Medicine, and The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Eyal Rosen
- Department of Endodontology, The Maurice and Gabriela Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel
| |
Collapse
|
39
|
Zhang C, Ma P, Qin A, Wang L, Dai K, Liu Y, Zhao J, Lu Z. Current Immunotherapy Strategies for Rheumatoid Arthritis: The Immunoengineering and Delivery Systems. RESEARCH (WASHINGTON, D.C.) 2023; 6:0220. [PMID: 39902178 PMCID: PMC11789687 DOI: 10.34133/research.0220] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/16/2023] [Indexed: 02/05/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease accompanied by persistent multiarticular synovitis and cartilage degradation. The present clinical treatments are limited to disease-modifying anti-rheumatic drugs (DMARDs) and aims to relieve pain and control the inflammation of RA. Despite considerable advances in the research of RA, the employment of current clinical procedure is enormous, hindered by systemic side effect, frequent administration, tolerance from long-lasting administration, and high costs. Emerging immunoengineering-based strategies, such as multiple immune-active nanotechnologies via mechanism-based immunology approaches, have been developed to improve specific targeting and to reduce adverse reactions for RA treatments. Here, we review recent studies in immunoengineering for the treatment of RA. The prospect of future immunoengineering treatment for RA has also been discussed.
Collapse
Affiliation(s)
- Chenyu Zhang
- School of Medicine, Shanghai University, Shanghai, China
- Clinical and Translational Research Center for 3D Printing Technology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peixiang Ma
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Degeneration and Regeneration in Skeletal System, Shanghai, China
| | - An Qin
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Degeneration and Regeneration in Skeletal System, Shanghai, China
| | - Liao Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kerong Dai
- Clinical and Translational Research Center for 3D Printing Technology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Liu
- School of Medicine, Shanghai University, Shanghai, China
| | - Jie Zhao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Degeneration and Regeneration in Skeletal System, Shanghai, China
| | - Zuyan Lu
- Clinical and Translational Research Center for 3D Printing Technology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
40
|
Xu Y, Luo Y, Weng Z, Xu H, Zhang W, Li Q, Liu H, Liu L, Wang Y, Liu X, Liao L, Wang X. Microenvironment-Responsive Metal-Phenolic Nanozyme Release Platform with Antibacterial, ROS Scavenging, and Osteogenesis for Periodontitis. ACS NANO 2023; 17:18732-18746. [PMID: 37768714 DOI: 10.1021/acsnano.3c01940] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Periodontitis is a chronic inflammatory disease deriving from dental plaque, characterized by the excessive accumulation of reactive oxygen species (ROS), matrix metalloproteinase (MMP) and other substances, resulting in the destruction of periodontal tissues. At present, the main therapeutic modalities, such as local mechanical debridement and antibiotic delivery, are not only difficult to solve the intractable bacterial biofilm effectively but also tricky to ameliorate the excessive inflammatory response as well as regenerate the impaired periodontal tissues. Herein, we have proposed the TM/BHT/CuTA hydrogel system formed by the self-assembly of the copper-based nanozyme (copper tannic acid coordination nanosheets, CuTA NSs) and the triglycerol monostearate/2,6-di-tert-butyl-4-methylphenol (TM/BHT) hydrogel. The negatively charged TM/BHT/CuTA can retain at the inflammation sites with a positive charge through electrostatic adsorption and hydrolyze in response to the increasing MMP of periodontitis, realizing the on-demand release of the CuTA nanozyme. The released CuTA nanozyme has antibacterial and antiplaque properties. Meanwhile, as a metal-phenolic nanozyme, it can scavenge multiple ROS by simulating the cascade process of superoxide dismutase (SOD) and catalase (CAT). Further, the CuTA nanozyme can modulate the macrophage polarization from M1 phenotype to M2 phenotype through the Nrf2/NF-κB pathway, which reduces the pro-inflammatory cytokines, increases the anti-inflammatory cytokines, and promotes the expression of osteogenetic genes successively, thus relieving the inflammation and accelerating the tissue regeneration of periodontitis. Altogether, this multifunctional nanozyme on-demand release platform (TM/BHT/CuTA) provides a desirable strategy for the treatment of periodontitis.
Collapse
Affiliation(s)
- Yingying Xu
- The Affiliated Stomatological Hospital, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Yifan Luo
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Zhenzhen Weng
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Haichang Xu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Wei Zhang
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Qun Li
- The Affiliated Stomatological Hospital, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Huijie Liu
- The Affiliated Stomatological Hospital, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Lubing Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Yanmei Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| | - Xuexia Liu
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
- College of Chemistry and Chemical Engineering, Jinggangshan University, Ji'an, Jiangxi 343009, P. R. China
| | - Lan Liao
- The Affiliated Stomatological Hospital, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Medical College, Jinggangshan University, Ji'an, Jiangxi 343009, P. R. China
| | - Xiaolei Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, Jiangxi 330088, P. R. China
| |
Collapse
|
41
|
Abbaszadeh S, Nosrati-Siahmazgi V, Musaie K, Rezaei S, Qahremani M, Xiao B, Santos HA, Shahbazi MA. Emerging strategies to bypass transplant rejection via biomaterial-assisted immunoengineering: Insights from islets and beyond. Adv Drug Deliv Rev 2023; 200:115050. [PMID: 37549847 DOI: 10.1016/j.addr.2023.115050] [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: 03/30/2023] [Revised: 06/14/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Novel transplantation techniques are currently under development to preserve the function of impaired tissues or organs. While current technologies can enhance the survival of recipients, they have remained elusive to date due to graft rejection by undesired in vivo immune responses despite systemic prescription of immunosuppressants. The need for life-long immunomodulation and serious adverse effects of current medicines, the development of novel biomaterial-based immunoengineering strategies has attracted much attention lately. Immunomodulatory 3D platforms can alter immune responses locally and/or prevent transplant rejection through the protection of the graft from the attack of immune system. These new approaches aim to overcome the complexity of the long-term administration of systemic immunosuppressants, including the risks of infection, cancer incidence, and systemic toxicity. In addition, they can decrease the effective dose of the delivered drugs via direct delivery at the transplantation site. In this review, we comprehensively address the immune rejection mechanisms, followed by recent developments in biomaterial-based immunoengineering strategies to prolong transplant survival. We also compare the efficacy and safety of these new platforms with conventional agents. Finally, challenges and barriers for the clinical translation of the biomaterial-based immunoengineering transplants and prospects are discussed.
Collapse
Affiliation(s)
- Samin Abbaszadeh
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Vahideh Nosrati-Siahmazgi
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran
| | - Kiyan Musaie
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Saman Rezaei
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran
| | - Mostafa Qahremani
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran
| | - Bo Xiao
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715 China.
| | - Hélder A Santos
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands; Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
42
|
Kumar A, Kanika, Kumar V, Ahmad A, Mishra RK, Nadeem A, Siddiqui N, Ansari MM, Raza SS, Kondepudi KK, Khan R. Colon-Adhering Delivery System with Inflammation Responsiveness for Localized Therapy of Experimental Colitis. ACS Biomater Sci Eng 2023; 9:4781-4793. [PMID: 37497615 DOI: 10.1021/acsbiomaterials.3c00480] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Ulcerative colitis (UC) is a chronic inflammation-related disease that severely affects the colon and rectum regions. A variety of therapy regimens are used for the treatment of UC. Clinically, therapeutic enema is the choice of therapy for UC patients. Irrespective of on-site administration, the major limitation of therapeutic enemas is the dispossession of the medicine followed by low drug availability for the therapeutic action. In our present work, we have developed an enzyme-responsive injectable hydrogel (ER-hydrogel) to overcome the limitations of therapeutic enema. The hydrogels possess two major advantages, which are being exploited for therapeutic drug delivery in UC: prolonged retention and enzyme responsiveness. The former is one of the prominent advantages of hydrogel compared to free drug enema and the latter controls the release of the drug or provides drug release on-demand. The ER-hydrogel was formulated by the heat-cool method and for therapeutic purposes, a corticosteroid drug, budesonide (Bud), was encapsulated into the ER-hydrogel and evaluated for its various physicochemical and therapeutic potentials in dextran sodium sulfate (DSS)-induced UC. In vitro and ex vivo adhesion studies confirm the retention or mucoadhesive nature of the ER-hydrogel, and the upsurge in Bud release from the Bud-loaded ER-hydrogel upon the addition of esterase enzyme confirms the enzyme-mediated drug release from the ER-hydrogel. Moreover, Bud-loaded ER-hydrogel exhibited promising results in alleviating the disease activity index of UC, and restored the length of the colon, which is the main hallmark of UC. In terms of the health of the colon tissue, the Bud-loaded ER-hydrogel restored the colonic tissue damage, as seen in the H&E-stained, AB-NR-stained, and HID-AB-stained colon sections. Finally, the Bud-loaded ER-hydrogel also markedly subsided the IL-1β, TNF-α, MPO, and nitrite levels in serum and colon tissues. Thus, the fabricated Bud-loaded ER-hydrogel possesses appreciable translational potential due to its ability to significantly ameliorate inflammatory changes compared to naive or water-based therapeutic enema in acute experimental colitis in mice.
Collapse
Affiliation(s)
- Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Vibhu Kumar
- National Agri-Food Biotechnology Institute, Mohali, Punjab 140306, India
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Rakesh Kumar Mishra
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Nahid Siddiqui
- Amity Institute of Biotechnology, Amity University, Noida 201303, India
| | - Md Meraj Ansari
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Syed Shadab Raza
- Department of Stem Cell Biology and Regenerative Medicine, Era University, Sarfarazganj, Lucknow 226003, India
| | | | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| |
Collapse
|
43
|
Pan J, Liao H, Gong G, He Y, Wang Q, Qin L, Zhang Y, Ejima H, Tardy BL, Richardson JJ, Shang J, Rojas OJ, Zeng Y, Guo J. Supramolecular nanoarchitectonics of phenolic-based nanofiller for controlled diffusion of versatile drugs in hydrogels. J Control Release 2023; 360:433-446. [PMID: 37422124 DOI: 10.1016/j.jconrel.2023.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 06/09/2023] [Accepted: 07/02/2023] [Indexed: 07/10/2023]
Abstract
Drug-dependent design of hydrogels is currently required for engineering the controlled release of therapeutics, which is a major contributor to the technical challenges relating to the clinical translation of hydrogel-drug systems. Herein, by integrating supramolecular phenolic-based nanofillers (SPFs) into hydrogel microstructures we developed a facile strategy to endow a range of clinically relevant hydrogels with controlled release properties for diverse therapeutic agents. The assembly of multiscale SPF aggregates leads to tunable mesh size and multiple dynamic interactions between SPF aggregates and drugs, which relaxes the available choices of drugs and hydrogels. This simple approach allowed for the controlled release of 12 representative drugs evaluated with 8 commonly used hydrogels. Moreover, the anesthetic drug lidocaine was loaded into SPF-integrated alginate hydrogel and demonstrated sustained release for 14 days in vivo, validating the potential for long-term anesthesia in patients.
Collapse
Affiliation(s)
- Jiezhou Pan
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Haotian Liao
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China; Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Guidong Gong
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yunxiang He
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Qin Wang
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China; School of Pharmacy, Southwest Minzu University, Chengdu, Sichuan 610065, China
| | - Lang Qin
- Key Laboratory of Birth Defects and Related of Women and Children of Ministry of Education, The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yaoyao Zhang
- Key Laboratory of Birth Defects and Related of Women and Children of Ministry of Education, The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hirotaka Ejima
- Department of Materials Engineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Blaise L Tardy
- Department of Chemical Engineering, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Joseph J Richardson
- Department of Chemical and Environmental Engineering, RMIT University, Melbourne, Victoria 3000, Australia
| | - Jiaojiao Shang
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| | - Orlando J Rojas
- Bioproducts Institute, Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Yong Zeng
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China; Bioproducts Institute, Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| |
Collapse
|
44
|
Xue S, Ruan G, Li J, Madry H, Zhang C, Ding C. Bio-responsive and multi-modality imaging nanomedicine for osteoarthritis theranostics. Biomater Sci 2023; 11:5095-5107. [PMID: 37305990 DOI: 10.1039/d3bm00370a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Osteoarthritis (OA) is one of the most common joint diseases currently, characterized by the gradual degradation of cartilage, remodeling of subchondral bone, development of synovitis, degenerative alterations in the menisci, and formation of osteophytes. Generally, loss of articular cartilage is the most common pathological manifestation of OA. However, owing to the lack of blood vessels and nerves, the damaged cartilage is unable to execute self-repair. Therefore, early detection and treatment of cartilage lesions are extremely vital. Given that precise diagnosis and therapeutic strategy are indispensable from the basic pathological features of OA, an ideal therapeutic strategy should cater to the specific features of the OA microenvironment to achieve disease-modifying therapy. To date, nanomedicine presents an opportunity to achieve the precisely targeted delivery of agents and stimuli-sensitive release at the optimum dose, which may be coupled with a controlled release profile and reduced side effects. This review mainly summarizes inherent and microenvironment traits of OA and outlines stimuli-responsive nanotherapies, including internal bio-responsive (e.g., reactive oxygen species, pH, and protease) and external (e.g., photo stimuli, temperature, ultrasound, and magnetic field) responsive nanotherapies. Furthermore, multi-targeted therapeutic strategies combined with multi-modality imaging are also discussed. In general, future exploration of more novel stimuli-responsive nanotherapies that can be used for early diagnosis and cartilage targeting may help ameliorate OA-related cartilage damage, decrease pain, and promote joint function.
Collapse
Affiliation(s)
- Song Xue
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guangfeng Ruan
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jia Li
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Henning Madry
- Center of Experimental Orthopedics, Saarland University and Saarland University Medical Center, Homburg, Germany
| | - Chao Zhang
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia.
| |
Collapse
|
45
|
Noddeland HK, Lind M, Petersson K, Caruso F, Malmsten M, Heinz A. Protease-Responsive Hydrogel Microparticles for Intradermal Drug Delivery. Biomacromolecules 2023. [PMID: 37307231 DOI: 10.1021/acs.biomac.3c00265] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Protease-responsive multi-arm polyethylene glycol-based microparticles with biscysteine peptide crosslinkers (CGPGG↓LAGGC) were obtained for intradermal drug delivery through inverse suspension photopolymerization. The average size of the spherical hydrated microparticles was ∼40 μm after crosslinking, making them attractive as a skin depot and suitable for intradermal injections, as they are readily dispensable through 27G needles. The effects of exposure to matrix metalloproteinase 9 (MMP-9) on the microparticles were evaluated by scanning electron microscopy and atomic force microscopy, demonstrating partial network destruction and decrease in elastic moduli. Given the recurring course of many skin diseases, the microparticles were exposed to MMP-9 in a flare-up mimicking fashion (multiple-time exposure), showing a significant increase in release of tofacitinib citrate (TC) from the MMP-responsive microparticles, which was not seen for the non-responsive microparticles (polyethylene glycol dithiol crosslinker). It was found that the degree of multi-arm complexity of the polyethylene glycol building blocks can be utilized to tune not only the release profile of TC but also the elastic moduli of the hydrogel microparticles, with Young's moduli ranging from 14 to 140 kPa going from 4-arm to 8-arm MMP-responsive microparticles. Finally, cytotoxicity studies conducted with skin fibroblasts showed no reduction in metabolic activity after 24 h exposure to the microparticles. Overall, these findings demonstrate that protease-responsive microparticles exhibit the properties of interest for intradermal drug delivery.
Collapse
Affiliation(s)
- Heidi K Noddeland
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, 2750 Ballerup, Denmark
| | - Marianne Lind
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, 2750 Ballerup, Denmark
| | - Karsten Petersson
- Explorative Formulation & Technologies, CMC Design and Development, LEO Pharma A/S, 2750 Ballerup, Denmark
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Martin Malmsten
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark
- Department of Physical Chemistry 1, University of Lund, SE-22100 Lund, Sweden
| | - Andrea Heinz
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
46
|
Jiang Q, Zhang S. Stimulus-Responsive Drug Delivery Nanoplatforms for Osteoarthritis Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206929. [PMID: 36905239 DOI: 10.1002/smll.202206929] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/16/2023] [Indexed: 06/08/2023]
Abstract
Osteoarthritis (OA) is one of the most prevalent age-related degenerative diseases. With an increasingly aging global population, greater numbers of OA patients are providing clear economic and societal burdens. Surgical and pharmacological treatments are the most common and conventional therapeutic strategies for OA, but often fall considerably short of desired or optimal outcomes. With the development of stimulus-responsive nanoplatforms has come the potential for improved therapeutic strategies for OA. Enhanced control, longer retention time, higher loading rates, and increased sensitivity are among the potential benefits. This review summarizes the advanced application of stimulus-responsive drug delivery nanoplatforms for OA, categorized by either those that depend on endogenous stimulus (reactive oxygen species, pH, enzyme, and temperature), or those that depend on exogenous stimulus (near-infrared ray, ultrasound, magnetic fields). The opportunities, restrictions, and limitations related to these various drug delivery systems, or their combinations, are discussed in areas such as multi-functionality, image guidance, and multi-stimulus response. The remaining constraints and potential solutions that are represented by the clinical application of stimulus-responsive drug delivery nanoplatforms are finally summarized.
Collapse
Affiliation(s)
- Qi Jiang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| | - Shufang Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| |
Collapse
|
47
|
Wang K, Yin C, Ye X, Chen Q, Wu J, Chen Y, Li Y, Wang J, Duan C, Lu A, Guan D. A Metabolic Driven Bio-Responsive Hydrogel Loading Psoralen for Therapy of Rheumatoid Arthritis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207319. [PMID: 36869654 DOI: 10.1002/smll.202207319] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/13/2023] [Indexed: 05/25/2023]
Abstract
Overexpressed matrix metalloproteinases, hypoxia microenvironment, and metabolic abnormality are important pathological signs of rheumatoid arthritis (RA). Designing a delivery carrier according to the pathological characteristics of RA that can control drug release in response to disease severity may be a promising treatment strategy. Psoralen is the main active ingredient isolated from Psoralea corylifolia L. and possesses excellent anti-inflammatory activities as well as improving bone homeostasis. However, the specific underlying mechanisms, particularly the possible relationships between the anti-RA effects of psoralen and related metabolic network, remain largely unexplored. Furthermore, psoralen shows systemic side effects and has unsatisfactory solubility. Therefore, it is desirable to develop a novel delivery system to maximize psoralen's therapeutic effect. In this study, a self-assembled degradable hydrogel platform is developed that delivers psoralen and calcium peroxide to arthritic joints and controls the release of psoralen and oxygen according to inflammatory stimulation, to regulate homeostasis and the metabolic disorder of the anoxic arthritic microenvironment. Therefore, the hydrogel drug delivery system based on the responsiveness of the inflammatory microenvironment and regulation of metabolism provides a new therapeutic strategy for RA treatment.
Collapse
Affiliation(s)
- Kexin Wang
- National Key Clinical Specialty/Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Neurosurgery Institute, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, P. R. China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
- Institute of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong, 999077, P. R. China
| | - Chuanhui Yin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Xiangmin Ye
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Quanlin Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jie Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yupeng Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yi Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Jun Wang
- School of Medicine, Foshan University, Foshan, Guangdong, 528225, P. R. China
| | - Chuanzhi Duan
- National Key Clinical Specialty/Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Neurosurgery Institute, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, P. R. China
| | - Aiping Lu
- Institute of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong, 999077, P. R. China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, 510000, P. R. China
| | - Daogang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
- Guangdong Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
48
|
Shen Q, Du Y. A comprehensive review of advanced drug delivery systems for the treatment of rheumatoid arthritis. Int J Pharm 2023; 635:122698. [PMID: 36754181 DOI: 10.1016/j.ijpharm.2023.122698] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 01/21/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023]
Abstract
Rheumatoid arthritis (RA), a chronic autoimmune disease, is characterized by articular pain and swelling, synovial hyperplasia, and cartilage and bone destruction. Conventional treatment strategies for RA involve the use of anti-rheumatic drugs, which warrant high-dose, frequent, and long-term administration, resulting in serious adverse effects and poor patient compliance. To overcome these problems and improve clinical efficacy, drug delivery systems (DDS) have been designed for RA treatment. These systems have shown success in animal models of RA. In this review, representative DDS that target RA through passive or active effects on inflammatory cells are discussed and highlighted using examples. In particular, DDS allowing controlled and targeted drug release based on a variety of stimuli, intra-articular DDS, and transdermal DDS for RA treatment are described. Thus, this review provides an improved understanding of these DDS and paves the way for the development of novel DDS for efficient RA treatment.
Collapse
Affiliation(s)
- Qiying Shen
- School of Pharmacy, Hangzhou Normal University, 2318 Yu-HangTang Road, Hangzhou 311121, China; Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-HangTang Road, Hangzhou 310058, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-HangTang Road, Hangzhou 310058, China.
| |
Collapse
|
49
|
Gu Z, Wang J, Fu Y, Pan H, He H, Gan Q, Liu C. Smart Biomaterials for Articular Cartilage Repair and Regeneration. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202212561] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Indexed: 01/06/2025]
Abstract
AbstractArticular cartilage defects bring about disability and worldwide socioeconomic loss, therefore, articular cartilage repair and regeneration is recognized as a global issue. However, due to its avascular and nearly acellular characteristic, cartilage tissue regeneration ability is limited to some extent. Despite the availability of various treatment methods, including palliative drugs and surgical regenerative therapy, articular cartilage repair and regeneration still face major challenges due to the lack of appropriate methods and materials. Smart biomaterials can regulate cell behavior and provide excellent tissue repair and regeneration microenvironment, thus inducing articular cartilage repair and regeneration. This process is adjusted by controlling drug/bioactive factors release via responding to exogenous/endogenous stimuli, tailoring materials’ structure and function similar to native cartilage or providing physiochemical and physical signaling factors. Herein, smart biomaterials, recently applied in articular cartilage repair and regeneration, are elaborated from two aspects: smart drug release system and smart scaffolds. Furthermore, articular cartilage and its defects and advanced manufacturing techniques of smart biomaterials are discussed in brief. Finally, perspectives for smart biomaterials used in articular cartilage repair and regeneration are presented and the clinical translation of smart biomaterials is emphasized.
Collapse
Affiliation(s)
- Zhanghao Gu
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Jiayi Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Yu Fu
- School of Aerospace Engineering and Applied Mechanics Tongji University Zhangwu Road 100 Shanghai 200092 P. R. China
| | - Hao Pan
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Hongyan He
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Qi Gan
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry East China University of Science and Technology Shanghai 200237 P. R. China
| |
Collapse
|
50
|
Dou X, Wang H, Yang F, Shen H, Wang X, Wu D. One-Step Soaking Strategy toward Anti-Swelling Hydrogels with a Stiff "Armor". ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206242. [PMID: 36683238 PMCID: PMC10037974 DOI: 10.1002/advs.202206242] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/01/2023] [Indexed: 06/17/2023]
Abstract
Double-network (DN) hydrogels consisting of noncovalent interacting networks are highly desired due to their well-controlled compositions and environmental friendliness, but the low water resistance always impairs their mechanical strength. Here, an anti-swelling hydrogel possessing the core/shell architecture through rational regulation of multiple weak noncovalent interactions is prepared. A composite hydrogel consists of chitosan (CS) and poly(N-acryloyl 2-glycine) (PACG), readily forming the shell-structured DN hydrogel after soaking in a FeCl3 solution because of in situ formation of chain entanglements, hydrogen bonds, and ionic coordination. The produced DN hydrogels exhibit excellent anti-swelling behaviors and mechanical durability for over half a year, even in some strict situations. Taking the merits of noncovalent bonds in adjustability and reversibility, the swelling property of these hydrogels can be easily customized through control of the ion species and concentrations. A dynamically reversible transition from super-swelling to anti-swelling is realized by breaking up and rebuilding the metal-coordination complexes. This facile but efficient strategy of turning the noncovalent interactions and consequently the mechanics and anti-swelling properties is imperative to achieve the rational design of high-performance hydrogels with specific usage requirements and expand their applicability to a higher stage.
Collapse
Affiliation(s)
- Xueyu Dou
- College of ChemistryChemical Engineering and Materials ScienceKey Laboratory of Molecular and Nano ProbesMinistry of EducationCollaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of ShandongInstitute of Molecular and Nano ScienceShandong Normal UniversityJinan250014China
- Beijing National Laboratory for Molecular SciencesInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hufei Wang
- Beijing National Laboratory for Molecular SciencesInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Fei Yang
- Beijing National Laboratory for Molecular SciencesInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hong Shen
- Beijing National Laboratory for Molecular SciencesInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xing Wang
- Beijing National Laboratory for Molecular SciencesInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Decheng Wu
- Beijing National Laboratory for Molecular SciencesInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- Department of Biomedical EngineeringSouthern University of Science and TechnologyShenzhen518055China
| |
Collapse
|